You are on page 1of 10

Journal of Drug Delivery Science and Technology 67 (2022) 102922

Contents lists available at ScienceDirect

Journal of Drug Delivery Science and Technology


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

Opinion Paper

Intranasal micellar curcumin for the treatment of chronic asthma


Ruchi Chawla a, *, Bhupendra Sahu a, Mohini Mishra a, Varsha Rani a, Rashmi Singh b
a
Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
b
Department of Zoology, MMV, Banaras Hindu University, Varanasi, 221005, India

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

Keywords: Curcumin is a natural phytoconstituent obtained from the rhizomes of Curcuma longa (turmeric) and is known for
Chronic asthma its diverse anti-oxidant and anti-inflammatory benefits, but its clinical utility is limited by its poor aqueous
Curcumin solubility and rapid metabolism, which ultimately affects its bioavailability. The present study is focused on the
Micelles
formulation of curcumin loaded micellar dispersion for intranasal delivery for the treatment of chronic asthma.
Intranasal
Micellar dispersion was prepared by film formation method using poly-(ethylene oxide)-block-distearoyl
Dispersion
Anti-oxidant phosphatidyl-ethanolamine (mPEG5000-DSPE) as lipid surfactant and characterized for its physic-chemical
Anti-inflammatory properties. The curcumin micelles were evaluated for their anti-asthmatic action in ovalbumin (OVA)-induced
allergic asthma model in male wistar rats against standard drug dexamethasone. The micelles showed mean
particle size in the range of 20.03 nm–26.48 nm. The micellar dispersion exhibited negative zeta potential in the
range of − 26.22 to − 25.52 mV. Incorporation of curcumin into micelles helped in enhancing the solubility of
curcumin besides providing it protection against degradation. In vitro studies showed sustained relase of cur­
cumin up to 36 h. A 14- fold increase in the bioaviability of the curcumin was measured when administered as
micelles. In addition, 4.4- fold higher concentration of drug was measured in the lungs for micellar curcumin.
Comparable reduction in the levels of intracellular ROS was observed for i.n. curcumin-micelles (approx.57.6%)
and dexamethasone (59.3%). Also, micellar curcumin produced significant supression (p < 0.05) in the release of
nitric oxide. Through the present study, we can suggest the potential application of curcumin micelles in the
treatment of asthma.

1. Introduction hydroxyeicosatetranoeic acid (HETE) and leukotriene B3 [3].


Drugs to manage chronic signs of asthma include inhaled cortico­
Amongst the inflammatory diseases, asthma is highly prevalent and steroids (ICs), leukotriene modifiers, long-acting beta-agonists (LABAs),
is marked by varying respiratory signs and airflow limitations like theophylline and combination inhalers containing corticosteroids and
hyper-responsiveness with repeated occurrence of wheezing, tightness LABA [2]. Despite effective therapeutic effects, toxicity associated with
of chest, coughing, difficulty in breathing, and airway remodeling. inhaled corticosteroid acts as a limiting factor.
Asthma is triggered by allergic stimuli (mites, cockroach residue, pol­ Different inflammatory signaling mechanisms involved in asthma
lens, mould, and animal scurf) and non-allergic stimuli (viral contagion, include Src family kinases (SFKs), protein kinase C (PKC), growth factor
subjection to tobacco smoke, exercise, cool air), which induce a series of tyrosine kinase receptors, nicotinamide adenine dinucleotide phosphate
episodes leading to chronic airway inflammation (swelling) [1]. (NADPH)/reactive oxygen species (ROS), mitogen activated protein ki­
The mast cell is the progenitor of inflammatory response to all sorts nase (MAPKs), nuclear factor-kappa B (NF-KB), and activator protein-1
of stimuli in the airways [2]. A very small proportion of the mast cells is (AP-1). MAPK pathway significantly influences and regulates cell
present in intra-luminal region, but activation and degranulation of growth and proliferation, chemotaxis, degranulation, and other secre­
these mast cells causes progressive mucosal permeability, with subse­ tary processes [5]. Airway remodeling is the resultant consequence of
quent activation of tissue mast cells [3]. The mast cells generate a va­ long term inflammation, so use of anti-inflammatory agents in the
riety of pro-inflammatory mediators like histamine, serotonin, treatment of asthma can help diminish the risk of aggravation of asthma
eicosanoides and platelet activating factors, cytokines and chemokines and its effects. Among the natural phytochemicals, curcumin consider­
[4], chemotactic factors for neutrophils and eosinophils, such as ably controls inflammation, apoptosis and cell growth, making it

* Corresponding author.
E-mail address: rchawla.phe@iitbhu.ac.in (R. Chawla).

https://doi.org/10.1016/j.jddst.2021.102922
Received 18 March 2020; Received in revised form 13 September 2020; Accepted 14 October 2021
Available online 27 October 2021
1773-2247/© 2021 Elsevier B.V. All rights reserved.
R. Chawla et al. Journal of Drug Delivery Science and Technology 67 (2022) 102922

beneficial for the prevention and treatment of inflammatory diseases. dissolved in 10 ml of ethanol. The mixture was sonicated for 2 min using
Curcumin is a vital curcuminoid present in Curcuma longa (a rhizome) probe sonicator and then vortex mixed thoroughly. The solvent was
and exhibits considerable antioxidant and anti-inflammatory properties eliminated using vacuum rotary evaporator operated at 100 rpm at
[6]. In one of our previous studies we have proposed the use of intra­ 45 ◦ C under vacuum (200 psig), to form a thin film of drug-containing
nasal curcumin as a complementary medication for prevention of airway lipid. Traces of remaining solvent were removed by applying vacuum
inflammations and bronchoconstrictions in asthma without any side overnight. The dried film was rehydrated with 10 ml of phosphate buffer
effect [7]. Studies have shown that intranasal delivery of drugs results in saline [10] alongwith gentle shaking at 40 ◦ C for 10 min. The unsolu­
fascinating results especially for treatment of respiratory tract disorders bilized drug was removed by filtration using 0.22 μm filter [15].
like asthma, chronic obstructive pulmonary disease, pulmonary infec­
tion, pulmonary hypertension, lung cancer, and cystic fibrosis. The 3. Characterization of curcumin loaded micellar dispersion
intra-nasal region provides highly vascularized surface area for ab­
sorption of lipophilic drugs and its eminence lies in sidestepping of 3.1. UV–Vis spectrophotometric method
first-pass metabolism by liver which in turn improves the bioavailability
of the drug [4]. The radical scavenging property of Curcumin was analyzed using UV–Vis spectrophotometric at 427 nm
polymer-glycerosomes of curcumin prepared using sodium hyaluronate λmax according to the method described elsewhere with modification
and trimethyl chitosan was observed in A549 cells stressed with [16].
hydrogen peroxide, in addition to suppressed production of cytokine IL6
and IL8. Nebulized glycerosomes delivered to rats showed significant
accumulation of curcumin in lungs [8]. 3.2. Particle size, polydispersity and zeta potential
Curcumin, though a wonder drug, with multi-targeting pharmaco­
logical potential suffers from poor aqueous solubility and low systemic The particle size, polydispersity, and zeta potential of micelles was
bioavailability [9]. In this present study, we have tried to improve the measured using DELSA™ NANO (Beckman Coulter, USA) based on dy­
pharmaceutical properties of curcumin by loading them into micelles for namic light scattering technique. After suitably diluting the samples in
treatment of asthma. Using coprecipitation rehydration (solvent evap­ ultra purified water and filtration through 0.22μ, size of the micelles was
oration) method, micelles were prepared using poly-(ethylene oxide)-­ measured at 25 ◦ C. For the measurement of zeta potential, samples (in
block-distearoyl phosphatidyl-ethanolamine (mPEG5000-DSPE)/MW triplicate) were diluted suitably in 1 mM NaCl solution and using elec­
5000 Da (mPEG5000-DSPE) [10]. The hydrophobic core of DSPE pro­ trophoretic light scattering technique the potential was recorded at
vides a suitable environment for incorporation of lipophilic drug (cur­ 25 ◦ C under applied electric field of 16 V/cm [17].
cumin) protecting it against degradation. Also, PEG5000-DSPE micelles
are effective drug carriers especially for poorly water soluble drugs
which show non-specific biodistribution and targeting, and poor oral 3.3. Determination of entrapment efficiency
bioavailability [11]. Moreover, PEGylation results in increase in the
molecular weight of carriers leading to increased half-life and further, The amount of drug entrapped in micelles provides an estimate of the
higher retention in the lungs [12]. Studies have shown that PEG mole­ percent entrapment efficiency (%EE). The micellar dispersion was ultra-
cules larger than 5ooo daltons stay longer in the lungs [12]. Further, use centrifuged at 10000 rpm at 4 ◦ C for 15 min. The clear supernatant was
of PEG5000-DSPE forms micelles with longer circulation time. Also, these discarded and dispersion was centrifuged again at 15000 rpm at 4 ◦ C for
structure escape complement activation and scavenging through opso­ 30 min. The pellet free from unentrapped drug was soaked in 10 ml of
nisation leading to better biodistribution [13]. The complement is acetonitrile and sonicated for 10 min. The drug released from micelles
further weakened, if the PEG-conjugated nanocarriers possess a negative was determined spectrophotometrically against similarly digested pla­
zeta potential [14]. cebo blank (without curcumin) [17]. The entrapment efficiency was
calculated using the formula:
2. Materials and methods
%EE = (Drug entrapped in micelles / total drug added) x 100.

2.1. Materials

Curcumin, Ovalbumin, aluminium hydroxide were generously pro­


vided by Sisco Research Laboratory Pvt. Ltd, India and mPEG5000-DSPE 3.4. Solid state characterization studies
was provided by Lipoid, Germany as a gift sample. Potassium dihy­
drogen orthophosphate, disodium hydrogen phosphate and sodium Surface morphology of prepared micelles was visualized using
hydroxide were purchased from Merck Life Science Pvt. Ltd, Mumbai. scanning electron microscope (ZEISS Supra 40, Germany) under normal
Analytical grade chemicals were used in the study. atmospheric conditions. For analysis, suitably diluted samples were
placed on a glass slide, spread evenly and allowed to dry. The dried
samples were then gold coated with the help of QUORUM (Q150RES)
2.2. Formulation of curcumin loaded micellar dispersion
sputter coater for 90 s at 20 mA, after which they were placed in the
sample holder and analyzed at 20 kV.
Curcumin-loaded micelles (C-mic) were prepared by co-precipitation
Interaction between the drug and the excipients can be studied using
rehydration (solvent evaporation) method. Varying ratios (1:2, 1:4, 1:6)
Attenuated Total Reflectance-Infrared (ATR-IR). The properties of a
of curcumin (drug) and mPEG5000-DSPE as shown in Table 1 were
drug are governed by the type of functional groups present in it, and in
case of any interaction with the excipients, it leads to alteration of the
Table 1 functional groups; thereby leading to appearance or disappearance of
Formulation batches prepared using different drug-polymer ratio.
characteristic peaks. In the present study, drug-excipient compatibility
Formulation (Drug: Polymer Amount of drug (in Amount of polymer (in was studied by obtaining ATR spectra (using infrared spectrophotom­
ratio) mg) mg)
eter; Bruker ALPHA ECO-ATR, USA) of curcumin, PEG5000-DSPE,
F1 (1:2) 20 40 physical mixture of curcumin and PEG5000-DSPE (in 1:1 ratio), and
F2 (1:4) 20 80 lyophilized C-mic. The spectra was analyzed and interpreted for the
F3 (1:6) 20 120
presence of characteristic peaks of the functional groups.

2
R. Chawla et al. Journal of Drug Delivery Science and Technology 67 (2022) 102922

3.5. Stability study 3.9. In-vivo antiasthmatic (anti-inflammatory) activity of curcumin in


OVA-induced asthma model
The prepared formulation samples were stored and sealed in USP
Type-1 amber colored glass vials and subjected to stability studies at 30 Reactive oxygen species serve as signaling molecules or indicators of
± 2 ◦ C/65% RH. The samples were evaluated at different time intervals inflammation. Asthma is an inflammatory condition of the respiratory
(15, 30, 45 days) and the stability was evaluated on the basis of changes tract and associated with increased generation of ROS and free-radical
in particle size, zeta potential and drug content [18]. mediated reactions resulting in oxidative stress in the respiratory tract
[21]. Increased levels of ROS cause oxidation of proteins, DNA, and
lipids and may also evoke secondary cellular reactive species. Curcumin
3.6. In-vitro drug release study acts as a scavenger of ROS by enhancing the activity of antioxidant
enzymes like catalase, superoxide dismutase [22], glutathione perox­
In-vitro drug release studies were performed using dialysis membrane idise and heme oxygenase-1 [22,23]. The extent to which curcumin can
technique. After hydrating the dialysis bag (cellulose membrane mol. regulate the generation of ROS was evaluated by measuring the level of
wt. cutoff 12–14000 Da, HIMEDIA, India) overnight in dissolution ROS in BALF cells [24].
media, it was filled with 2 ml of micellar dispersion and both its ends
were tied and firmly clamped. The sealed dialysis bag was incubated in 3.10. Animal studies
150 ml of release medium, (phosphate buffer; pH 7.4), under continuous
stirring at 100 rpm at 37 ± 0.5 ◦ C. 5 ml aliquots (in triplicate) were 3.10.1. Sensitization and challenge protocol
withdrawn at pre-determined time intervals with simultaneous The animals were allowed to acclimatize for 5 days prior to treat­
replacement of equivalent volume of fresh media to maintain sink ment. The animals were randomly divided into six groups (n = 6/group)
condition. After suitably diluting the samples, they were analyzed as: control group (Group I), vehicle (saline) treated (Group II), blank
spectrophotometrically at 427 nm to determine the extent of release of micelles treated (Group III), curcumin suspension treated (Group IV),
curcumin [15]. curcumin micelles treated (Group V), and dexamethasone treated
(Group VI). All groups except control were sensitized intraperitoneally
3.7. In-vivo studies (I.P.) with 1 mg OVA adsorbed on 20 mg Al(OH)3 gelatinous on days 0,
7, and 14. All the animal groups were challenged with 1.1% OVA so­
Male Wistar rats (160 ± 30 g) bred at Central Animal House (CAH), lution in 200 μL normal saline by intratracheal instillation from day
Institute of Medical Science, Banaras Hindu University (IMS BHU), were 15–20 as shown in Fig. 1 [25].
used for the study. The animals were housed under standard laboratory
conditions (25 ± 2 ◦ C, 60–70% humidity), with alternate 12-h dark and 3.10.2. Treatment protocol
light cycle, with free access to food and water. The animals were accli­ OVA sensitized and challenged animals were treated from days
matized to laboratory conditions before the study. The experimental 15–20 as follows: Group I and II were treated with normal saline i.n.;
protocol was approved by the Institutional Animal Ethical Committee group III was treated with i.n. blank micelles, group IV was treated with
(IAEC), IMS, BHU with registration number 542/GO/ReBi//S/02/ C-susp (5 mg/kg i.n.), group V was treated with prepared C-mic (5 mg/
CPCSEA. kg i.n.) and group VI was treated with dexamethasone (1 mg/kg i.p.)
(shown in Fig. 1) [26].

3.8. Pharmacokinetic study of curcumin 3.10.3. Collection of BAL-Fluid


After euthanization (pentobarbital sodium 200 mg/kg, i.p.) the an­
Animals were fasted overnight before the study. The animal was imals were sacrificed on day21. After intratracheal cannulation, 1 ml of
divided into two groups with 6 animals each; Group I was treated with chilled BPS was injected into the lungs and applying gentle aspiration
curcumin loaded micellar dispersion (C-mic) and Group II with curcu­ BAL fluid was collected. The procedure was repeated twice and the fluid
min suspension (C-susp). Each group was administered 5 mg/kg of body was centrifuged at 3000 rpm for 10 min at 4 ◦ C and the cell pellet was
weight of curcumin intranasally. Blood samples (500 μl) were collected collected.
from the femoral vein at designated time intervals (0, 0.5, 1, 2, 4, 6, 8, 12
and 24 h) and stored in Eppendorf tubes containing heparin (0.2 mg/ 3.10.4. Measurement of intracellular reactive oxygen species (iROS)
ml). The samples were centrifuged at 1500 rpm for 15 min and the Total number of viable cells present in BALF were counted and then,
supenatant collected was deproteinized with acetonitrile. The depro­ the cells were washed with chilled phosphate buffer saline. For labelling
teinzed samples were vortexed for 2 min and then centrifuged at 14000 of iROS, cells at a concentration of 1 X 105 were incubated in 100 μl of
rpm for 5 min. The supernatant was then filtered through 0.22-μm sy­ 20 mM dichlorofluorescein diacetate (DCFH-DA) for 45 min at 37 ◦ C in
ringe filter and subjected to chromatographaic analysis [7,15,14]. Iso­ dark. Further, using fluorescence spectrometer (λex: 490 nm; λem: 515
cratic elution was performed on Waters XTerra RP18, 5 μm, 250 mm × nm), the fluorescence intensity was measured with the values being
4.6 mm i.d C18 analytical column, coupled with a PDA detector at a flow normalized against the fluoresence intensity of normal cells [5].
rate of 1 ml/min, using ACN: Water (60:40) as mobile phase at 425 nm
[19]. 3.10.5. Detection of nitrite level in serum
Nitric oxide is one of the potential makers of inflammation and
3.8.1. Measurement of the concentration of curcumin in bronchoalveolar higher levels of nitric oxide are produced in asthamatic condition. Three
fluid isoforms (two constitutive and one inducible) of nitric oxide enzyme are
At the end of the study, rats were sacrificed, and bronchoalveolar responsible for production of neuronal or type I (nNOS), inducible iso­
fluid was collected by the tracheal cannulation method [20]. 1 ml of form or type II (iNOS), and the endothelial isoform or type 3 (eNOS)
chilled BPS was injected into the lungs through cannula and rinsed [26–31]. Studies have shown that curcumin acts as a scavenger of nitric
twice. The broncioalveolar fluid was mixed with acetonitrile in the ratio oxide and modulates signaling pathways mediated via (NF-KB)and
1:3, vortex mixed. After centrifugation at 3000 rpm for 10 min, super­ mitogen-activated protein kinase, which prevent bronchial inflamma­
natant (organic phase) was collected and filtered through a 0.22-μm tion in asthmatic patients [30]. Further, studies have also shown that
syringe filter and analyzed by chromatographic method to determine the curcumin decreases the nitric oxide synthase activity [32–34]. Studies
concentration of curcumin. have shown that curcumin may be used as an adjuvant in the treatment

3
R. Chawla et al. Journal of Drug Delivery Science and Technology 67 (2022) 102922

Fig. 1. Study protocol for OVA-induced asthma model.

of asthma as it suppresses iNOS and NO. In the present study, the effect 3.02%, 83.4 ± 3.82% and 88.8 ± 4.14% respectively. With increase in
of intranasally delivered curcumin micelles on the relase of nitric oxide the proportion of lipid, an increase in % entrapment efficiency was
was studied. At room temperature, 100 μl of serum was mixed with 100 observed, probably due to availability of larger micellar core for the
μl of vanadium chloride, followed by addition of 100 μl of Griess reagent incorporation of drug.
to the mixture. The 96 microwell plate was incubated for 45 min at
37 ◦ C, and the absorbance was measured at 540 nm using microplate 4.1.3. Solid state characterization studies
reader. The concentration in samples was estimated against standard The scanning electron micrographs of curcumin-loaded micelles
curve of sodium nitrite [35]. have been shown in Fig. 2. Spherical micelles with a core-shell structure
were observed within size range of 50 nm as a result of minimization of
4. Result and discussion interfacial free energy of block copolymers, which tends towards the
hydrophobic part i.e. the core of micelles [38].
4.1. Evaluation of curcumin loaded micellar dispersion IR spectra of curcumin, PEG5000-DSPE, physical mixture of curcumin
and PEG5000-DSPE, and C-mic (batch F2; data for batches F1 and F3 not
4.1.1. Particle size, polydispersity and zeta potential mentioned) was taken (Fig. 3). Characteristic peaks of curcumin corre­
Table 2, shows the mean particle size, and polydispersity of curcumin sponding to –OH stretching was observed at 3588.23 cm− 1. Peaks for
loaded micellar dispersion. The mean particle size of formulations F1, stretching of aromatic C-H and aliphatic C-H in curcumin were observed
F2, and F3 were found to be 20.03 nm, 24.92 nm, and 26.48 nm at 3115.78 cm− 1 and 2879.01 cm− 1 respectively. Peak corresponding to
respectively with a zeta potential of − 26.22, − 24.68, and − 25.52 C=O was found at 1697.37 cm− 1 and that due to C-O stretching was
respectively. Increase in lipid amount resulted in an increase of particle found between 1288.89 cm− 1 and 1394.33 cm− 1. All the characteristic
size. Sub-micron sized particles with diameter less than 100 nm stay peaks of curcumin were retained and no extra peaks were observed in
dissolve in the alveolar fluid and evade phagocytosis and mucociliary the physical mixture and the formulation, indicating absence of any
clearance thereby increasing the time for which micelles stay in the physical or chemical incompatibility between drug and excipients used
lungs [17]. The carbamate linkage by which PEG is conjugated to dis­ in formulation of micelles.
tearoyl phosphatidylethanolamine (DSPE) imparts a net negative charge
to the phosphate moiety resulting in negative zeta potential of the 4.1.4. Stability study
formulation. A large negative charge provides stability against aggre­ The curcumin-loaded micelles were subjected to stability testing and
gation and as per already published reports retention of negatively the results were compared with freshly prepared batches corresponding
charged entities is significantly higher in the lungs in comparison to to the formulations F1, F2 and F3. No significant difference in particle
their positively charged counterparts [35,36]. The dispersion exhibited size, zeta potential, and drug content were observed between the sta­
as narrow size distribution indicating a homogenous formulation bility batches and fresh batch. Polyethyleneglycol (PEG) conjugated
(Table 2). Monodisperse lower-sized particles are absorbed highly micelles have lower critical micellar concentration (CMS) in comparison
through the nasal mucosa resulting in higher bioavailability. to conventional micelles. Studies have shown CMC of PEG conjugated
micelles to be around 10− 7 M, while in conventional micelles it is about
4.1.2. Entrapment efficiency 10− 3 to 10− 4 M. So, conjugation with PEG result in the formation of a
Percentage ratio of the amount of drug loaded in micelles to the stable micellar system, which shows slow dissociation and allows
theoretical quantity of the drug added during preparation of micelles incorporation and retention of drugs for longer duration providing
can be indicated as Percentage drug entrapment efficiency (%EE). The higher drug concentration in the target tissues [39].
drug entrapment efficiency of F1, F2 and F3 was found to be 73.1 ±
4.2. In-vitro drug release study
Table 2
Fig. 4, shows the cumulative percentage release of curcumin from the
Particle size, polydispersity, zeta potential, and %entrapment efficiency of
formulation batches. prepared micellar dispersion (F1, F2, and F3) in phosphate buffered
saline (pH 7.4). The study performed by the dialysis membrane method
Formulation Drug: Average PDI Zeta Entrapment
at 37◦ ± 0.5 ◦ C with continuous stirring at 100 rpm, showed slow and
Lipid Particle Size Potential Efficiency
ratio (nm) (mV) sustained release for more than 36 h with a burst release during the first
hour. Surface bound drug resulted in burst release followed by diffusion
FI 1:2 20.03 ± 1.92 0.238 − 26.22 ± 73.1 ±
2.02 3.02% controlled sustained release of curcumin from the core to the surface.
F2 1:4 24.92 ± 2.19 0.194 − 24.68 ± 83.4 ± Approximately, 60.38%, 73.19%, and 58.04% of curcumin was released
1.89 3.82% from formulations F1, F2 and F3 respectively after 36 h of study. The
F3 1:6 26.48 ± 1.51 0.141 − 25.52 ± 88.8 ± t50% (time at which 50% of the drug was released in medium) of the
2.11 4.14%
formulations were about 14h, 10h, and 12h for the F1, F2, and F3,

4
R. Chawla et al. Journal of Drug Delivery Science and Technology 67 (2022) 102922

Fig. 2. Scanning electron micrographs of formulation batches of C-mic F1, F2 and F3.

5
R. Chawla et al. Journal of Drug Delivery Science and Technology 67 (2022) 102922

Fig. 3. ATR-IR spectra of curcumin, PEG5000-DSPE, physical mixture of curcumin and PEG5000-DSPE (1:1), and C-mic formulation batch F2.

Fig. 4. Drug release profile of curcumin from formulation batches F1, F2 and F3.

respectively. The slow and sustained release of the drug could be evi­ coefficient (R2) of around 0.99. The pharmacokinetic parameters of
denced by the values of t50% [37]. intranasally administered curcumin suspension and curcumin loaded
micellar dispersion have been shown in Table 3. Micelles produced
almost double concentration of curcumin at Cmax in comparison to
4.3. In-vivo studies
suspension. The reference chromatograms have been shown in Figs. 5
and 6.
4.3.1. Pharmacokinetic study of curcumin in rat
Formulation batch F2 with highest entrapment efficiency (83.4%)
and in vitro drug release (73.012%) was used for further pharmacoki­
Table 3
netic evaluation. The concentration of curcumin in biological samples
Pharmacokinetic parameters of curcumin.
was determined by RP-HPLC method [40]. For preparation of standard
Product Cmax AUC0–24 h Tmax Ke (1/ T1/2 MRT
calibration curve, working stock solutions were prepared by spiking
(μg/ml) (μg⋅h/mL) (h) h) (h) (h)
blank plasma/BALF with known amounts of drugs in the concentration
range of 2 μg/ml to 50 μg/ml. The calibration curve showed linear C-susp 6.94 66.79 2 0.0958 7.232 7.889
C-mic 13.28 954.33 4 0.0797 8.688 10.266
correlation between drug concentration and peak area with a correlation

6
R. Chawla et al. Journal of Drug Delivery Science and Technology 67 (2022) 102922

Fig. 5. HPLC Chromatogram of (a) blank and (b) pure curcumin (30 μg) analyzed at 425 nm.

Entrapment of curcumin within the micellar core protected it against was examined in the experimental groups by applying repeated measure
degradation and resulted in its increased availability, as can be corre­ One Way ANOVA with Newman-Keuls Multiple Comparision Test.
lated with a 14-fold increase in the bioavailability of curcumin. The Fig. 9 shows significant difference in level of ROS between control
plasma concentration versus time profile has been shown in Fig. 7. and treatment groups (p < 0.05). Production of ROS increased after OVA
Further, slower rate of elimination from micelles resulted in increased sensitization and challenge in groups II and III as compared to the
residence (MRT) of curcumin in body. Control group. Results has showed decrease level of intracellular ROS
(iROS) that is 57.6% and 59.3% in curcumin-micelle i.n administred
4.3.2. Measurement of the concentration of curcumin in BALF group V, and i.p. dexamethasone administred group VI animals
respectively. The levels of iROS decreased to 46.5% and 44.2% respec­
4.3.2.1. In-vivo antiasthmatic (anti-inflammatory) activity of curcumin in tively in group II and III (Table 4). These study shows that intranasally
OVA-induced asthma model. After 24 h of i.n. administration of curcu­ administered curcumin micelles exhibit higher anti-oxidant effect as
min formulations (micelles and suspension), the concentration of cur­ compared to curcumin suspension and exert a comparatively similar
cumin was measured in BALF (Fig. 8). A significantly higher effect in reference to standard drug dexamethasone (Fig. 7).
concentration of curcumin 2.02 μg/ml was measured in BALF of the
animals to which micelles were administered. However, for the other 4.3.2.3. Effect of curcumin on nitrite level. The nitrite levels are indica­
group, concentration of curcumin in the BALF was found to be 0.456 μg/ tive of the extent of nitric oxide present in the serum. The effect of
ml. Approximately, 4.4 fold higher concentration of curcumin was intranasally administrated curcumin-loaded micelles in regulating ni­
measured in the lungs indicating the efficacy of micelles in carrying the trite levels was examined and statistically analyzed by applying
drug to the lungs and also protecting it against degradation accounting repeated measure one way ANOVA with Newman-Keuls Multiple
for its higher concentration in the lungs. Comparision Test. Significantly higher nitrite levels were observed in
Group II and III asthmatic groups as compared to Control (group I).
4.3.2.2. Effect of curcumin on the generation of intracellular reactive ox­ Significant supression (p < 0.05) in the ] nitric oxide release to the
ygen species. ROS serves as a signaling molecule that could be directly extent of 15% and 37.7% after intransally administration of curcumin
linked to multiple cell functions and inflammation. The extent of regu­ suspension (Group IV) and curcumin micelles (Group V) respectively.
lation of ROS on intranasal administration of curcumin-loaded micelles However, comparable decrease in the nitrite levels were observed for

7
R. Chawla et al. Journal of Drug Delivery Science and Technology 67 (2022) 102922

Fig. 6. HPLC Chromatogram of curcumin released at 12 h from (a) cur-suspension (retention time: 1.295 min) and (b) curcumin micelles (retention time: 1.285 min)
analyzed at 425 nm.

Fig. 7. Plasma concentration-time profile of curcumin loaded micelles and cur-suspension.

Groups V and VI (37.7% and 40.7% respectively) (Fig. 10 and Table 4). 5. Conclusion
The ability of drug-loaded polymeric micelles to penetrate across the
epithelial cells is further strengthened by the presence of PEG which Curcumin is a pleiotropic herb which interacts with several targets
facilitates penetration across biological barriers tissue extracellular and controls the progression of disease. Of the various reported effects,
matrix, cellular barriers, and biological fluids such as mucus to reach the curcumin exhibits potent antioxidant and anti-inflammatory properties
inflamed site and decrease inflammation (as compared to non- and helps control inflammation especially in inflammatory diseases like
PEGylated formulations) [41,42]. asthma. The therapeutic potential of curcumin is limited by its poor

8
R. Chawla et al. Journal of Drug Delivery Science and Technology 67 (2022) 102922

Fig. 8. HPLC Chromatogram of curcumin analyzed in bronchoalveolar fluid on administration of (a) curcumin suspension (retention time: 1.285 min) and (b)
curcumin micelles (retention time: 1.285 min) at 425 nm.

Table 4
Percentage decrease in level of ROS and nitrite in different treatment groups.
S. Treatment % decrease in ROS level % decrease in Nitrite level
N. Group compared to the asthmatic compared to the asthmatic
group group

1. C-Susp 29.84 15
2. C-Mic 46.51 37.7
3. Dexamethasone 59.37 40.7

Fig. 9. Effect of different treatments on levels of ROS in BALF of rat. Exposure


to OVA increased ROS level in the BALF cells, and reduction of which was
observed on intranasal administration of curcumin micelles. The results are
shown as mean ± SEMs (ap<0.05 as compared to Control, b p < 0.05 as
compared to Group II, cp < 0.05 as compared to Group III, d p < 0.05 as
compared to Group IV and e p < 0.05 as compared to Group IV).

aqueous solubility, rapid metabolism, and quick degradation at acidic


pH. In one of our previous studies we have reported the effectiveness of
intranasally administered curcumin in asthma [7]. Loading of curcumin Fig. 10. Effect of different treatments on nitrite level in serum of rat. The
into micelles enhances the biopharmaceutical properties of curcumin. micelles produced effects similar to that of standard drug dexamethasone. The
Besides protecting it against degradation, the solubility of curcumin is results are shown as mean ± SEMs (ap<0.05 as compared to Control, b p < 0.05
as compared to Group II, cp < 0.05 as compared to Group III, and d p < 0.05 as
increased up to 40% due to micellar solubilization [43,44]. Also, mi­
compared to Group IV).
celles provide sustained release of the drug. The study shows that

9
R. Chawla et al. Journal of Drug Delivery Science and Technology 67 (2022) 102922

micellar curcumin shows significantly better attenuation of [19] L. Ji, J. Yunyun, W. Jun, F. Guorong, W. Yutian, Z. Chuan, A rapid and simple
HPLC method for the determination of curcumin in rat plasma: assay development,
OVA-induced chronic asthma in comparison to curcumin administered
validation and application to a pharmacokinetic study of curcumin liposome,
as suspension. Also, curcumin micelles showed significant attenuation in Biomed Chromatogr (2009 Nov) [cited 2020 Feb 26];23(11):1201–7. Available
generation of ROS and nitric oxide, similar to that obtained on treatment from: http://www.ncbi.nlm.nih.gov/pubmed/19488971.
of dexamethasone. Significantly higher concentration of curcumin was [20] W. Qamar, Technical considerations and precautions in in situ bronchoalveolar
lavage and alveolar infiltrating cells isolation in rats, Toxicol. Mech. Methods 25
observed in the BAL fluid when administered as micelles in comparison (7) (2015 Sep) 547–551.
to curcumin suspension. Overall, the micelles provide an efficient sys­ [21] R. Dworski, Oxidant stress in asthma, in: Thorax, BMJ Publishing Group, 2000,
tem for delivery of drug via intranasal route. p. S51.
[22] A.C. Pulla Reddy, B.R. Lokesh, Effect of dietary turmeric (curcuma longa) on iron-
induced lipid peroxidation in the rat liver, Food Chem. Toxicol. 32 (3) (1994)
Author declaration 279–283.
[23] G.S. Jeong, G.S. Oh, H.O. Pae, S.O. Jeong, Y.C. Kim, M.K. Shin, Y.S. Byeong, Y.
H. Sang, S.L. Ho, J.G. Jeong, J.S. Koh, H.T. Chung, Comparative effects of
The authors are aware of the modifications made in the revised curcuminoids on endothelial heme oxygenase-1 expression: ortho-methoxy groups
manuscript. are essential to enhance heme oxygenase activity and protection, Exp. Mol. Med.
38 (4) (2006 Aug) 393–400.
[24] D. Paola Rosanna, C. Salvatore, Reactive oxygen species, inflammation, and lung
Declaration of interest diseases, Curr. Pharmaceut. Des. 18 (26) (2012 Jul) 3889–3900.
[25] F. Dong, C. Wang, J. Duan, W. Zhang, D. Xiang, M. Li, Puerarin attenuates
The authors declare that they have no known competing financial ovalbumin-induced lung inflammation and hemostatic unbalance in rat asthma
model, Evidence-Based Complement Altern Med. 2014 (2014) 1–9.
interests or personal relationships that could have appeared to influence
[26] T. Hamaguchi, Y. Matsumura, M. Suzuki, K. Shimizu, R. Goda, I. Nakamura,
the work reported in this paper. The author(s) received no financial I. Nakatomi, M. Yokoyama, K. Kataoka, T. Kakizoe, NK105, a paclitaxel-
support for the research. incorporating micellar nanoparticle formulation, can extend in vivo antitumour
activity and reduce the neurotoxicity of paclitaxel, Br. J. Cancer 92 (7) (2005 Apr)
1240–1246.
References [27] D.S. Bredt, P.M. Hwang, S.H. Snyder, Localization of nitric oxide synthase
indicating a neural role for nitric oxide, Nature 347 (6295) (1990) 768–770.
[1] K.J.H. Jaclyn Quirt, Asthma. J Allergy Clin Immunol. 14 (Suppl 2) (2018) 57–70. [28] Q.W. Xie, H.J. Cho, J. Calaycay, R.A. Mumford, K.M. Swiderek, T.D. Lee, A. Ding,
[2] Chung KF. Role of Inflammation in the Hyperreactivity of the Airways in Asthma. T. Troso, C. Nathan, Cloning and characterization of inducible nitric oxide synthase
Vol. vol. 41, Thorax. BMJ Publishing Group; 1986. p. 657–662. from mouse macrophages, Science (80- ) 256 (5054) (1992 Apr) 225–228.
[3] J. Bienenstock, Immunology of the Lung and Upper Respiratory Tract, McGraw- [29] Q. Hamid, Springall, J. Polak, V. Riveros-Moreno, P. Chanez, J. Bousquet,
Hill, 1984, p. 414. P. Godard, S. Holgate, P. Howarth, A. Redington, Induction of nitric oxide synthase
[4] D. Proud, J. Sweet, P. Stein, R.A. Settipane, A. Kagey-Sobotka, M.H. Friedlaender, in asthma, Lancet 342 (8886–8887) (1993 Dec) 1510–1513.
L.M. Lichtenstein, Inflammatory mediator release on conjunctival provocation of [30] C.M. Prado, M.A. Martins, I.F.L.C. Tibério, Nitric oxide in asthma physiopathology,
allergic subjects with allergen, J. Allergy Clin. Immunol. 85 (5) (1990 May) ISRN Allergy 2011 (2011) 1–13.
896–905. [31] S. Lamas, P.A. Marsden, G.K. Li, P. Tempst, T. Michel, Endothelial nitric oxide
[5] P.S. Chauhan, D.K. Singh, D. Dash, R. Singh, Intranasal curcumin regulates chronic synthase: molecular cloning and characterization of a distinct constitutive enzyme
asthma in mice by modulating NF-ĸB activation and MAPK signaling, isoform, Proc. Natl. Acad. Sci. U. S. A. 89 (14) (1992 Jul) 6348–6352.
Phytomedicine 51 (2018) 1–16. [32] D.O. Moon, M.O. Kim, H.J. Lee, Y.H. Choi, Y.M. Park, M.S. Heo, G.Y. Kim,
[6] I. Stankovic, Curcumin, Chem Tech Assess 1 (8) (2004) 1–8. Curcumin attenuates ovalbumin-induced airway inflammation by regulating nitric
[7] Subhashini, P.S. Chauhan, S. Kumari, J.P. Kumar, R. Chawla, D. Dash, M. Singh, oxide, Biochem. Biophys. Res. Commun. 375 (2) (2008 Oct) 275–279.
R. Singh, Intranasal curcumin and its evaluation in murine model of asthma, Int. [33] S. Biswas, I. Rahman, Modulation of steroid activity in chronic inflammation: a
Immunopharm. 17 (3) (2013) 733–743. novel anti-inflammatory role for curcumin, in: Molecular Nutrition and Food
[8] M.L. Manca, J.E. Peris, V. Melis, D. Valenti, M.C. Cardia, D. Lattuada, E. Escribano- Research, vol. 52, Wiley-VCH Verlag, 2008, pp. 987–994.
Ferrer, A.M. Fadda, M. Manconi, Nanoincorporation of curcumin in polymer- [34] P. Nilani, N. Kasthuribai, B. Duraisamy, P. Dhamodaran, S. Ravichandran,
glycerosomes and evaluation of their in vitro-in vivo suitability as pulmonary K. Ilango, B. Suresh, Invitro antioxidant activity of selected antiasthmatic herbal
delivery systems, RSC Adv. 5 (127) (2015 Dec) 105149–105159. constituents, Ancient Sci. Life 28 (4) (2009 Apr) 3–6.
[9] L. Zhao, J. Du, Y. Duan, Y. Zang, H. Zhang, C. Yang, F. Cao, G. Zhai, Curcumin [35] K.M. Miranda, M.G. Espey, D.A. Wink, A rapid, simple spectrophotometric method
loaded mixed micelles composed of Pluronic P123 and F68: preparation, for simultaneous detection of nitrate and nitrite, Nitric Oxide - Biol Chem. 5 (1)
optimization and in vitro characterization, Colloids Surf. B Biointerfaces 97 (2012) (2001) 62–71.
101–108. [36] I.J. Fidler, A. Raz, W.E. Fogler, R. Kirsh, P. Bugelski, G. Poste, Design of liposomes
[10] Masoud J, Abdulla A, Tze-Fung Tan Y, Darwis Y. Rehydrated lyophilized to improve delivery of macrophage-augmenting agents to alveolar macrophages,
rifampicin-loaded mPEG-DSPE formulations for nebulization. Cancer Res. 40 (12) (1980 Dec) 4460–4466.
[11] K.K. Gill, A. Kaddoumi, S. Nazzal, PEG–lipid micelles as drug carriers: [37] M.N. Sahib, S. Abdalwahed, S.A. Abdulameer, Y. Darwis, K.K. Peh, Y.T.F. Tan,
physiochemical attributes, formulation principles and biological implication. https Solubilization of beclomethasone dipropionate in sterically stabilized phospholipid
://doi.org/10.3109/1061186x.2014.997735, 2015. nanomicelles (SSMs): physicochemical and in vitro evaluations, Drug Des. Dev.
[12] H. Gursahani, J. Riggs-Sauthier, J. Pfeiffer, D. Lechuga-Ballesteros, C.S. Fishburn, Ther. 6 (2012) 29–42.
Absorption of polyethylene glycol (PEG) polymers: the effect of PEG size on [38] N. Nishiyama, H. Takemoto, Polymeric micelles, Encycl Polym Nanomater 1–7
permeability, J Pharm Sci (2009 Aug) [cited 2020 Feb 26];98(8):2847–56. (2014).
Available from: http://www.ncbi.nlm.nih.gov/pubmed/19408293. [39] K. Kazunori, S.K. Glenn, Y. Masayuki, O. Teruo, S. Yasuhisa, Block copolymer
[13] M.N. Sibata, A.C. Tedesco, J.M. Marchetti, Photophysicals and photochemicals micelles as vehicles for drug delivery, J. Contr. Release 24 (1–3) (1993 May)
studies of zinc(II) phthalocyanine in long time circulation micelles for 119–132.
photodynamic therapy use, Eur. J. Pharmaceut. Sci. 23 (2) (2004 Oct) 131–138. [40] J. Li, Y. Jiang, J. Wen, G. Fan, Y. Wu, C. Zhang, A rapid and simple HPLC method
[14] A. Vonarbourg, C. Passirani, P. Saulnier, P. Simard, J.C. Leroux, J.P. Benoit, for the determination of curcumin in rat plasma: assay development, validation
Evaluation of pegylated lipid nanocapsules versus complement system activation and application to a pharmacokinetic study of curcumin liposome, Biomed
and macrophage uptake, J. Biomed. Mater. Res. Part A [Internet] (2006 Sep 1) Chromatogr (2009 Nov), https://doi.org/10.1002/bmc.1244 [cited 2020 Feb 26];
[cited 2020 Feb 26];78(3):620–8. Available from: http://www.ncbi.nlm.nih.gov/p 23(11):1201–7. Available from:.
ubmed/16779767. [41] J.S. Suk, Q. Xu, N. Kim, J. Hanes, L.M. Ensign, PEGylation as a strategy for
[15] M.N. Sahib, S. Abdalwahed, Abdulameer, Y. Darwis, K.K. Peh, Y.T.F. Tan, improving nanoparticle-based drug and gene delivery, in: Advanced Drug Delivery
Solubilization of beclomethasone dipropionate in sterically stabilized phospholipid Reviews, vol. 99, Elsevier B.V., 2016, pp. 28–51.
nanomicelles (SSMs): physicochemical and in vitro evaluations, Drug Des. Dev. [42] I.M. El-Sherbiny, N.M. El-Baz, M.H. Yacoub, Inhaled nano- and microparticles for
Ther. 6 (2012) 29–42. drug delivery, Glob Cardiol Sci Pract 2015 (1) (2015 Jan) 2.
[16] S. Warule Pooja, P.G.S.A.D. Patel Vipul, Development and validation of UV [43] S.M.H. Rahman, T.C. Telny, T.K. Ravi, S. Kuppusamy, Role of Surfactant and pH in
spectrophotometric method for the estimation of curcumin in an ayurvedic Dissolution of Curcumin, Indian J Pharm Sci [Internet, 2009 [cited 2020 Sep 5];71
formulation haridrakhand, Int J Pharm DRUG Anal 5 (5) (2017) 193–197. (2):139–42. Available from: www.ijpsonline.com.
[17] K.K. Gill, S. Nazzal, A. Kaddoumi, Paclitaxel loaded PEG5000-DSPE micelles as [44] S. Rani, S. Mishra, M. Sharma, A. Nandy, S. Mozumdar, Solubility and stability
pulmonary delivery platform: formulation characterization, tissue distribution, enhancement of curcumin in Soluplus® polymeric micelles: a spectroscopic study,
plasma pharmacokinetics, and toxicological evaluation, Eur. J. Pharm. Biopharm. J Dispers Sci Technol (2020 Mar 20) [cited 2020 Sep 5];41(4):523–36. Available
79 (2) (2011 Oct) 276–284. from: https://www.tandfonline.com/doi/abs/10.1080/01932691.2019.1592687.
[18] S. Ugwu, A. Zhang, M. Parmar, B. Miller, T. Sardone, V. Peikov, I. Ahmad,
Preparation, characterization, and stability of liposome-based formulations of
mitoxantrone, Drug Dev. Ind. Pharm. 31 (2) (2005) 223–229.

10

You might also like