You are on page 1of 12

Colloids and Surfaces B: Biointerfaces 205 (2021) 111838

Contents lists available at ScienceDirect

Colloids and Surfaces B: Biointerfaces


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

Systematically designed chitosan-coated solid lipid nanoparticles of ferulic


acid for effective management of Alzheimer’s disease: A
preclinical evidence
Sumant Saini a, Teenu Sharma a, Atul Jain b, Harmanjot Kaur a, O.P. Katare a,
Bhupinder Singh a, b, *
a
University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Studies, Panjab University, Chandigarh, 160014, India
b
UGC-Centre of Excellence in Applications of Nanomaterials, Nanoparticles and Nanocomposites (Biomedical Sciences), Panjab University, Chandigarh, 160014, India

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

Keywords: Ferulic acid (FA) is a ubiquitous natural plant bioactive with distinctive promise in neurodegenerative disorders.
Chitosan However, its therapeutic efficacy gets compromised owing to its poor aqueous solubility, inadequate perme­
Lipid nanoparticles ability across lipophilic barriers, and extensive first-pass metabolism. The current studies, therefore, were un­
Neurodegenerative disease
dertaken to systematically develop chitosan-coated solid lipid nanoparticles (SLNs) using QbD paradigms for
Intranasal delivery
improved efficacy of FA in the management of Alzheimer’s disease (AD). SLNs of FA were formulated employing
Quality by Design (QbD)
Blood-brain barrier Compritol as lipid and polysorbate 80 as surfactant and optimised using a 32 Central Composite Design (CCD).
The optimized formulation, surface-coated with chitosan using ionic gelation, exhibited particle size of 185 nm,
entrapment efficiency of 51.2 % and zeta potential of 12.4 mV. FTIR and DSC studies verified the compatibility of
FA with formulation excipients, PXRD construed significant loss of drug crystallinity, while FESEM depicted
existence of uniform spherical nanoparticles with little aggregation. Notable improvement in ex vivo mucoad­
hesion and permeation studies using goat nasal mucosa, coupled with extension in in vitro drug release, was
obtained with SLNs. Substantial improvement with SLNs in cognitive ability through the reduction in escape
latency time during behavioural studies, together with significant improvement in various biochemical param­
eters and body weight gain was observed in AD-induced rats. Histopathological images of different rat organs
showed no perceptible change(s) in tissue morphology. Overall, these preclinical findings successfully demon­
strate improved anti-AD efficacy, superior nasal mucoadhesion and permeation, extended drug release, improved
patient compliance potential, safety and robustness of the developed lipidic nanoconstructs of FA through
intranasal route.

1. Introduction 16 million by 2050 [4].


Among the myriad treatment strategies currently adopted for man­
Alzheimer’s disease (AD), one of the most prevalent neurodegener­ agement of AD, the primary ones include cholinesterase inhibitors and
ative disorders, affects almost 3% of the world’s population [1]. Char­ NMDA receptor antagonists. A multitude of factors, such as inadequate
acterized by serious but gradual diminution in cognitive functions, and inconsistent drug oral bioavailability, owing to their low aqueous
incidences of AD get markedly augmented among the aged population of solubility, poor permeability across the biological membranes and high
over 65 years, thereby posing a huge health and economic burden on the first-pass metabolism, together with poor brain penetration and reten­
society [2]. Neuronal degeneration in AD results in substantial loss of tion, need for repeated dosing, and high frequency of side effects, tend to
memory, communication skills, judgment and reasoning [3]. Albeit the limit the optimum usage of such drugs [5,6]. This calls for an exploration
prevalence of AD has been rising all across, the number of people of newer therapeutic strategies for the effective management of AD [7,
inflicted with AD in the USA alone is expected to range between 11 and 8].

* Corresponding author at: University Institute of Pharmaceutical Sciences, National UGC Centre for Excellence in NanoBiomedical Applications, Panjab Uni­
versity, Chandigarh, 160 014, India.
E-mail addresses: bsbhoop@yahoo.com, bsbhoop@pu.ac.in (B. Singh).

https://doi.org/10.1016/j.colsurfb.2021.111838
Received 21 January 2021; Received in revised form 28 April 2021; Accepted 8 May 2021
Available online 14 May 2021
0927-7765/© 2021 Published by Elsevier B.V.
S. Saini et al. Colloids and Surfaces B: Biointerfaces 205 (2021) 111838

Various plant bioactives, in this regard, have been documented to used, throughout the current studies, were of analytical grades(s).
possess anti-AD potential, which include, quercetin [9], morin [10],
apigenin [11], kaempferol [12], rutin [13], ferulic acid (FA) [14],
berberine [15], curcumin [16] and resveratrol [17]. FA, 3-hydroxy 2.2. Methods
cinnamic acid, one of such promising bioactive explored, is abun­
dantly found in various fruits and vegetables [18]. It is reported to 2.2.1. Selection of lipids and surfactants for formulation
possess antioxidant, anticancer, cardioprotective and neuroprotective Solubility studies on FA were conducted in various lipids, viz.,
potential [19,20]. However, the therapeutic efficacy of FA gets thwarted Compritol, stearic acid, Dynasan, GMS, Inwitor and Precirol, and sur­
by its poor aqueous solubility (i.e., soluble only in hot water) and factants, viz., Transcutol, Tween 40, Tween 80, Span 80, Gelucire 39/01,
inability to traverse the lipophilic barriers (with log P of 0.78), it being a Brij and poloxamer 188, to select apt excipients for the formulation of
potentially BCS class IV molecule [21–23]. Moreover, extensive SLNs. Briefly, the lipids were heated to a temperature just above their
first-pass metabolism of FA further diminishes its efficacy [24]. respective melting points, using a thermostatically-controlled water
Nanostructured systems have demonstrated stellar drug delivery bath (Riviera Glass Pvt. Ltd., Mumbai, India). An excess of FA was added
merits like high drug loading, enhanced stability and extended disso­ to each flask and shaken overnight. The samples were then withdrawn,
lution profiles. The lipidic nanocarriers, further, come with several ad­ centrifuged (10 min) and the concentration of FA was determined in the
vantages like controlled release, improved permeability across supernatant, following apt dilution, using a UV visible spectrophotom­
biological barriers, excellent biocompatibility, improved drug stability, eter (Jenway-6315, Cole-Parmer, Staffordshire, UK) at 322 nm.
high encapsulation, ease to scalability, sterilizability, and augmented
oral bioavailability of bioactive compounds [25,26]. Solid lipid nano­ 2.2.2. Preparation of SLNs
particles (SLNs), in this regard, are considered promising lipidic nano­ The SLNs were prepared using hot homogenization technique [36].
carriers. However, these are easily distinguishable by the immune The lipid (Compritol) was heated above its melting point (i.e., 70–75
system, due to their hydrophobicity and surface negativity, which

C). FA, dissolved in ethanol, was added to it until it turned transparent.
determine their opsonization and subsequent clearance. Surface func­ The aqueous phase, comprising of water and Tween 80, was mixed to
tionalization of nanoparticles with polymers like chitosan, therefore, obtain a clear solution and heated. Under isothermal conditions, the
induces a positive charge, decreases opsonization and enhances their lipidic phase was added drop-wise to the aqueous phase, employing a
residence time [27–29]. Various routes of administration have been high-speed homogenizer (IKA® T18 digital Ultra Turrax®, Washington,
experimented for delivery of the therapeutic agents to the brain. Intra­ USA) until a clear homogenous emulsion was obtained. The dispersion
nasal administration, in this regard, has proved to be a quite promising was probe-sonicated (Misonix Sonicator® 3000, Farmingdale, NY, USA)
route, as it not only bypasses the blood-brain barrier (BBB) and delivers and then stirred on a magnetic stirrer (Remi, Mumbai, India) at 2000
drug molecules directly to the brain via olfactory and transgeminal rpm for 1 h to allow solvent diffusion from the lipid phase [37,38]. The
pathways, but also circumnavigates the hepatic first-effect, resulting in dispersion was then lyophilized (Alpha, 2–4 LD plus, Martin Christ,
rapid onset of drug action [30,31]. Chitosan, in this context, is consid­ Osterode am Harz, Germany) and stored in glass vials until further use
ered an ideal polymer owing to its promising mucoadhesive character­ [39,40].
istics, high biocompatibility and non-toxic, and above all, its ability to
induce reversible opening of tight junctions in the nasal epithelial cells 2.2.3. Systematic development of SLNs
[32,33]. Chitosan coating of SLNs is generally carried out using elec­
trogelation technique owing to its myriad benefits like uniform coating, 2.2.3.1. Formulation objectives. According to the established QbD par­
high entrapment efficiency (being water-based), safety and adigms, a quality target product profile (QTPP) was outlined enumer­
cost-effectiveness [34,35]. ating variegated desirable quality traits of FA-loaded SLNs to achieve
The current studies, accordingly, report our successful endeavours to maximal efficacy for therapeutic management of AD, as delineated in
deliver FA across BBB using chitosan-coated SLNs. The lipidic nano­ Table S1. Appropriate justifications envisioned for the selection of
constructs were explored vis-a-vis pure FA employing a battery of in vitro various quality attributes (CQAs) are outlined in Table S2.
evaluation studies like particle size, zeta potential, entrapment effi­
ciency and drug release; diverse spectroscopic, calorimetric and X-ray 2.2.3.2. Risk estimation studies. As elementary risk(s) assessment is
diffractometric studies; ex vivo mucoadhesive strength and permeability pivotal during systematic development of SLNs, an Ishikawa fishbone
studies on goat nasal mucosa; and finally, for their anti-Alzheimer effi­ diagram was conceptualized highlighting cause-effect relationships be­
cacy and safety potential through variegated in vivo investigations like tween key material and process attributes with various CQAs, using
behavioural, biochemical, brain level and histopathological studies in Minitab® 17 software (Minitab, LLC, USA). Further, a risk estimation
the disease-induced rats. Nanostructured systems were systematically matrix (REM) was established, classifying these input factors among
designed using Quality by Design (QbD) approach, encompassing initial low, medium, and high-risk parameters, based upon the risk(s) allied
risk assessment, factor screening and optimization using response sur­ with each factor [37,39,41].
face mapping (RSM).
2.2.3.3. Factor screening studies. For selecting “influential few factors”
2. Materials and methods from a plethora of variables, a 7-factors, 8 run Taguchi design (TgD),
operating at low (-1) and high (+1) levels, was employed. Pareto and
2.1. Materials half-normal plots, thus obtained, were analysed to discern significant
variables using Design-Expert® 12 software (StatEase Inc., Minneapolis,
Compritol 888 ATO, Transcutol and Gelucire 39/01 were obtained ex USA). Table S3 illustrates the design matrix for all the prepared for­
gratis from M/s Gattefosse, France. Dynasan 114, Imwitor 960 K and mulations investigated, along with their factor levels [41].
Precirol ATO were obtained ex gratis from M/s Cremer Oleo GmbH & Co,
Germany. FA, Tween 20, Tween 40, Tween 80, Span 80, Brij 35, sodium 2.2.3.4. Systematic optimization of SLNs. Statistically significant factors,
triphosphate (STPP) and poloxamer 188 were purchased from M/s SRL selected from screening studies, were optimized using a Central Com­
Chemicals, India. Stearic acid and glyceryl monostearate (GMS) were posite Design (CCD) for 2 factors, accounting for 13 experimental trials,
purchased from M/s SD Fine Chemicals, India and Fischer Scientific, including quintuplicate runs around the centre point (0,0), as illustrated
India, respectively. Chitosan (medium grade) was purchased from M/s in Table 1. The model fitness was analysed by computing the magnitudes
HiMedia Laboratories Pvt Ltd, India. All the solvents and chemicals of the probability of type I error (i.e., p-value) and correlation coefficient

2
S. Saini et al. Colloids and Surfaces B: Biointerfaces 205 (2021) 111838

Table 1 temperature (◦ C) [51].


Design matrix as per a 2-factor Central Composite Design for optimization of the
prepared SLNs formulations of ferulic acid, along with employed factor levels in 2.2.4.5. Powder X-ray diffraction (PXRD). PXRD studies were conduct­
coded and actual form. ed for lyophilized SLNs and cSLNs using X’Pert PRO® diffractometer
Run A: Lipid B: Surfactant system (PANalytical, Netherlands) employing Cu-Ka radiation at 1.5406
1 0 1 Å at an accelerating voltage of 45 kV. Samples, packed in aluminum pan,
2 1 1 scanned between 4 and 50◦ values of 2θ [52,53].
3 1 0
4 0 0
5 1 − 1
2.2.4.6. Drug entrapment efficiency (EE). The EE of coated and uncoated
6 0 0 SLNs was measured as per a method reported previously [36]. Aliquots
7 0 − 1 (2 mL) were centrifuged (ThermoFisher Scientific, Legend™ XTR Sor­
8 − 1 − 1 vall™ USA) at 12,000 rpm (16128xg) for 30 min at 4 ◦ C. The superna­
9 − 1 0
tant was then pipetted, filtered and suitably diluted for the
10 0 0
11 0 0 quantification of unentrapped FA using UV–vis spectrophotometer at
12 0 0 322 nm. The amount of entrapped FA was calculated using Eq. 1.
13 − 1 `1
Total amount of FA added − Free FA in supernatant
Factors Low(-1) Medium(0) High(+1) EE = × 100 (1)
Total amount of FA added
Lipid (mg) 150 275 400
Surfactant (g) 1.50 2.75 4.00 2.2.4.7. Drug release studies. Dialysis sac technique was used to study
the in vitro release behaviour of FA from the formulations for a period of
(i.e., R). Factor-response relationship(s) were graphically represented as 48 h, vis-à-vis pure drug suspended in sodium carboxymethyl cellulose
3-D response surfaces and corresponding 2-D contour plots for each of solution (0.25 %, w/v). The dissolution was performed in phosphate
the CQAs [42]. buffer pH 6.5 (50 mL) at 37 ± 0.5 ◦ C, 100 rpm. SLNs (1 mg of FA), were
placed in the dialysis sac (Molecular weight cut-off of 12 kDa), and
sample aliquots (2 mL) were removed at pre-scheduled time-intervals.
2.2.3.5. Coating of optimized SLNs. The optimized SLNs dispersion was
An equal volume of fresh dissolution medium was replenished and the
further coated with chitosan employing ionic gelation technique [29,35,
dissolution data were corrected for losses of drug and/or drug release
43,44]. Briefly, SLNs was added drop-wise into an equal volume (10 mL)
volume occurred during sampling, and cumulative percent of FA release
of chitosan solution (0.175 %, w/v) under stirring (25 ◦ C, 20 min).
was computed likewise [54].
Subsequently, ionic crosslinker, STPP (4.5 mL of 0.5 %, w/v), was added
drop-wise and stirred further at 600 rpm for 30 min, to ensure uniform
coating. Chitosan-coated SLNs (cSLNs) dispersion was lyophilized and 2.2.4.8. Determination of mucoadhesive strength. Goat nasal mucosa was
stored, until further use [28,33,34,45,46]. The chitosan coating effi­ procured from a local slaughter-house (Nayagaon, Chandigarh). The
ciency in the current studies was determined as per a previously re­ mucosa was washed with normal saline solution to remove adherent
ported method [47]. connective tissues and stored in simulated nasal fluid (SNF), pH 6.5, till
tested for its mucoadhesive strength, for at least 12 h, as per a method
2.2.4. Characterization of optimized and cSLNs reported previously [55,56] on TA-XT plus Texture Analyser (Stable
Micro Systems, Godalming, UK) in an adhesive test mode.
2.2.4.1. Particle size and zeta potential. Mean values of particle size
(Snm) and zeta potential (ZP) for optimized SLNs and cSLNs were 2.2.4.9. Ex vivo permeability studies. Ex vivo nasal mucosal permeation
determined by dynamic light scattering at an angle of 90◦ employing was determined on Franz diffusion cell (PermGear Inc., Pennsylvania,
Zetasizer® (NANO ZS-90, Malvern, Worcestershire, UK). Prior to the USA) [55]. SLNs and cSLNs, dispersed in 1 mL of SNF, pH 6.5 (5 mL),
analysis, the samples were suitable diluted [48]. were added to donor compartment, which was fitted and clamped onto
the receptor compartment containing fresh SNF. Periodically, the sam­
ples were withdrawn and suitably replaced with fresh medium.
2.2.4.2. Field emission scanning electron microscopy (FESEM). The sur­
Isothermal conditions and physiological temperature were maintained
face topology of the SLNs was studied using FESEM (8010, Hitachi,
through a water jacket system. The samples were quantified using an
Japan). The sample was analysed at a working gap of 7.2 mm, and im­
HPLC method, previously reported by us [57].
ages were obtained at a suitable accelerating voltage of 5.0 kV [49].
FA permeating per unit surface area (A/S) was plotted against time
(t). The flux (J) was computed from the graph’s slope for all the com­
2.2.4.3. Fourier-transform infrared spectroscopy (FTIR). The drug-
binations using Eq. 2 [58,59].
excipient interactions were investigated using FTIR analysis. The sam­
ples for FTIR analysis was prepared by finely grinding the sample with A Pcoff Atot h2
= [t − ] (2)
KBr which was then compressed into pellets. The spectra were recorded S V 6D
on an FTIR spectrophotometer (Perkin Elmer, MA, USA), and were then
interpreted for changes in the spectral peaks [50]. Where, Atot, V, h and D depict the quantity of FA, volume of donor cell,
thickness of dialysis membrane, and diffusion coefficient, respectively.
2.2.4.4. Differential scanning calorimetry (DSC). DSC analysis of FA and Herein, PcoffAtot/V, delineates the slope (J), while, h2/6D, denotes the
its blend with each of the excipients was carried out by triturating them abscissa intercept, furnishing the value of lag time.
with a pestle in mortar. Samples, 3–5 mg each, were weighed in a
Tzero™ aluminum pan, which was heated at a rate of 10 ◦ C/min. Three 2.2.4.10. Stability studies. The lyophilized formulations, packed in air-
runs were conducted for each sample with nitrogen flow of 20 mL/min. tight glass vials were evaluated for any significant alteration(s) in
The eventual thermograms of FA and physical mixture were analysed drug assay and other CQAs at 5 ± 2 ◦ C, 25 ± 2 ◦ C and 60 ± 5% RH, 40 ±
using Star and Universal, while that of SLNs using DSC-TA Q20 (all by 2 ◦ C and 75 ± 5% RH (ICH guidelines Q1(R2)). The formulation was
M/s TA instruments, New Castle, DE). Appropriate scans were recorded, evaluated for a storage period of 3 months, at monthly intervals, for
and the corresponding plots were obtained between heat flow (W/g) and various parameters to establish stability of surface-modified colloidal

3
S. Saini et al. Colloids and Surfaces B: Biointerfaces 205 (2021) 111838

system, viz., % assay, particle size, EE, ZP, % release and f2 values. Table 2
Constitution of various animal treatment groups for pharmacodynamic studies
2.2.4.11. In vivo studies in rats. To investigate the in vivo efficacy of of ferulic acid and its SLNs formulations in rat (n = 3).
optimized SLNs, the formulations were tested on Wistar rats (180− 200 GROUP TREATMENT DESCRIPTION
g), as per the protocols approved by Panjab University Institutional 1 Naïve Animals receiving normal saline orally.
Animal Ethics Committee (vide Letter No. PU/45/99/CPCSEA/IAEC/ 2 Control /sham Animals injected with bilaterally
2019/242). Animals were obtained from Central Animal House of Uni­ intracerebroventricular (ICV)-artificial
versity, housed in animal cages, and acclimatized for 7 days with a 12 h cerebrospinal fluid
3 Positive control /test Animals injected with ICV-STZ bilaterally
day-and-night cycle (27 ± 3 ◦ C and 55 ± 5% RH), before the conduct of 4 FA Animals injected with ICV-STZ bilaterally along
studies. Rats were fed with standard diet along with drinking water, ad with administration of pure drug suspension,
libitum. intranasally
2.2.4.11.1. Drug treatment and dosing schedule. For pharmacody­ 5 FA SLN i.n. Animals injected with ICV-STZ bilaterally along
with administration of optimized formulation,
namic evaluation of FA and its formulations, animals were distributed
intranasally
randomly into 8 groups, each comprising of three rats, as described in 6 CS-coated optimized ICV-STZ bilaterally along with administration of
Table S4. AD was induced in rats by intracerebroventricular (ICV) in­ FA SLNs i.n. chitosan-coated optimized SLNs of FA,
jection of streptozocin (STZ) [60,61]. The drug was administered via intranasally
oral and intranasal route. All the experimental studies were carried out 7 FA suspension p.o. ICV-STZ bilaterally and FA administered peroral
suspension in 0.25 %w/v sodium
for 28 days (STZ); 3 mg/mL, FA; 80 mg/kg.
carboxymethylcellulose solution
2.2.4.11.2. Induction of AD. Animals were anaesthetized with 8 CS-coated optimized ICV-STZ bilaterally along with administration of
intraperitoneally injected ketamine (80 mg/kg) and xylazine (10 mg/ FA SLNs p.o. optimized SLNs formulation perorally
kg) on a stereotaxic apparatus. Following midline sagittal incision in rat
skull, holes were drilled for Hamilton syringe into the lateral ventricle
microscopical [71].
(0.8 mm on the coronal axis (C), 1.5 mm on sagittal axis (S), and 3.6 mm
horizontal axis (H); from the zero-reference point (Bregma)). Artificial
cerebrospinal fluid and/or STZ were infused into the rat brain [62]. The 3. Results and discussion
incision was then sutured, dental fixer and neosporin were applied. Rats
were allowed access to water and food ad libitum. 3.1. Selection of formulation lipids and surfactants
2.2.4.11.3. Behavioural studies. 2.2.4.11.3.1. Spatial navigation and
learning Selection of lipids and surfactants was made based on their solubi­
Improvement in the cognitive ability of AD-induced rats after treat­ lization potential for FA. As inferred from the solubility profiles of FA in
ment with developed SLNs was analysed using previously reported various lipids (Figure S1), the trend observed was Compritol 888ATO >
behavioural tests [63]. Morris water maze (MWM) studies were con­ stearic acid > Dynasan 114 > Imwitor 960 K > Precirol ATO > glyceryl
ducted to assess animal behaviour in each group. A large circular pool, monostearate, whereas the solubility of FA in various surfactants was
with dimensions of 150 × 45 × 30 cm, was filled with water. The pool found to follow the order as Tween 80 > Gelucire 39/01 > Brij 35 >
was partitioned into 4 equal quadrants, and a target quadrant was Tween 20 > Tween 40 > poloxamer 188 > Transcutol > Span 80. Thus,
selected. A submerged platform was placed in the pool, and the rats were Compritol and Tween 80 were selected as the solid lipid and surfactant,
trained in the acquisition phase at intervals of 30 s. Acquisition index or respectively, for subsequent development of SLNs.
escape latency time (ELT) was noted to locate the hidden platform in
water maze. Successively, the probe trial was performed wherein the 3.2. Postulation of QTPP and CQAs
spatial memory was consolidated by removing the platform 24 h after
the last training session, and the time spent by the subject in target Table S1 outlines the QTPP, based on brainstorming and literature
quadrant (TSTQ) was determined. search, enumerating various intended quality characteristics to achieve
2.2.4.11.3.2. Biochemical assessment desired efficacy and safety of FA-loaded SLNs. Table S2 delineates
Various biochemical markers were estimated as per standard pro­ suitable justifications for selecting various CQAs of the proposed
tocols in supernatant, viz., lipid peroxidation (LPO) [64] and the levels nanocarriers.
of glutathione (GSH) [65], nitrite [66], protein [67], superoxide dis­
mutase (SOD) [68], and acetylcholinesterase (AChEs) [69]. Brains har­ 3.3. Risk estimation studies
vested after euthanasia (cervical dislocation) were rinsed with chilled
isotonic saline and homogenized. The homogenates were centrifuged at An Ishikawa fishbone diagram, delineating various plausible factors
10,000 rpm (11,200xg) for 10 min at 4 ◦ C. The data were analysed using impacting the chosen CQAs, is portrayed in Figure S2. Further, a risk
one-way ANOVA at p < 0.05 to infer any statistically significant change estimation matrix (REM) (Figure S3) was constructed. As input vari­
in the biochemical parameters among various treatment groups. ables like amount of lipid (mg), amount of surfactant (g), homogeniza­
2.2.4.11.3.3. Estimation of brain levels tion time (min), homogenization speed (rpm), probe sonication time
Rats were randomly divided into 3 treatment groups (Table 2), each (min), stirring speed (rpm), and suspension stirring time (h) were
with 3 animals. The supernatant (2 mL; as per the procedure mentioned associated with medium to high risk; these were further prioritized using
in Section 2.2.4.11.3.2.) was mixed with 2 mL of acetonitrile [70]. The factor screening investigations.
solution was then centrifuged at 10,000 rpm (11,200 x g) for 5 min, and
the supernatant was analysed by our previously reported HPLC method 3.4. Factor screening studies
[57].
Table 2 Figure S4 portrays half-normal and Pareto charts obtained during
2.2.4.11.3.4. Histopathological studies factor screening studies using TgD. TgD was selected for factor
Various organs like brain, lung, heart, kidney and liver were isolated, screening, it being an efficient first-order experimental design, since
fixed in formalin and subjected to histopathological examinations. Tis­ only the main effects were to be identified and studied [72]. Amounts of
sue specimens were fixed in paraffin block, sectioned, sliced (5 μm lipid and surfactant were found to significantly (p < 0.05 each) impact
thick), stained with hematoxylin and eosin, and observed under optical various CQAs; hence were chosen as the critical material attributes
(CMAs). Multiple linear regression analysis was performed using Eq. 3,

4
S. Saini et al. Colloids and Surfaces B: Biointerfaces 205 (2021) 111838

wherein the main effects were only considered. subsequently optimized using a CCD, as it could unearth intricate factor-
response relationship(s) as well as various possible factor interaction(s)
Y = β0 + β1X1 + β2X2 + β3X3 + β4X4 + β5X5 + β6X6 + β7X7 (3) [72,73]. Data were fitted into a second-order quadratic model,
Here, β0 represents the intercept, and β1− 7 are the coefficients for linear comprising of main effects, interaction effects and quadratic terms, as
terms for factors X1− 7, respectively. As observed from the Pareto charts represented in Eq. 4.
and half-normal plots, the amounts of lipid and surfactant had the t Y = β0 + β1X1 + β2X2 + β3X1X2 + β4X21 + β5X22 (4)
values higher than their respective threshold limits for all the CQAs
analysed, viz., EE, Snm and ZP. In the aforesaid equation, β0 represents the intercept, β 1 and β2 as the
coefficients of linear terms X1 and X2 respectively, β3 as the coefficient of
interaction term, and β4 and β5 as the coefficients of respective quadratic
3.5. Systematic optimization studies on SLNs terms X21 and X 22, respectively. Table S5 enlists the magnitudes of various
model coefficients, along with the ANOVA parameters.
The two influential CMPs, viz., amounts of lipid and surfactant, were

Fig. 1. 3-D response surface plots and corresponding 2-D contour plots for various CQAs; Particle size (A,B); Zeta potential (C,D); Entrapment efficiency (E,F); and
Drug Release (G,H).

5
S. Saini et al. Colloids and Surfaces B: Biointerfaces 205 (2021) 111838

The 3-D RSM plot and the corresponding 2-D RSM plot for Snm (Fig. 1 in the vibrational frequency of –C– – O group stretch in the FTIR spectra
a and b, respectively) revealed that at low levels of surfactant, Snm tends of the physical mixture of FA with the selected excipients [76]. This
to increase with increasing lipid levels. However, at high levels of sur­ indicates that the selected excipients were quite compatible with FA and
factant, Snm values tend to decrease. These observations indicate the can be employed, in combination, for further studies.
presence of a sufficient amount of surfactant to reduce the interfacial
tension with increasing amounts of lipid, thus leading to improved sta­ 3.6.3. DSC studies
bilization of the nanoparticulate dispersion. Also, at high levels of lipid, The DSC thermogram of FA depicts a single characteristic endo­
it was observed that increasing the surfactant amount tended to reduce thermic peak at 176.58 ◦ C (Figure S8A). The thermograms of physical
Snm. mixtures of FA with Compritol (Figure S8B), Tween 80 (Figure S8C),
The 3-D RSM plot and the corresponding 2-D RSM plot for ZP, as and Compritol and Tween 80 together (Figure S8D), portray no new
depicted in Fig. 1c and d, respectively, show that increasing the levels of peak(s), along with an intact peak of FA, thus refuting any plausible
lipid leads to a rise in ZP values, followed by a decline in their values at distinct interaction(s) between FA and chosen excipients. The physical
the higher levels. In contrast, at higher levels of lipid, on increasing the mixture(s) showed characteristic peaks of the drug, which were absent
amount of surfactant, ZP tends to decrease, potentially ascribable to the in the DSC thermogram of formulation, plausibly attributable to the
non-ionic character of Tween 80 as the surfactant. However, at low entrapment of FA into the formulation (Figure S8E). The melting point
levels of lipid, this increasing trend in ZP is observed to be less peaks were found to be broadened, reduced in intensity, and slightly
pronounced. shifted from their actual melting points, ascribable to the lower
The 3-D RSM plot for % EE is depicted in Fig. 1e and f, respectively. It enthalapy and the plasticizing effect of the surfactants and co-
shows a curvilinear interactive and declining trend in % EE values at surfactants, respectively, in consonance with various literature reports
lower and higher levels, though an increasing trend is observed at their [77,78]. Further, the endothermic peak of FA in SLNs indicates the
intermediate levels. presence of FA in stable crystalline form in the lipidic matrix of the
Fig. 1 (g and h) illustrate the influence of the levels of lipid and formulation.
surfactant on the values of % RelFA. The 3-D RSM plot (Fig. 1 g) portrays
a linear descent in the magnitude of % RelFA at all the levels of lipid, 3.6.4. PXRD studies
indicating sustained kind of drug release of the nanoconstructs. Analo­ Figure S9 illustrates PXRD patterns of FA (a), lyophilized SLNs (b)
gous information can also be deduced from the corresponding 2-D and cSLNs (c). FA exhibits sharp peaks at 2θ scattered angles of 9.04,
contour plot (Fig. 1h). However, a rise in % RelFA is observed on 12.83, 15.64, 25.97, 26.43 and 29.48, which are characteristic of its
increasing Tween 80, apparently owing to its emulsification crystalline nature. However, these peaks were conspicuously absent in
characteristics. lyophilized formulations (Figure S9(B and D)), vividly indicating the
RSM facilitated establishing relations(s) among the CMAs and CQAs, loss of crystallinity in the corresponding lipidic nanoconstructs.
chiefly using these 3-D and 2-D RSM plots. Design space, as a definitive
outcome of graphical optimization, demarcated the composition of the 3.6.5. FESEM studies
optimum formulation of prepared nanocarriers [36,37]. The FESEM images of optimized SLNs (Figure S10 (a and b)) and
cSLNs (Figure S10 (c and d)) indicate their uniform spherical geometry
3.5.1. Search for the optimum solution with minuscule aggregation. However, surface coating was found to
Figure S5 portrays the overlay plot and location of the optimal introduce irregularities on surface of SLNs, thereby increasing the
formulation within the design space. The optimized SLNs formulation effective contact surface area, which could plausibly enhance its nasal
comprised of Compritol (180 mg) and Tween 80 (2.25 g), with Snm of mucoadhesion.
181.44 nm and ZP of -2.72 mV. The EE of optimized formulation was
found to be 38.66 %, while cumulative % RelFA at 12 h was 74.01 %. 3.6.6. Drug entrapment studies
The system exhibited EE of 38.92 and 51.18 % for uncoated and
3.5.2. Coating of SLNs with chitosan coated optimized SLNs, respectively. Lower values of EE can be ascribed
The chitosan coating efficiency of cSLNs was found to be 61.7 %, to the hydrophilic nature of molecule and reduced entrapment of FA in
quite in concurrence with literature [47]. the lipidic coat [74]. Augmented EE values of cSLNs, however, may be
ascribed to the presence of chitosan coating, which could have pre­
3.6. Characterization studies on optimized SLNs and cSLNs vented leaching out of FA from the SLNs [79].

3.6.1. Particle size and zeta potential studies 3.6.7. In vitro drug release studies
As illustrated in Figure S6 (a and c), Snm of SLNs and cSLNs was Fig. 2 portrays nearly sustained-release profile for the SLNs. Only 19
observed to be 182.6 and 184.9 nm, respectively, while the corre­ % of FA was observed to be released within 12 h in its naïve form,
sponding values of ZP were -2.63 and 12.4 mV (Figure S6 (b and d)), whereas encapsulating FA in SLNs and subsequent chitosan coating
respectively. The charge on ZP values got altered from negative to significantly improved its release to 72 % and 64 %, respectively, in 12
positive, following chitosan coating, apparently owing to cationic nature h. Drug release kinetic modelling using Korsmeyer-Peppas model fur­
of the polymer. Though ZP got augmented in magnitude considerably nished the release exponent values of 0.392 and 0.454 for optimized
with cSLNs, yet it was not adequate to prevent agglomeration; hence the uncoated and coated SLNs, indicating that FA release followed Fickian
formulation was lyophilised [74,75]. and quasi-Fickian mechanisms, respectively. Encapsulating FA in SLNs
improved in vitro dissolution performance, which is quite evident from
3.6.2. FTIR studies the enhanced rate and extent of dissolution. Chitosan coating impeded
The vibrational frequency of the − OH group stretch of FA is observed dissolution profile vis-à-vis uncoated optimized SLNs, which may be
at 3437.48 cm− 1 (Figure S7A) of Supplementary Information). The attributed to the formation of a barrier layer of chitosan around the
FTIR spectra of physical mixtures of FA with Compritol (Figure S7B), SLNs, thereby increasing the effective path length for FA to get released
Tween 80 (Figure S7C), individually, and with both Compritol and into the dissolution medium vis-à-vis the uncoated SLNs dispersion.
Tween 80 (Figure S7D) depict no appreciable change in the charac­ Hence, the rate, as well as the extent of dissolution, was observed to be
teristic vibrational frequency of the –OH group stretch of FA, indicating comparatively lowered, following chitosan coating, indicating relatively
the absence of any interaction. The vibrational frequency of –C– –O superior controlled release characteristics of cSLNs.
group stretch of FA was observed at 1689.26 cm− 1, delineating no shift

6
S. Saini et al. Colloids and Surfaces B: Biointerfaces 205 (2021) 111838

Fig. 2. Mean % cumulative drug release profiles of uncoated optimized SLNs and chitosan-coated optimized SLNs, along with pure drug suspension (n = 3).

3.6.8. Ex vivo mucoadhesion studies epithelial mucosa, thus leading eventually to their increased residence
Fig. 3a depicts enhanced mucoadhesive strength of SLNs, on chitosan time in the nasal cavity [83].
coating. The results construe that the detachment force, indicative of
nasal mucoadhesive strength, increased from 6.88 to 8.55 N, after 3.6.9. Ex vivo permeation studies
coating. cSLNs are known to possess a positive surface charge, which Distinct augmentation in the extent of FA permeating through the
tends to facilitate their binding to the anionic mucosal membranes [80, nasal mucosa was observed with uncoated optimized SLNs (i.e., 25.35
81]. Further, mucoadhesion is documented to decrease drug loss due to %) as well as after their being coated with chitosan (i.e., 35.49 %) vis-
mucociliary clearance [82], thus enhancing the availability of drug(s) at à-vis 2.29 % of pure drug suspension, in the first 12 h. This remarkable
the site of the absorptive surface for prolonged periods. The intranasal enhancement in drug permeation through nasal mucosa with SLNs could
application of mucoadhesive nanoparticles causes them to swell, on be ascribed to their lipidic architectural constitution [84]. However, as
coming in contact with the nasal mucosal secretions and adhere to the portrayed in Fig. 3b, drug permeation rate in case of uncoated optimized
formulation (permeation coefficient of 0.00046) was found to be rela­
tively higher than with coated optimized formulation (permeation co­
efficient of 0.00036), as more drug may leach out of the uncoated
system.

3.6.10. Stability studies


The values of assayed drug content, as well as of various physical and
dissolution parameters, studied as CQAs, along with the f2 values
thereof, observed during the 90-days stability studies are enumerated in
Table S6. The freeze-dried cSLNs stored at 25 ± 2 ◦ C and 40 ± 2 ◦ C
demonstrated significant rise in Snm and ZP, along with reduction in EE
values, after reconstitution. Increasing growth in the size of nano­
particulate systems with time and temperature is well-established. The
escalating trend of ZP values at 25 and 40 ◦ C, on the other hand, can
potentially be attributed to the quite spontaneous crystalline re-
orientation of the formulations during three months of storage, result­
ing eventually in shifting the electronic charge on the surface of the
particle(s) from the erstwhile negative to positive ones [57,61]. Storage
of nanoparticles at 40 ◦ C/75 % RH, likewise, resulted in a much higher
increase of the ZP values vis-à-vis with samples stored at 25 ◦ C/60 % RH,
corroborating their far lower stability at high temperatures. Further,
entrapment of FA was observed to decline at all the storage conditions
with time, though minimal influence was seen at the refrigerated con­
ditions, i.e., around 5 ◦ C. Drug expulsion from solid lipid matrix, owing
to crystallization of lipid, is already documented to cause reduction in
EE on storage at 25 ◦ C as well as at 40 ◦ C [84]. Marginal alterations in
drug content and other studied parameters, viz., %EE, globule size and
Q12h, along with miniscule variations in the f2 values, well within the
Fig. 3. (a) Ex vivo mucoadhesive strength of (A) uncoated optimized SLNs; and desirable range of 50 and 100; all corroborate the robustness of the
(B) chitosan-coated optimized SLNs. (b) Mean permeation coefficient of pre­ formulated SLNs under the refrigerated conditions, in consonance with
pared SLNs as compared to pure FA. Each crossbar indicates mean +1 SD (n various literature reports on SLNs [36]. Accordingly, on the basis of the
= 3).

7
S. Saini et al. Colloids and Surfaces B: Biointerfaces 205 (2021) 111838

aforesaid observations, the refrigerated conditions can be vividly 0.001), and AChE activity (p < 0.001), while caused depletion of GSH
construed as most favourable for the storage of SLNs [85]. levels (p < 0.001) and SOD activity (p < 0.001), in hippocampus and
cortex, vis-à-vis sham group (Fig. 5). A 28-day i.n. administration
3.6.11. In vivo studies regimen of uncoated and coated optimized SLNs significantly attenuated
oxidative stress parameters (LPO, nitrite concentration, protein levels
3.6.11.1. Behavioural studies. Animals administered with pure drug, i.n. and AChE activity, and restored GSH levels and SOD activity) vis-à-vis
and orally, with ICV-STZ treatment, showed a notable decline in the positive control group. Further, i.n. administration of coated optimized
cognitive abilities as compared to sham group (p < 0.001 on Day 26 and SLNs also showed distinct improvement in oxidative stress parameters as
27). An insignificant change was observed in mean escape latency time compared to the uncoated optimized SLNs via i.n. treatment.
with pure drug (p > 0.05 on Days 25–27). In a nutshell, there was a nearly 4.9-folds improvement in TSTQ, 3.9-
Meanwhile, treatment of uncoated and coated optimized SLNs i.n., folds reduction in MDA levels, 4.4-folds elevation in GSH levels, 6.6-
along with ICV-STZ treatment, significantly improved cognitive per­ folds reduction in nitrite levels, 4.5-folds reduction in protein levels,
formance (i.e., shortened ELT) vis-à-vis pure FA i.n. (p < 0.001 each on 4.1-folds reduction in AChE activity, 5.2-folds improvement in SOD
Day 27). Moreover, i.n. treatment with coated optimized SLNs vis-à-vis activity and 6.9-folds increase in brain levels of FA in animals with
uncoated ones, significantly reduced mean escape latency time was intranasally administered coated SLNs vis-à-vis pure drug. This signifies
observed on Day 27 (p < 0.001), indicating superior efficacy of the that the developed nanosystem substantially improved the brain access
former in improving cognitive performance. Similarly, animals treated of FA through i.n. route, by potentially circumnavigating BBB.
with uncoated SLNs orally, along with ICV-STZ treatment, showed sig­
nificant improvement in cognitive performance vis-à-vis pure drug oral 3.6.11.3. In vivo brain drug level estimation studies. As observed from
treatment on Day 27 (p < 0.001). Also, a significant difference was Fig. 6, significantly enhanced FA concentration was observed in brain
noticed between uncoated and coated SLNs i.n. vis-à-vis uncoated SLNs homogenates of animals treated intranasally with uncoated optimized
oral treatment (Day 27, p < 0.001). SLNs (5.42-folds) as well as with coated SLNs (6.91-folds) vis-à-vis in
Animals receiving pure drug i.n. and pure drug oral treatment also those treated with the pure drug (p < 0.001 each). Improved trans­
failed to locate the submerged platform as compared to the sham group, cellular transport of FA from the SLNs via olfactory route to brain might
devoting significantly less TSTQ (p < 0.001) (Fig. 4). Significant dif­ cause such augmentation. Surface coating of chitosan on SLNs furnished
ference was perceptible between i.n. administration on of prepared SLNs remarkably higher brain levels, plausibly owing to extended nasal
vis-à-vis oral intake of prepared SLNs in TSTQ as well as % change in retention time, opening of tight junctions of nasal epithelial cells, pos­
TSTQ wrt positive control (p < 0.001). Nearly 4.32- to 4.93-folds itive charge of chitosan and improved stability in the biological milieu of
improvement in TSTQ was seen in the animals treated with uncoated the developed nanosystem [27]. The brain: plasma level ratio, following
and coated optimized SLNs i.n., respectively, vis-à-vis pure drug. The i.n. administration, with cSLNs (i.e., 2.35), was also found to be
uncoated optimized SLNs on oral treatment, however, showed only considerably higher compared to uncoated SLNs (i.e., 1.29) and pure FA
2.66-folds improvement in TSTQ vis-à-vis the corresponding pure drug (i.e., 0.95) administered via i.n. route. The results vividly indicate
(Fig. 4). Overall, following administration of the formulated SLNs, noteworthy contribution of SLNs in direct nose-to-brain transport of FA
significantly improved the navigational abilities of the treated rats to by bypassing the BBB and ratifying the superior delivery of cationic
memorize the platform position thus ratifying enhanced anti-Alzheimer nanoparticles via nasal route.
potential of surface-coated SLNs vis-à-vis pure FA, both administered i.n. 3.6.11.3.1. Histopathological findings. Figure S11 portrays histo­
(p < 0.001). pathological images (at 100X magnification) of different rat organs, viz.,
brain, lung, heart, kidney and liver, treated with normal saline (i.e.,
3.6.11.2. Biochemical estimation studies. Sham-treated rats did not naïve group) (Figure S11 A, C, E, G and I), and those with coated
exhibit any notable change(s) in the biochemical levels in hippocampus optimized SLNs i.n. (Figure S11 B, D, F, H and J). Hippocampus (A and
and cortex brain regions, vis-à-vis naïve group. However, single ICV-STZ B) exhibited no significant change. Bronchioles with lymphoid cells and
administration (positive control group) significantly raised the LPO alveoli (Figure S11 C and D) are observed to be normal. Heart tissue
levels (p < 0.001), nitrite concentration (p < 0.001), protein levels (p < (Figure S11 E and F)) showed normal anatomy. Further, the glomeruli

Fig. 4. Effect on various treatments on time spent in the quadrant during the retention phase of Morris Maze test. Data expressed as mean + 1 SD (n = 3). Data
analysed using one-way ANOVA. ***p < 0.001; N.S. Not Significant.

8
S. Saini et al. Colloids and Surfaces B: Biointerfaces 205 (2021) 111838

Fig. 5. Change in the levels of various biochemical parameters on formulation treatment. Data are expressed as mean +1 SD (n = 3). Data were analysed using one-
way ANOVA. ***p < 0.001; N.S. as Not Significant.

and tubules in kidneys (Figure S11 G and H), were found to be normal to augment its access to the brain, but also to improve its oral
too. Cell cords, portal tracts, central veins and Kupffer cells of liver bioavailability by possible circumnavigation of hepatic first-pass effect
(Figure S11 I and J) portrayed normal anatomy. Overall, these findings and prolongation of retention in the body. Application of rational QbD
indicated no histopathological change(s) in all the vital organs studied paradigms, during drug delivery development, delineated various cause-
between both of the animal groups. Accordingly, the results establish the effect relationships, prioritized the selection of vital few influential
biosafety and tolerability of the developed formulation. variables, embarked upon the demarcation of a robust optimized
formulation and finally validation of DoE application. Evaluation of
4. Conclusions stability studies of SLNs formulations indicated that the refrigerated
storage conditions (i.e., around 5 ◦ C) were quite suitable for maintaining
A number of plant bioactives, including FA, hold enormous promise the robustness of the nanocoutered formulations. in vivo pharmacody­
in alleviating various neurodegenerative disorders but are marked with namic behavioural studies in rats, employing Morris water maze test, as
several physicochemical, biopharmaceutical and biodistributional pit­ well as alteration in the animal body weight as a biomarker, demon­
falls. The current research work was accordingly orchestrated; investi­ strated markedly improved cognition with the coated optimized SLNs,
gating plausible improvement in the anti-Alzheimer’s potential of FA followed by uncoated SLNs vis-à-vis pure drug. Intranasal administration
using novel lipidic nanocarriers through nasal route. Novel mucoadhe­ also showed superior results vis-à-vis oral route. Estimation of apt
sive SLNs of FA, surface-coated with chitosan, were formulated not only biochemical parameters in rat brain homogenates indicated markedly

9
S. Saini et al. Colloids and Surfaces B: Biointerfaces 205 (2021) 111838

Fig. 6. Drug concentrations in brain and plasma of different animal groups. Data expressed are as mean + 1 SD (n = 3). Data analysed using one-way ANOVA. ***p <
0.001; **p < 0.01.

improved regulation of MDA, GSH, nitrite, protein levels, AChE and SOD [5] P. Hrelia, G. Sita, M. Ziche, E. Ristori, A. Marino, M. Cordaro, R. Molteni, V. Spero,
M. Malaguti, F. Morroni, S. Hrelia, Common protective strategies in
activity in the animals receiving uncoated and coated optimized SLNs
neurodegenerative disease: focusing on risk factors to target the cellular redox
intranasally vis-à-vis the pure drug. FA intake via intranasal route was system, Oxid. Med. Cell. Longev. 2020 (2020), 8363245-8363245.
also found to be much more beneficial in upregulating the biochemical [6] B. Heigle, A. Khan, R. Ottwell, M. Vassar, Use of exaggerated language in news
parameters vis-à-vis oral route treatment. Histopathological studies stories to describe drugs for treatment of Alzheimer’s disease, Alzheimers. Dement.
6 (2020) e12062-e12062.
established the safety and tolerability of the coated SLNs. The successful [7] NINDS., Alzheimer’s Disease Information Page, Accessed from https://www.ninds.
outcomes of the present work on QbD-enabled chitosan-coated SLNs nih.gov/Disorders/All-Disorders/Alzheimers-Disease-Information-Page, Accessed
using lipidic nanoconstructs of FA via nose-to-brain delivery with on November, 02, 2020.
[8] National Institute on Aging., Alzheimer’s Disease Fact Sheet, Accessed from https
distinctly superior pharmacodynamic performance in AD can also be ://www.nia.nih.gov/health/alzheimers-disease-fact-sheet, Accessed on November,
rationally extrapolated to other problematic drugs and bioactives 12, 2020.
encountering similar biodistributional and biopharmaceutical issues. [9] M.A. Ansari, H.M. Abdul, G. Joshi, W.O. Opii, D.A. Butterfield, Protective effect of
quercetin in primary neurons against Aβ (1–42): relevance to Alzheimer’s disease,
J. Nutr. Biochem. 20 (2009) 269–275.
[10] J.A. Lemkul, D.R. Bevan, Destabilizing Alzheimer’s Aβ42 protofibrils with morin:
Declaration of Competing Interest mechanistic insights from molecular dynamics simulations, Biochemistry 49
(2010) 3935–3946.
[11] M. Venigalla, E. Gyengesi, G. Münch, Curcumin and apigenin–novel and promising
The authors report no declarations of interest. therapeutics against chronic neuroinflammation in Alzheimer’s disease, Neural
Regen. Res. 10 (2015) 1181–1185.
Acknowledgements [12] T. Beg, S. Jyoti, F. Naz, F. Ali, S.K. Ali, A.M. Reyad, Y.H. Siddique, Protective effect
of kaempferol on the transgenic drosophila model of Alzheimer’s disease, CNS
Neurol. Disord. Drug Targets 17 (2018) 421–429.
The author (B.S.) gratefully appreciates the bestowed generosity of [13] P.X. Xu, S.W. Wang, X.L. Yu, Y.J. Su, T. Wang, W.W. Zhou, H. Zhang, Y.J. Wang, R.
M/s Stat-Ease Inc., USA, in gifting a perpetual and multiple annual T. Liu, Rutin improves spatial memory in Alzheimer’s disease transgenic mice by
reducing Aβ oligomer level and attenuating oxidative stress and
licenses of Design-Expert software to him and his research team as a neuroinflammation, Behav. Brain Res. 264 (2014) 173–180.
mark of recognition to his unmatched contribution in the application of [14] J.J. Yan, J.S. Jung, T.K. Kim, M.A. Hasan, C.W. Hong, J.S. Nam, D.K. Song,
QbD paradigms in developing diverse novel and nanostructured drug Protective effects of ferulic acid in amyloid precursor protein plus presenilin-1
transgenic mouse model of Alzheimer disease, Biol. Pharm. Bull. 36 (2013)
delivery systems. The first author (S.S.) acknowledges the requisite 140–143.
funding provided by the University Grants Commission, New Delhi, [15] S. Lohan, K. Raza, S.K. Mehta, G.K. Bhatti, S. Saini, B. Singh, Anti-Alzheimer’s
India, to him under Meritorious Research Fellowship scheme. potential of berberine using surface decorated multi-walled carbon nanotubes: a
preclinical evidence, Int. J. Pharm. 530 (2017) 263–278.
[16] S. Mishra, K. Palanivelu, The effect of curcumin (turmeric) on Alzheimer’s disease:
Appendix A. Supplementary data an overview, Ann. Indian Acad. Neurol. 11 (2008) 13–19.
[17] C. Sawda, C. Moussa, R.S. Turner, Resveratrol for Alzheimer’s disease, Ann. N. Y.
Acad. Sci. 1403 (2017) 142–149.
Supplementary material related to this article can be found, in the [18] PubChem., Ferulic acid, Accessed from https://pubchem.ncbi.nlm.nih.gov/compo
online version, at doi:https://doi.org/10.1016/j.colsurfb.2021.111838. und/Ferulic-acid, Accessed on October, 1, 2020.
[19] G. Meng, X. Meng, X. Ma, G. Zhang, X. Hu, A. Jin, Y. Zhao, X. Liu, Application of
ferulic acid for Alzheimer’s disease: combination of text mining and experimental
References validation, Front. Neuroinform. 12 (2018) 1–31.
[20] T. Mori, N. Koyama, J. Tan, T. Segawa, M. Maeda, T. Town, Combined treatment
[1] C. Behl, Alzheimer’s disease and oxidative stress: implications for novel therapeutic with the phenolics (-)-epigallocatechin-3-gallate and ferulic acid improves
approaches, Prog. Neurobiol. 57 (1999) 301–323. cognition and reduces Alzheimer-like pathology in mice, J. Biol. Chem. 294 (2019)
[2] W. Zhang, Q. Zhang, Q. Yang, P. Liu, T. Sun, Y. Xu, X. Qian, W. Qiu, C. Ma, 2714–2731.
Contribution of Alzheimer’s disease neuropathologic change to the cognitive [21] S. Kaur, M. Dhiman, A.K. Mantha, Ferulic acid: a natural antioxidant with
dysfunction in human brains with Lewy Body-related pathologyy, Neurobiol. Aging application towards neuroprotection against alzheimer’s disease, in: V. Rani,
91 (2020) 56–65. U. Yadav (Eds.), Functional Food and Human Health, Springer, Singapore, 2018,
[3] S. Karantzoulis, J.E. Galvin, Distinguishing Alzheimer’s disease from other major pp. 576–586.
forms of dementia, Expert Rev. Neurother. 11 (2011) 1579–1591. [22] N. Kumar, V. Pruthi, Potential applications of ferulic acid from natural sources,
[4] A. Alzheimer’s, Alzheimer’s disease facts and figures, Alzheimers. Dement. 11 Biotechnol. Rep. Amst. (Amst) 4 (2014) 86–93.
(2015) (2015) 332–384.

10
S. Saini et al. Colloids and Surfaces B: Biointerfaces 205 (2021) 111838

[23] M. Srinivasan, A.R. Sudheer, V.P. Menon, Ferulic Acid, Therapeutic potential [52] C. Anselmi, M. Centini, M. Ricci, A. Buonocore, P. Granata, T. Tsuno, R.M. Facino,
through its antioxidant property, J. Clin. Biochem. Nutr. 40 (2007) 92–100. Analytical characterization of a ferulic acid/ϒ-cyclodextrin inclusion complex,
[24] Z. Zhao, Y. Egashira, H. Sanada, Ferulic acid is quickly absorbed from rat stomach J. Pharm. Biomed. Anal. 40 (2006) 875–881.
as the free form and then conjugated mainly in liver, J. Nutr. 134 (2004) [53] Y.T. Sohn, J.H. Oh, Characterization of physicochemical properties of ferulic acid,
3083–3088. Arch. Pharm. Res. 26 (2003) 1002–1008.
[25] V. Jenning, S.H. Gohla, Encapsulation of retinoids in solid lipid nanoparticles [54] B. Singh, S. Singh, A comprehensive computer program for the study of drug
(SLN), J. Microencapsul. 18 (2001) 149–158. release kinetics from compressed matrices, Indian J. Pharm. Sci. 60 (1998)
[26] A. Zielińska, N.R. Ferreira, A. Feliczak-Guzik, I. Nowak, E.B. Souto, Loading, 358–362.
release profile and accelerated stability assessment of monoterpenes-loaded solid [55] F.C. Carvalho, M.S. Barbi, V.H.V. Sarmento, L.A. Chiavacci, F.M. Netto, M.P.
lipid nanoparticles (SLN), Pharm. Dev. Technol. 25 (2020) 832–844. D. Gremião, Surfactant systems for nasal zidovudine delivery: structural,
[27] P. Fonte, F. Andrade, F. Araujo, C. Andrade, J. das Neves, B. Sarmento, Chitosan- rheological and mucoadhesive properties, J. Pharm. Pharmacol. 62 (2010)
coated solid lipid nanoparticles for insulin delivery. Methods in Enzymology, 430–439.
Elsevier, 2012, pp. 295–314. [56] A.R. Madgulkar, M.R. Bhalekar, K.D. Asgaonkar, A.A. Dikpati, Synthesis and
[28] A.C. Vieira, L.L. Chaves, S. Pinheiro, S. Pinto, M. Pinheiro, S.C. Lima, D. Ferreira, characterization of a novel mucoadhesive derivative of xyloglucan, Carbohydr.
B. Sarmento, S. Reis, Mucoadhesive chitosan-coated solid lipid nanoparticles for Polym. 135 (2016) 356–362.
better management of tuberculosis, Int. J. Pharm. 536 (2018) 478–485. [57] S. Saini, T. Sharma, A. Patel, R. Kaur, S.K. Tripathi, O.P. Katare, B. Singh, QbD-
[29] Y. Luo, Z. Teng, Y. Li, Q. Wang, Solid lipid nanoparticles for oral drug delivery: steered development and validation of an RP-HPLC method for quantification of
chitosan coating improves stability, controlled delivery, mucoadhesion and cellular ferulic acid: rational application of chemometric tools, J. Chromatogr. B, Biomed.
uptake, Carbohydr. Polym. 122 (2015) 221–229. Appl. 1155 (2020), 122300.
[30] F. Erdő, L.A. Bors, D. Farkas, Á. Bajza, S. Gizurarson, Evaluation of intranasal [58] C. Salamanca, A. Barrera-Ocampo, J. Lasso, N. Camacho, C. Yarce, Franz diffusion
delivery route of drug administration for brain targeting, Brain Res. Bull. 143 cell approach for pre-formulation characterisation of ketoprofen semi-solid dosage
(2018) 155–170. forms, Pharmaceutics 10 (2018) 148.
[31] X. Dong, Current strategies for brain drug delivery, Theranostics 8 (2018) [59] H. Zhang, J.R. Robinson, In vitro methods for measuring permeability of the oral
1481–1493. mucosa, in: M.J. Rathbone (Ed.), Oral Mucosal Drug Delivery, Marcel Dekker Inc.,
[32] L. Casettari, L. Illum, Chitosan in nasal delivery systems for therapeutic drugs, N.Y, 1996, pp. 85–100.
J. Control. Release 190 (2014) 189–200. [60] M.G. Fronza, R. Baldinotti, M.C. Martins, B. Goldani, B.T. Dalberto, F.S. Kremer,
[33] V. Trotta, B. Pavan, L. Ferraro, S. Beggiato, D. Traini, L.G. Des Reis, S. Scalia, K. Begnini, L. da Silva Pinto, E.J. Lenardão, F.K. Seixas, T. Collares, Rational
A. Dalpiaz, Brain targeting of resveratrol by nasal administration of chitosan- design, cognition and neuropathology evaluation of QTC-4-MeOBnE in a
coated lipid microparticles, Eur. J. Pharm. Biopharm. 127 (2018) 250–259. streptozotocin-induced mouse model of sporadic Alzheimer’s disease, Sci. Rep. 9
[34] S. Pedroso-Santana, N. Fleitas-Salazar, Ionotropic gelation method in the synthesis (2019) 7276.
of nanoparticles/microparticles for biomedical purposes, Polym. Int. 69 (2020) [61] K.G. Ravelli, B. dos Anjos Rosário, R. Camarini, M.S. Hernandes, L.R. Britto,
443–447. Intracerebroventricular streptozotocin as a model of Alzheimer’s Disease:
[35] M.L. Tsai, S.W. Bai, R.H. Chen, Cavitation effects versus stretch effects resulted in neurochemical and behavioral characterization in mice, Neurotox. Res. 31 (2017)
different size and polydispersity of ionotropic gelation chitosan–sodium 327–333.
tripolyphosphate nanoparticle, Carbohydr. Polym. 71 (2008) 448–457. [62] S. Shree, R. Bhardwaj, Kashis, R. Deshmukh, Non-transgenic animal models of
[36] M. Bhalekar, P. Upadhaya, A. Madgulkar, Formulation and characterization of alzheimer’s disease, in: P.K. Bansal, R. Deshmukh (Eds.), Animal Models of
solid lipid nanoparticles for an anti-retroviral drug darunavir, Appl. Nanosci. 7 Neurological Disorders: Principle and Working Procedure for Animal Models of
(2017) 47–57. Neurological Disorders, Springer, Singapore, Singapore, 2017, pp. 3–22.
[37] S. Beg, S. Jain, V. Kushwah, G.K. Bhatti, P.S. Sandhu, O.P. Katare, B. Singh, Novel [63] G. Kamalinia, F. Khodagholi, F. Atyabi, M. Amini, F. Shaerzadeh, M. Sharifzadeh,
surface-engineered solid lipid nanoparticles of rosuvastatin calcium for low-density R. Dinarvand, Enhanced brain delivery of deferasirox-lactoferrin conjugates for
lipoprotein-receptor targeting: a Quality by Design-driven perspective, iron chelation therapy in neurodegenerative disorders: In vitro and in vivo studies,
Nanomedicine 12 (2017) 333–356. Mol. Pharm. 10 (2013) 4418–4431.
[38] S. Dhawan, R. Kapil, B. Singh, Formulation development and systematic [64] E.D. Wills, Mechanisms of lipid peroxide formation in animal tissues, Biochem. J.
optimization of solid lipid nanoparticles of quercetin for improved brain delivery, 99 (1966) 667–676.
J. Pharm. Pharmacol. 63 (2011) 342–351. [65] G.L. Ellman, Tissue sulfhydryl groups, Arch. Biochem. Biophys. 82 (1959) 70–77.
[39] P. Nirbhavane, G. Sharma, B. Singh, G.K. Khuller, V.G. Goni, A.B. Patil, O. [66] L.C. Green, D.A. Wagner, J. Glogowski, P.L. Skipper, J.S. Wishnok, S.
P. Katare, Preclinical explorative assessment of celecoxib-based biocompatible R. Tannenbaum, Analysis of nitrate, nitrite, and [15N]nitrate in biological fluids,
lipidic nanocarriers for the management of CFA-induced rheumatoid arthritis in Anal. Biochem. 126 (1982) 131–138.
Wistar rats, AAPS PharmSciTech 19 (2018) 3187–3198. [67] A.G. Gornall, C.J. Bardawill, M.M. David, Determination of serum proteins by
[40] S. Trombino, R. Cassano, T. Ferrarelli, E. Barone, N. Picci, C. Mancuso, Trans- means of the biuret reaction, J. Biol. Chem. 177 (1949) 751–766.
ferulic acid-based solid lipid nanoparticles and their antioxidant effect in rat brain [68] Y. Kono, Generation of superoxide radical during autoxidation of hydroxylamine
microsomes, Colloids Surf. B Biointerfaces 109 (2013) 273–279. and an assay for superoxide dismutase, Arch. Biochem. Biophys. 186 (1978)
[41] B. Singh, R. Bhatowa, C.B. Tripathi, R. Kapil, Developing micro-/nanoparticulate 189–195.
drug delivery systems using “design of experiments”, Int. J. Pharm. Investig. 1 [69] G.L. Ellman, K.D. Courtney, V. Andres Jr, R.M. Featherstone, A new and rapid
(2011) 75–87. colorimetric determination of acetylcholinesterase activity, Biochem. Pharmacol. 7
[42] B. Singh, A. Rani, N. Ahuja, R. Kapil, Formulation optimization of (1961) 88–95.
hydrodynamically balanced oral controlled release bioadhesive tablets of tramadol [70] U. Bickel, How to measure drug transport across the blood-brain barrier, NeuroRx
hydrochloride, Sci. Pharm. 78 (2010) 303–324. 2 (2005) 15–26.
[43] T.A. Ahmed, B.M. Aljaeid, Preparation, characterization, and potential application [71] G.S. Travlos, R.W. Morris, M.R. Elwell, A. Duke, S. Rosenblum, M.B. Thompson,
of chitosan, chitosan derivatives, and chitosan metal nanoparticles in Frequency and relationships of clinical chemistry and liver and kidney
pharmaceutical drug delivery, Drug Des. Devel. Ther. 10 (2016) 483–507. histopathology findings in 13-week toxicity studies in rats, Toxicology 107 (1996)
[44] J.Y. Wang, Y. Wang, X. Meng, Chitosan nanolayered cisplatin-loaded lipid 17–29.
nanoparticles for enhanced anticancer efficacy in cervical cancer, Nanoscale Res. [72] B. Singh, S. Saini, S. Lohan, S. Beg, Systematic development of nanocarriers
Lett. 11 (2016) 1–8. employing quality by design paradigms. Nanotechnology-Based Approaches for
[45] O. Gartziandia, E. Herran, J.L. Pedraz, E. Carro, M. Igartua, R.M. Hernandez, Targeting and Delivery of Drugs and Genes, Academic Press, Massachusetts, US,
Chitosan coated nanostructured lipid carriers for brain delivery of proteins by 2017, pp. 110–148.
intranasal administration, Colloids Surf. B Biointerfaces 134 (2015) 304–313. [73] B. Singh, T. Sharma, S. Saini, R. Kaur, A. Jain, K. Raza, S. Beg, Systematic
[46] G. Rassu, E. Soddu, A.M. Posadino, G. Pintus, B. Sarmento, P. Giunchedi, E. Gavini, development of drug nanocargos using formulation by design (FbD): an updated
Nose-to-brain delivery of BACE1 siRNA loaded in solid lipid nanoparticles for overview, Crit. Rev. Ther. Drug Carrier Syst. 37 (2020) 229–269.
Alzheimer’s therapy, Colloids Surf. B Biointerfaces 152 (2017) 296–301. [74] M.L. Bondi, G. Montana, E.F. Craparo, P. Picone, G. Capuano, M.D. Carlo,
[47] G. Guizhen, J. Qingcai, D. Yilei, Study on quantitative assay of chitosan in G. Giammona, Ferulic acid-loaded lipid nanostructures as drug delivery systems for
compound sample with a spectrophotometric method, Food Ferment. Ind. 29 Alzheimer’s disease: Preparation, characterization and cytotoxicity studies, Curr.
(2003) 49–52. Nanosci. 5 (2009) 26–32.
[48] K. Oehlke, D. Behsnilian, E. Mayer-Miebach, P.G. Weidler, R. Greiner, Edible solid [75] Malvern., Zetasizer Malvern: nano series, Accessed from https://www.chem.uci.
lipid nanoparticles (SLN) as carrier system for antioxidants of different edu/~dmitryf/manuals/Malvern%20Zetasizer%20ZS%20DLS%20user%
lipophilicity, PLoS One 12 (2017), e0171662. 20manual.pdf, Accessed on Nov, 10, 2020.
[49] R. Kumar, P.F. Siril, P. Soni, Tuning the particle size and morphology of high [76] D.L. Pavia, G.M. Lampman, G.S. Kriz, J.R. Vyvyan, Infrared spectroscopy,
energetic material nanocrystals, Def. Technol. 11 (2015) 382–389. Introduction to spectroscopy (2001) 15–104.
[50] PerkinElmer Frontier., IR ready for an challenge, Accessed from https://www.per [77] T. Delmas, A.-C. Couffin, P.A. Bayle, F. De Crecy, E. Neumann, F. Vinet, M. Bardet,
kinelmer.com/lab-solutions/resources/docs/BRO_FrontierFTIR.pdf, Accessed on J. Bibette, I. Texier, Preparation and characterization of highly stable lipid
April, 1, 2021. nanoparticles with amorphous core of tuneable viscosity, J. Colloid Interface Sci.
[51] N.R. Pani, L.K. Nath, S. Acharya, B. Bhuniya, Application of DSC, IST, and FTIR 360 (2011) 471–481.
study in the compatibility testing of nateglinide with different pharmaceutical [78] C. Freitas, R. Müller, Correlation between long-term stability of solid lipid
excipients, J. Therm. Anal. 108 (2012) 219–226. nanoparticles (SLN™) and crystallinity of the lipid phase, Eur. J. Pharm. Biopharm.
47 (1999) 125–132.

11
S. Saini et al. Colloids and Surfaces B: Biointerfaces 205 (2021) 111838

[79] F.Z. Wang, M.W. Zhang, D.S. Zhang, Y. Huang, L. Chen, S.M. Jiang, K. Shi, R. Li, [83] S. Liu, S. Yang, P.C. Ho, Intranasal administration of carbamazepine-loaded
Preparation, optimization, and characterization of chitosan-coated solid lipid carboxymethyl chitosan nanoparticles for drug delivery to the brain, Asian J.
nanoparticles for ocular drug delivery, J. Biomed. Res. 32 (2018) 411–423. Pharm. Sci. 13 (2018) 72–81.
[80] K. Divya, M.S. Jisha, Chitosan nanoparticles preparation and applications, Environ. [84] G. Nava, E. Piñón, L. Mendoza, N. Mendoza, D. Quintanar, A. Ganem, Formulation
Chem. Lett. 16 (2018) 101–112. and in vitro, ex vivo and in vivo evaluation of elastic liposomes for transdermal
[81] A.B. Nair, N. Sreeharsha, B.E. Al-Dhubiab, J.G. Hiremath, P. Shinu, M. Attimarad, delivery of ketorolac tromethamine, J. Pharm. Drug Deliv. Res. 3 (2011) 954–970.
K.N. Venugopala, M. Mutahar, HPMC- and PLGA-based nanoparticles for the [85] P.A. Makoni, K. Wa Kasongo, R.B. Walker, Short term stability testing of efavirenz-
mucoadhesive delivery of sitagliptin: optimization and in vivo evaluation in rats, loaded solid lipid nanoparticle (SLN) and nanostructured lipid carrier (NLC)
Materials Basel (Basel) 12 (2019) 4239. dispersions, Pharmaceutics 11 (2019) 1–21.
[82] J. Leal, H.D.C. Smyth, D. Ghosh, Physicochemical properties of mucus and their
impact on transmucosal drug delivery, Int. J. Pharm. 532 (2017) 555–572.

12

You might also like