Molecules 28 01833
Molecules 28 01833
Article
Development of Computational In Silico Model for Nano Lipid
Carrier Formulation of Curcumin
Omar Waleed Abduljaleel Albasri, Palanirajan Vijayaraj Kumar * and Mogana Sundari Rajagopal
Faculty of Pharmaceutical Sciences, Department of Pharmaceutical Technology, UCSI University, Jalan Menara
Gading, Taman Connaught, Cheras, Kuala Lumpur 56000, Malaysia
* Correspondence: [email protected]
Abstract: The oral delivery system is very important and plays a significant role in increasing the
solubility of drugs, which eventually will increase their absorption by the digestive system and
enhance the drug bioactivity. This study was conducted to synthesize a novel curcumin nano lipid
carrier (NLC) and use it as a drug carrier with the help of computational molecular docking to
investigate its solubility in different solid and liquid lipids to choose the optimum lipids candidate for
the NLCs formulation and avoid the ordinary methods that consume more time, materials, cost, and
efforts during laboratory experiments. The antiviral activity of the formed curcumin–NLC against
SARS-CoV-2 (COVID-19) was assessed through a molecular docking study of curcumin’s affinity
towards the host cell receptors. The novel curcumin drug carrier was synthesized as NLC using a
hot and high-pressure homogenization method. Twenty different compositions of the drug carrier
(curcumin nano lipid) were synthesized and characterized using different physicochemical techniques
such as UV–Vis, FTIR, DSC, XRD, particle size, the zeta potential, and AFM. The in vitro and ex
vivo studies were also conducted to test the solubility and the permeability of the 20 curcumin–NLC
formulations. The NLC as a drug carrier shows an enormous enhancement in the solubility and
permeability of the drug.
are combined with solid lipids to make NLCs. Normally, tween 80 and poloxamer 188 are
typically used as surfactants [11].
Curcumin is mostly derived from the natural plant. The richest and most cost-effective
source of crude curcumin is Curcuma longa [12]. It has also been used to treat a range of
problems in various patients, including diabetes, liver disease, cancer, and rheumatoid
disease [13]. Curcumin is only sparsely soluble in water [14]. Chemically, it is an unstable
molecule with a limited biological half-life because it is poorly absorbed and rapidly
metabolized [15]. All these factors contribute to curcumin’s lower bioavailability when
taken as a whole [16]. It is categorized as GRAS (generally recognized as safe) by the
US Food and Drug Administration [17]. The most attractive and vital reason for the
therapeutic use of curcumin is its superior safety profile. It has been demonstrated that
curcumin has a very low toxicity [18]. The bioactivity of orally taken curcumin has been the
most fundamental basis for this apprehension. Thus, it can be used as an anti-inflammatory,
anti-microbial, anti-oxidant, anti-viral, and anti-cancer agent [19–21].
In the last two decades, much research and many experiments were carried out to
investigate the optimal characteristics of the drug [22]. The previous trials were not effi-
cient and expensive [23], thus virtual screening was applied as a new approach based on
structural information [24]. The method of virtual screening can be classified as structure-
based and ligand-based drug designing methods [25,26]. The first one describes molecular
docking while the second method deals with the relationship between the quantitative
structure’s activity and pharmacophore modeling [27,28]. The structural information in-
creases the range of molecular targets of proteins and protein–ligand complexes via certain
steps, starting with chemical synthesis techniques, purification, X-ray crystallography, and
nuclear magnetic resonance spectroscopy (NMR) [29].
Generally, the molecular docking method determines the interaction between the
ligand and target molecule [30,31]. It can also predict the ligand binding affinity in the
formation of a stable complex with protein in minimum free binding energy [32,33]. The
binding can occur via non-covalent interactions such as hydrogen bonds, ionic bonds,
hydrophobic, and van der Waals forces [34].
The current research proposed the development of NLC molecules to improve the
oral solubility and permeability of curcumin. The research also focused on developing
a computational molecular docking to study the solubility of curcumin in different solid
and liquid lipids to choose the optimum candidates for the NLCs formulation rather than
the frequent experimental method, which consumes much time, efforts, and high costs.
The molecular docking model was applied to study and enhance the antiviral activity of
curcumin against COVID-19 by studying the binding affinity to the host cell receptors of
the angiotensin-converting enzyme (ACE2) [35].
types of bond interaction between omega-3 and curcumin were examined. The results
represent one hydrogen bond as a conventional hydrogen bond (distance 2.398), one
hydrogen bond as a carbon–hydrogen bond (distance 3.537), and three hydrophobic pi–
alkyl bonds (distance range 5.066–5.472). For linoleic acid (obtained from grape seeds
oil) CID 5280450, the types of bond interaction between linoleic acid and curcumin were
examined. The results represent one hydrophobic pi–sigma (distance 3.573) bond and three
hydrophobic pi–alkyl bonds (distance range 3.792–4.982). For ethyl palmitate (obtained
from olive oil) CID 12366, the types of bond interaction between ethyl palmitate and
curcumin were examined. The results represent one hydrophobic pi–sigma (distance 3.536)
bond and four hydrophobic pi–alkyl bonds (distance range 3.658–5.498). For ricinoleic acid
(obtained from castor oil) CID 14030006, the types of bond interaction between ricinoleic
acid and curcumin were examined. The results represent three hydrophobic pi–alkyl bonds
(distance range 3.879–5.082) as shown in Figure 1.
Figure 1. 3D molecular interaction between curcumin (CUR) with different liquid lipids (highlighted
in yellow) for solubility prediction. The formed bonds are described as black short lines.
Oleic acid shows the types of bond interaction between oleic acid and curcumin. The
results represent four hydrogen bonds (distance range 3.464–3.514) and twelve hydrophobic
pi–alkyl bonds (distance range 3.911–5.433). Figure 2 represents the 3D interaction between
the curcumin molecule and the oleic acid molecule with bond types and distances.
Figure 2. 3D interaction between curcumin molecule (gray) and oleic acid oil (yellow). The formed
bonds are described as black short lines.
Considering solid lipids (SL), the types of bond interaction between Cetostearyl alcohol
and curcumin were examined. The results represent four hydrophobic pi–alkyl bonds
(distance range 4.015–4.306), while the types of bond interaction between stearic acid and
curcumin were examined. The results represent four hydrophobic pi–alkyl bonds (distance
range 3.827–5.029). The types of bond interaction between palmitic acid and curcumin
were examined, and the results represent two hydrogen bonds (distance 3.545 and 3.643)
and four hydrophobic pi–alkyl bonds (distance range 3.654–5.101). The types of bond
Molecules 2023, 28, 1833 4 of 23
interaction between tripalmitin and curcumin were examined and the results represent
two hydrogen bonds as conventional hydrogen bonds (distance 2.664 and 2.701) and two
hydrophobic pi–alkyl bonds (distance 3.917 and 4.729), as presented in Figure 3.
Figure 3. 3D molecular interaction between curcumin (CUR) with different solid lipids (highlighted
in yellow) for solubility prediction. The formed bonds are described as black short lines.
As observed in Table 1, the types of bond interaction between GMS and curcumin
represent six hydrogen bonds as conventional hydrogen bonds, one hydrophobic pi–sigma
(C–H) bond, and one hydrophobic pi–alkyl bond (pi-orbital). Figure 4 represents the 3D
and 2D interaction between the curcumin molecule and GMS molecule, respectively; the
bonds type and distances were also described.
The scale of root means square deviation (RMSD) clustering for the solubility construal
docking for GMS was set to nine and was used to determine how well scoring combinations
pose and calculate the ligand (CUR) in the lipid site (GMS). Discovery Studio software
was used to interpret the obtained docking results. Molecular docking insights revealed
that CUR bound to GMS via six hydrogen bonds, one pi–sigma C–H hydrophobic bond,
and one pi–alkyl hydrophobic bond. The values for the binding energy of each lipid with
curcumin were obtained and used as an indicative tool to expect the stability of the formed
formulations and was approximately −8.6 Kcal/mol for GMS, which was significantly
lower than the other compared ten lipids (range from −6.5 to −1.8).
Molecules 2023, 28, 1833 5 of 23
Figure 4. 3D and 2D molecular interaction between curcumin molecule (gray) and GMS (yellow).
The lowest binding energy of GMS has the highest binding affinity to curcumin and
indicates that the ligand binds strongly to the receptor, which proves that GMS has the
highest solubility tendency for curcumin. BIOVIA Discovery Studio software confirmed
the interaction between CUR and GMS. It was reported that conventional hydrogen bond-
ing shows the strongest interaction between molecules and the results showed that six
conventional H-bonding were generated between CUR as the H-donner and GMS as the
H-accepter in the type of OH–H bond, which shows fundamental solubilizing services in a
biomolecular structure [36].
Hydrogen bonds perform as facilitators to CUR–GMS binding and offer an inordinate
chance for the solubilization of CUR in GMS since it can endorse the ligand binding
affinity among the donor and acceptor. It was conveyed that hydroxyl groups of GMS
play a crucial role in binding through hydrogen bonding generation and solubilizing CUR.
Therefore, this demonstrated that CUR could bind and have an affinity to GMS through
strong conventional hydrogen bonding [37]. In addition, the shortest distance of these
conventional hydrogen bonds compared to another type of bond present (less than 3000 ◦ A)
proves that the presence of such bonds gives a higher binding affinity and solubility
between curcumin and GMS molecules [38]. These findings were not reported with the
other compared 10 lipids. Moreover, the presence of a pi–alkyl bond in the interaction
between curcumin and GMS provides the stability for the system, and the presence of the
pi–sigma bond provides an extra stability to the interaction between the molecules [39]. On
the other hand, oleic acid was found to be the best liquid lipid candidate to be mixed with
GMS for the NLCs formulation since the high compatibility between these two lipids was
found experimentally and due to the obtained docking results, which showed that oleic
acid was superior compared to other liquid lipids in demonstrating a lower binding energy
of −6.5 kcal/mol. Moreover, the presence of four C–H bonds in the oleic acid–curcumin
interaction allows solubility [40].
A comparison between the predicted solubility of curcumin in different solid and
liquid lipids obtained from the molecular docking study (through calculating the binding
energy) and the actual solubility results obtained from laboratory experiments (by calculat-
ing the amount of curcumin dissolved in these lipids) was conducted to see how far this
computational docking system can be recommended for choosing the optimum solid and
liquid lipid. The results were promising and showed a similar order of solubility (from
highest to lowest) for both solid and liquid lipids, indicating the accuracy of the docking
study applied, as shown in Table 2.
Molecules 2023, 28, 1833 6 of 23
Table 3. Formulations results of EE%, particle size, PDI, and zeta potential.
Table 3. Cont.
Figure 5. FTIR spectra of (A) GMS and (B) oleic acid, plot of %Transmittance vs. 1/cm.
Figure 6. FTIR for pure curcumin (A), the optimized formulation of curcumin–NLC F1 (B), which
shows no chemical interaction with curcumin major peaks, plot %Transmittance vs. 1/cm.
Molecules 2023, 28, 1833 8 of 23
The lack of additional peaks/broadening of the peaks suggests that the process param-
eters did not cause the curcumin to be stressed, resulting in degradation. Curcumin also
had no chemical interactions and was confirmed to be compatible with the formulation
excipients [47,48].
Figure 7. DSC studies of pure curcumin (A), the prepared curcumin NLCs-F1 (B) with the absence
melting peak of curcumin at 180 ◦ C confirming amorphous state plot of mW vs. temp.
Figure 8. Powder X-ray diffraction studies of pure curcumin (A), and curcumin NLCs-F1 (B) presenting
significant lowering intensity due to high amorphization, the plot of Counts vs. Theta-2 Theta (deg).
Molecules 2023, 28, 1833 9 of 23
Figure 9. Counter and response surface plots of entrapment efficiency (stirring time and lipid
concentration), n = 6.
Figure 10. Comparison of cumulative % drug release NLC formulas (F1, F2, F4, F12, and F19) and
pure curcumin suspension versus time (h), mean ± SD (n = 6).
Figure 11. Atomic force microscopy image of optimum curcumin–NLC F1 determining the morphol-
ogy of the prepared nano lipids (size area 3 × 3 µm).
Figure 12. Intestinal permeation study representing permeation of curcumin suspension and
curcumin-NLCs versus time (h), mean ±SD (n = 6).
2.8. Molecular Docking Studies of the Anti-Viral Activity of Curcumin NLC against SARS-CoV-2
(COVID-19)
The docking scores of the binding affinity between the different host cell receptors
and curcumin in the curcumin–NLC complex are tabulated in Table 4. A comparison was
applied for the obtained docking results of the curcumin interaction with the host cell
Molecules 2023, 28, 1833 12 of 23
receptors of ACE 2 and the curcumin interaction with NLC in the curcumin–NLC–host
cell receptor complex to study the influence of the formed curcumin–NLC on the binding
affinity for the target host cell receptors. First, the PyRx software was applied for the
docking of curcumin with the NLC components and it was then explored through the
Discovery Studio visualizer. Again, the obtained micelles were subjected to docking with
the three host cells through the same process as described earlier.
Table 4. The docking scores of curcumin with different targets (host cell receptors) in the curcumin–
NLC complex.
The anti-viral potential of curcumin against COVID-19 using different targets was
investigated. The results showed a significant reduction in the binding energy, indicating
an enhancement in the binding affinity of the ligand toward the targets. The curcumin
interaction with NLC in a complex has shown an interaction potential and exhibited
−6.8 kcal/mol binding energy and formed one conventional H-bond with C: ASP994, one
carbon–hydrogen bond C: GLY999, two pi–sigma C: THR998 and A: THR998, and five pi–
alkyl bonds B: VAL991, C: PHE970, A: TYR756, B: ARG995, and C: ARG995. Tables 5 and 6
epitomize the results of the data obtained from the active residues, bond length (A0), bond
types, and bond categories involved in the molecular interactions of curcumin with NLC
and curcumin with different receptors (7KMB,7KNB, and 7KNH) in the curcumin–NLC–
receptor complex.
Table 5. The active residues, bond length (A0), bond types, and bond categories involved in the
molecular interactions of curcumin with NLC in a complex.
Table 6. The active residues, bond length (A0), bond types, and bond categories involved in the
molecular interactions of curcumin with ACE2 receptors (7KMB, 7KNB, and 7KNH) in a complex.
Table 6. Cont.
For the 7KMB receptor, the curcumin interaction with the receptor in a complex
represents a significant enhancement in the binding affinity through an energy reduction
to −9.1 kcal/mol; the interaction also formed an additional four H-bonds compared to
the two H-bonds formed for curcumin with NLC, indicating an increase in the curcumin
affinity towards the receptor. The formed conventional H-bond showed a great increase
through six formed bonds F: TYR202, F: SER511, F: SER511, F: ARG514, F: LYS562, and F:
GLY395. Moreover, pi–anion, pi–pi stacked, and pi–alkyl were formed with F: ASP206, F:
TYR202, and F: TYR202, respectively (Figure 13).
For the 7KNB receptor, interestingly, the curcumin interaction with the receptor in
a complex represents a strong enhancement of the binding affinity through an energy
reduction to −8.6 kcal/mol and formed a new extra five H-bonds, indicating a high increase
in the curcumin affinity towards the receptor. The interaction showed six conventional
H-bonds C: THR998, A: ARG995, A: ARG995, A: ARG995, B: ARG995, and B: VAL991, one
carbon–hydrogen bond C: ASP994, one amide pi-stacked bond A: ASP994:C, O;ARG995,
and 3 pi–alkyl bonds C: VAL991, A: ARG995 and B: ARG995 (Figure 14).
Molecules 2023, 28, 1833 14 of 23
Figure 13. 2D interaction of curcumin–NLC with ACE 2 binding receptor (7KMB) in a complex.
Figure 14. 2D interaction of curcumin–NLC with ACE 2 binding receptor (7KNB) in a complex.
For the 7KNH receptor, a study of the curcumin interaction with the receptor in a
complex represents an enhancement in the binding affinity through an energy reduction
to −8.4 kcal/mol and formed new additional six H-bonds and carbon–hydrogen bonds,
indicating a high increase in the curcumin affinity towards the receptor. The interaction
showed five conventional H-bonds C: GLN755, B: ASN969, B: ARG995, B: ARG995, and B:
GLY971, three carbon–hydrogen bonds C: ASP994, B: ALA972, and B: ILE973, one pi–anion
bond C: GLU990, and one pi–alkyl bond C: LEU752 (Figure 15).
The results in Table 6 show a significant tendency of the binding affinity of curcumin
to the three binding receptors of ACE2 when formulated as an NLC due to the low binding
energy and the increased H-bonding [72]. Consequently, these increase the antiviral
property of curcumin against SARS-CoV-2 and disrupt SARS-CoV-2 binding to ACE2
binding receptors and prevent their entry into the cell. It is anticipated that enhancing the
curcumin solubility through the NLCs formulation will provide a suitable concentration of
the medicine for its absorption and bioavailability, which enhances the curcumin’s anti-viral
activity in the treatment of COVID-19 [73].
Figure 15. 2D interaction of curcumin–NLC with ACE 2 binding receptor (7KNH) in a complex.
3.2. Molecular Dynamic Study for Curcumin Solubility and Interaction with Solid and
Liquid Lipids
The interaction of curcumin (CUR) lipids for the solubility prediction [SDF, PDB]
was studied. Low molecular weight curcumin was derived from the PubChem website
(https://pubchem.ncbi.nlm.nih.gov/, accessed on 3 January 2023) and downloaded as
SDF and converted to pdb format using Discovery Studio Software (version- 19.1.0.18287).
Eleven lipid structures including solid lipids (GMS, tripalmitin, stearic acid, palmitic acid,
and sitosterol alcohol) and liquid lipids (oleic acid, apocarotenal, ethyl palmitate, omega-3,
ricinoleic acid, and linoleic acid) were also downloaded as SDF from the PubChem website
and prepared for docking through the same process as curcumin. All the structures of the
selected compounds that aided as molecules for modeling studies were adjusted using the
Auto-Dock software (PyRx software version 0.8, python.exe) before docking. Using PyRx
software, the lipid molecules were subjected to auto-dock to build a macromolecule.
The ligand curcumin was dragged to the software in SDF format and subjected to
minimizing energy (E = 234.8) and was saved as the pdbq format. After selecting each lipid
molecule involved (which was used in experimental laboratory studies in this project) and
ligand, run vina was applied to study the interaction between lipid and ligand curcumin
through PyRx software. The docking outcomes were scrutinized to recognize and assess
the binding capability between these compounds. Discovery Studio software was used to
analyze the type of bond interaction between lipid and curcumin and we chose the best
fit interaction for the optimum solubility. Auto-dock vina in PyRx software is the most
preferable software for molecular docking. The molecular docking approach can be used to
model the interaction between small molecules and proteins at the atomic level, allowing
for the characterization of the behavior of small molecules in the binding site of target
proteins, as well as elucidating the fundamental biochemical processes [74,75]. Discovery
Studio software is an agglomeration to transcribe small molecules and macromolecule
systems. It is developed by Dassault Systems BIOVIA (Accelrys) [76]. Discovery Studio is a
single unified, graphical interface for advanced drug design and protein modeling research.
This software provides a plethora of viewers for display plots and graphical representations
of data [77]. In the present study, there was an investment in this software to predict the
interaction and solubility between hydrophobic drugs such as curcumin and lipids.
Table 7. Cont.
detector wavelength was set at 425 nm. The injection volume was 10 µL. Methanol was
used as the diluent.The lipid-specific NLCs blanks were all the same [79].
ζ = εµ/η
where ζ is the zeta potential, µ is the electrophoretic mobility, ε is the electric permittivity
of the liquid, and η is the viscosity of the liquid.
3.9. Molecular Docking Studies of the Anti-Viral Activity of Curcumin against SARS-CoV-2
(COVID-19)
The molecular docking was conducted on the Lenovo ThinkPad T440p using the
PyRx-Virtual Screening Tool. The structure of curcumin (sdf file format) was downloaded
from the official website of the National Center for Biotechnology Information PubChem
(https://pubchem.ncbi.nlm.nih.gov/, accessed on 3 January 2023). The energy minimiza-
tion (optimization) was performed by a Universal Force Field (UFF). The 3D structure of
curcumin–NLC constituents GMS, oleic acid, poloxamer 188, and tween 80 were obtained
from drawing these chemical structures through ChemSchetch software and transferred
from 2D to 3D and saved (as.mol file format). Then, they were converted to PDB format
using the Discovery Studio visualizer 2019.
These structures were prepared for docking through PyRx software. The three host
cell receptor structures of ACE2 (PDB ID: 7KMB, 7KNB, and 7KNH) were obtained from
the RCSB PDB site (https://www.rcsb.org/, accessed on 3 January 2023). The receptor
structures, with the aid of the Discovery Studio Visualizer 2021, were optimized, purified,
and prepared for molecular docking. Autodock vina 1.1.2 in PyRx 0.8 was used to perform
the molecular docking studies. A large number of glycosylated S proteins cover the surface
of SARS-CoV-2 and bind to the host cell receptor angiotensin-converting enzyme 2 (ACE2),
Molecules 2023, 28, 1833 19 of 23
mediating a viral cell entry. Once the virus enters the cell, the viral RNA is released.
Polyproteins are translated from the RNA genome, and the replication and transcription
of the viral RNA genome occurs via the protein cleavage and assembly of the replicase–
transcriptase complex. Viral RNA is replicated, and structural proteins are synthesized,
assembled, and packaged in the host cell, after which viral particles are released [92]. For
molecular docking, both ligands (PDBQT files), as well as the targets, were selected. The
active binding receptors were examined and lightened using the BIOVIA Discovery Studio
Visualizer (version-19.1.0.18287).
4. Conclusions
The computational molecular docking denotes that the binding energy for GMS was
very low (−8.6 Kcal/mol), which indicates the highest binding affinity to curcumin and a
strong binding to the receptor with the highest solubility tendency for curcumin. Therefore,
glyceryl monostearate (GMS) was chosen as the best solid lipid with the highest drug
solubility among the screened solid lipids. Oleic acid was selected as a liquid lipid for the
NLCs formulation due to its high binding affinity to curcumin and compatibility with GMS
as well. The major absorption peaks of the FTIR with a prominent peak were observed at
1629 cm−1 as identified by the prepared NLC-curcumin. The reduced melting point in the
DSC detection and the XRD results shows a lower peak intensity of the curcumin–NLC
formula, indicating the amorphization of the curcumin crystals.
The low particle size of the novel NLC-curcumin provides a high surface area of
contact with the dissolution medium, which enhanced the solubility of curcumin. The
optimum NLCs formulas had an entrapment efficacy percent (EE%) = 84.23 ± 1.35. The
AFM image confirmed the spherical shape and the low particle size of the prepared NLC.
The novel NLC-curcumin shows an increase in the drug release from the best formula
compared to the other formulations. This could be attributed to the much smaller particle
size, which increases the surface area and, as a result, the drug release rate was increased
with the suspension of curcumin-NLCs. The permeability and penetration release rate in
the intestine and the gut was reported after 8 h and it was found to be 6.83 ± 1.53 and
42.52 ± 3.15 mg/cm2 /h, respectively, due to the small size of NPs and to the presence of
permeation enhancers (poloxamer 188 and tween 80). The future perspectives of the study
include the requirements of determination of the industrial feasibility of the proposed
delivery system, scale-up, and pilot plant studies.
References
1. Quintanar-Guerrero, D.; Leyva-Gómez, G.; Vergara, N.E.M.; Muñoz, N.M. Nanopharmaceuticals. Nanomater. Nanotechnol. Med.
2022, 87–114. [CrossRef]
2. Thi, T.T.H.; Suys, E.J.; Lee, J.S.; Nguyen, D.H.; Park, K.D.; Truong, N.P. Lipid-based nanoparticles in the clinic and clinical trials:
From cancer nanomedicine to COVID-19 vaccines. Vaccines 2021, 9, 359. [CrossRef] [PubMed]
3. Sheng, T.M.; Kumar, P.V. A New Approach for β-cyclodextrin Conjugated Drug Delivery System in Cancer Therapy. Curr. Drug
Deliv. 2022, 19, 266–300. [PubMed]
4. Sakellari, G.I.; Zafeiri, I.; Batchelor, H.; Spyropoulos, F. Formulation design, production and characterisation of solid lipid
nanoparticles (SLN) and nanostructured lipid carriers (NLC) for the encapsulation of a model hydrophobic active. Food Hydrocoll.
Health 2021, 1, 100024. [CrossRef] [PubMed]
5. Li, N.; Li, X.; Cheng, P.; Yang, P.; Shi, P.; Kong, L.; Liu, H. Preparation of Curcumin Solid Lipid Nanoparticles Loaded with
Flower-Shaped Lactose for Lung Inhalation and Preliminary Evaluation of Cytotoxicity In Vitro. Evid. Based Complement. Altern.
Med. 2021, 2021. [CrossRef]
6. Baldim, I.; Souza, C.R.F.; Oliveira, W.P. Encapsulation of Essential Oils in Lipid-Based Nanosystems. In Phytotechnology; CRC
Press: Boca Raton, FL, USA, 2021; pp. 197–230.
7. Gupta, T.; Singh, J.; Kaur, S.; Sandhu, S.; Singh, G.; Kaur, I.P. Enhancing bioavailability and stability of curcumin using solid lipid
nanoparticles (CLEN): A covenant for its effectiveness. Front. Bioeng. Biotechnol. 2020, 8, 879. [CrossRef]
8. Zhong, Q.; Zhang, L. Nanoparticles fabricated from bulk solid lipids: Preparation, properties, and potential food applications.
Adv. Colloid Interface Sci. 2019, 273, 102033. [CrossRef]
9. Mura, P.; Maestrelli, F.; D’Ambrosio, M.; Luceri, C.; Cirri, M. Evaluation and comparison of solid lipid Nanoparticles (SLNs)
and Nanostructured lipid carriers (NLCs) as vectors to develop Hydrochlorothiazide effective and safe pediatric oral liquid
formulations. Pharmaceutics 2021, 13, 437. [CrossRef]
10. Ban, C.; Jo, M.; Park, Y.H.; Kim, J.H.; Han, J.Y.; Lee, K.W.; Kweon, D.-H.; Choi, Y.J. Enhancing the oral bioavailability of curcumin
using solid lipid nanoparticles. Food Chem. 2020, 302, 125328. [CrossRef]
11. Wang, G.; Li, R.; Parseh, B.; Du, G. Prospects and challenges of anticancer agents’ delivery via chitosan-based drug carriers to
combat breast cancer: A review. Carbohydr. Polym. 2021, 268, 118192. [CrossRef]
12. Degot, P.; Huber, V.; El Maangar, A.; Gramüller, J.; Rohr, L.; Touraud, D.; Zemb, T.; Gschwind, R.M.; Kunz, W. Triple role of
sodium salicylate in solubilization, extraction, and stabilization of curcumin from Curcuma Longa. J. Mol. Liq. 2021, 329, 115538.
[CrossRef]
13. Bagherniya, M.; Darand, M.; Askari, G.; Guest, P.C.; Sathyapalan, T.; Sahebkar, A. The clinical use of curcumin for the treatment
of rheumatoid arthritis: A systematic review of clinical trials. In Studies on Biomarkers and New Targets in Aging Research in Iran;
Springer International Publishing: Cham, Switzerland, 2021; pp. 251–263.
14. Wünsche, S.; Yuan, L.; Seidel-Morgenstern, A.; Lorenz, H. A Contribution to the Solid State Forms of Bis (demethoxy) curcumin:
Co-Crystal Screening and Characterization. Molecules 2021, 26, 720. [CrossRef]
15. Lohani, A.; Verma, A. Lipid vesicles: Potential nanocarriers for the delivery of essential oils to combat skin aging. In Nanotechnology
for the Preparation of Cosmetics Using Plant-Based Extracts; Elsevier: Amsterdam, The Netherlands, 2022; pp. 131–156.
16. Sabet, S.; Rashidinejad, A.; Melton, L.D.; McGillivray, D.J. Recent advances to improve curcumin oral bioavailability. Trends Food
Sci. Technol. 2021, 110, 253–266. [CrossRef]
17. Komal, K.; Chaudhary, S.; Yadav, P.; Parmanik, R.; Singh, M. The therapeutic and preventive efficacy of curcumin and its
derivatives in esophageal cancer. Asian Pac. J. Cancer Prev. APJCP 2019, 20, 1329. [CrossRef]
18. Wang, F.; Chen, J.; Dai, W.; He, Z.; Zhai, D.; Chen, W. Pharmacokinetic studies and anticancer activity of curcumin-loaded
nanostructured lipid carriers. Acta Pharm. 2017, 67, 357–371. [CrossRef]
19. Pramanik, S.; Sali, V. Connecting the dots in drug delivery: A tour d’horizon of chitosan-based nanocarriers system. Int. J. Biol.
Macromol. 2021, 169, 103–121. [CrossRef]
20. Luis Abreu, J. Artemisia annua+ Zinc for the Treatment of COVID-19: A Potential Successful Combination Therapy with
Ivermectin. Rev. Daena Rev. Daena (Int. J. Good Conscienc.) 2021, 16.
21. Fai, T.K.; Yee, G.H.; Kumar, P.V.; Elumalai, M. Preparation of Chitosan Particles as a Delivery System for Tetrahydrocurcumin:
β-cyclodextrin Inclusive Compound for Colorectal Carcinoma. Curr. Drug Ther. 2021, 16, 430–438. [CrossRef]
22. Obergrussberger, A.; Friis, S.; Brüggemann, A.; Fertig, N. Automated Patch Clamp in Drug Discovery: Major Breakthroughs and
Innovation in the Last Decade; Taylor & Francis: Milton Park, UK, 2021; Volume 16, pp. 1–5.
Molecules 2023, 28, 1833 21 of 23
23. Luo, Y.; Peng, J.; Ma, J. Next Decade’s AI-Based Drug Development Features Tight Integration of Data and Computation. Health
Data Sci. 2022, 2022, 9816939. [CrossRef]
24. Sabanés Zariquiey, F.; Jacoby, E.; Vos, A.; van Vlijmen, H.W.; Tresadern, G.; Harvey, J. Divide and Conquer. Pocket-Opening
Mixed-Solvent Simulations in the Perspective of Docking Virtual Screening Applications for Drug Discovery. J. Chem. Inf. Model.
2022, 62, 533–543. [CrossRef]
25. Bredewout, M. Virtual Screening in Multi-Target Drug Design. Master’s Thesis, Utrecht University, Utrecht, The Netherlands,
2022.
26. Bragina, M.E.; Daina, A.; Perez, M.A.; Michielin, O.; Zoete, V. The SwissSimilarity 2021 Web Tool: Novel Chemical Libraries
and Additional Methods for an Enhanced Ligand-Based Virtual Screening Experience. Int. J. Mol. Sci. 2022, 23, 811. [CrossRef]
[PubMed]
27. Elisi, G.M.; Scalvini, L.; Lodola, A.; Bedini, A.; Spadoni, G.; Rivara, S. In silico drug discovery of melatonin receptor ligands with
therapeutic potential. Expert Opin. Drug Discov. 2022, 17, 343–354. [CrossRef] [PubMed]
28. Maki, M.A.A.; Kumar, P.V.; Cheah, S.-C.; Siew Wei, Y.; Al-Nema, M.; Bayazeid, O.; Majeed, A.B.B.A. Molecular modeling-based
delivery system enhances everolimus-induced apoptosis in Caco-2 cells. ACS Omega 2019, 4, 8767–8777. [CrossRef] [PubMed]
29. Galazzo, L.; Bordignon, E. Electron Paramagnetic Resonance Spectroscopy in structural-dynamic studies of large protein
complexes. In Progress in Nuclear Magnetic Resonance Spectroscopy; Elsevier: Amsterdam, The Netherlands, 2022.
30. Kabir, A.; Muth, A. Polypharmacology: The Science of Multi-targeting Molecules. Pharmacol. Res. 2022, 176, 106055. [CrossRef]
[PubMed]
31. Semple, S.C.; Leone, R.; Barbosa, C.J.; Tam, Y.K.; Lin, P.J. Lipid Nanoparticle Delivery Systems to Enable mRNA-Based Therapeu-
tics. Pharmaceutics 2022, 14, 398. [CrossRef]
32. Ferreira, L.G.; Dos Santos, R.N.; Oliva, G.; Andricopulo, A.D. Molecular docking and structure-based drug design strategies.
Molecules 2015, 20, 13384–13421. [CrossRef]
33. Boufissiou, A.; Abdalla, M.; Sharaf, M.; Al-Resayes, S.I.; Imededdine, K.; Alam, M.; Yagi, S.; Azam, M.; Yousfi, M. In-silico
investigation of phenolic compounds from leaves of Phillyrea Angustifolia L. as a potential inhibitor against the SARS-CoV-2
main protease (Mpro PDB ID: 5R83) using a virtual screening method. J. Saudi Chem. Soc. 2022, 26, 101473. [CrossRef]
34. Zhan, J.; Lei, Z.; Zhang, Y. Non-covalent interactions of graphene surface: Mechanisms and applications. Chem 2022, 8, 947–979.
[CrossRef]
35. Yashvardhini, N.; Samiksha, S.; Jha, D.K. Pharmacological intervention of various Indian medicinal plants in combating COVID-19
infection. Biomed. Res. Ther. 2021, 8, 4461–4475.
36. Pandeya, K.; Ganeshpurkar, A.; Mishra, M.K. Natural RNA dependent RNA polymerase inhibitors: Molecular docking studies of
some biologically active alkaloids of Argemone mexicana. Med. Hypotheses 2020, 144, 109905. [CrossRef]
37. Tee, Y.N.; Kumar, P.V.; Maki, M.A.; Elumalai, M.; Rahman, S.A.; Cheah, S.-C. Mucoadhesive Low Molecular Chitosan Complexes
to Protect rHuKGF from Proteolysis: In-vitro Characterization and FHs 74 Int Cell Proliferation Studies. Curr. Pharm. Biotechnol.
2021, 22, 969–982. [CrossRef]
38. Kinzhalov, M.A.; Kashina, M.V.; Mikherdov, A.S.; Mozheeva, E.A.; Novikov, A.S.; Smirnov, A.S.; Ivanov, D.M.; Kryukova, M.A.;
Ivanov, A.Y.; Smirnov, S.N. Dramatically Enhanced Solubility of Halide-Containing Organometallic Species in Diiodomethane:
The Role of Solvent Complex Halogen Bonding. Angew. Chem. 2018, 130, 12967–12971. [CrossRef]
39. Hasib, R.A.; Ali, M.C.; Rahman, M.S.; Rahman, M.M.; Ahmed, F.F.; Mashud, M.A.A.; Islam, M.A.; Jamal, M.A.H.M. A com-
putational biology approach for the identification of potential SARS-CoV-2 main protease inhibitors from natural essential oil
compounds. F1000Research 2022, 10, 1313. [CrossRef]
40. Nakmode, D.; Bhavana, V.; Thakor, P.; Madan, J.; Singh, P.K.; Singh, S.B.; Rosenholm, J.M.; Bansal, K.K.; Mehra, N.K. Fundamental
Aspects of Lipid-Based Excipients in Lipid-Based Product Development. Pharmaceutics 2022, 14, 831. [CrossRef]
41. Azmi, N.A.N.; Elgharbawy, A.A.; Motlagh, S.R.; Samsudin, N.; Salleh, H.M. Nanoemulsions: Factory for food, pharmaceutical
and cosmetics. Processes 2019, 7, 617. [CrossRef]
42. Awais, M.; Ullah, N.; Ahmad, J.; Sikandar, F.; Ehsan, M.M.; Salehin, S.; Bhuiyan, A.A. Heat transfer and pressure drop performance
of Nanofluid: A state-of-the-art review. Int. J. Thermofluids 2021, 9, 100065. [CrossRef]
43. Saghafi, Z.; Mohammadi, M.; Mahboobian, M.M.; Derakhshandeh, K. Preparation, characterization, and in vivo evaluation of
perphenazine-loaded nanostructured lipid carriers for oral bioavailability improvement. Drug Dev. Ind. Pharm. 2021, 47, 509–520.
44. Ajala, T.O.; Abraham, A.; Keck, C.M.; Odeku, O.A.; Elufioye, T.O.; Olopade, J.O. Shea butter (Vitellaria paradoxa) and Pentaclethra
macrophylla oil as lipids in the formulation of Nanostructured lipid carriers. Sci. Afr. 2021, 13, e00965.
45. Erawati, T.; Agustin, E.D.; Meldaviati, G.; Rohmah, N.N.; Purwanti, T.; Miatmoko, A.; Hendradi, E.; Rosita, N.; Hariyadi, D.M.
Effect of Peppermint Oil on the Characteristics and Physical Stability of Nanostructured Lipid Carrier-CoQ10. Trop. J. Nat. Prod.
Res. 2022, 6, 303–307.
46. Chen, P.; Zhang, H.; Cheng, S.; Zhai, G.; Shen, C. Development of curcumin loaded nanostructured lipid carrier based thermosen-
sitive in situ gel for dermal delivery. Colloids Surf. A Physicochem. Eng. Asp. 2016, 506, 356–362. [CrossRef]
47. Rapalli, V.K.; Kaul, V.; Waghule, T.; Gorantla, S.; Sharma, S.; Roy, A.; Dubey, S.K.; Singhvi, G. Curcumin loaded nanostructured
lipid carriers for enhanced skin retained topical delivery: Optimization, scale-up, in-vitro characterization and assessment of
ex-vivo skin deposition. Eur. J. Pharm. Sci. 2020, 152, 105438. [CrossRef] [PubMed]
Molecules 2023, 28, 1833 22 of 23
48. Selvaraj, K.; Yoo, B.-K. Curcumin-loaded nanostructured lipid carrier modified with partially hydrolyzed ginsenoside. AAPS
PharmSciTech 2019, 20, 1–9. [CrossRef] [PubMed]
49. Rajasekar, A.; Devasena, T.; Suresh, S.; Senthil, B.; Sivaramakrishnan, R.; Pugazhendhi, A. Curcumin nanospheres and nanorods:
Synthesis, characterization and anticancer activity. Process Biochem. 2022, 112, 248–253. [CrossRef]
50. Lin, D.; Xiao, L.; Qin, W.; Loy, D.A.; Wu, Z.; Chen, H.; Zhang, Q. Preparation, characterization and antioxidant properties of
curcumin encapsulated chitosan/lignosulfonate micelles. Carbohydr. Polym. 2022, 281, 119080. [CrossRef]
51. Muntean, P.; Socaciu, C.; Socaciu, M.A. Lipid Nanostructured Particles as Emerging Carriers for Targeted Delivery of Bioactive
Molecules: Applications in Food and Biomedical Sciences (An Overview). Bull. UASVM Food Sci. Technol. 2020, 77, 37–46.
[CrossRef]
52. Behbahani, E.S.; Ghaedi, M.; Abbaspour, M.; Rostamizadeh, K.; Dashtian, K. Curcumin loaded nanostructured lipid carriers:
In vitro digestion and release studies. Polyhedron 2019, 164, 113–122. [CrossRef]
53. Beraldo-Araújo, V.L.; Vicente, A.F.S.; van Vliet Lima, M.; Umerska, A.; Souto, E.B.; Tajber, L.; Oliveira-Nascimento, L. Levofloxacin
in nanostructured lipid carriers: Preformulation and critical process parameters for a highly incorporated formulation. Int. J.
Pharm. 2022, 626, 122193. [CrossRef]
54. Agrawal, M.; Saraf, S.; Pradhan, M.; Patel, R.J.; Singhvi, G.; Alexander, A. Design and optimization of curcumin loaded nano lipid
carrier system using Box-Behnken design. Biomed. Pharmacother. 2021, 141, 111919. [CrossRef]
55. Sharma, A.; Baldi, A. Nanostructured lipid carriers: A review. J. Dev. Drugs 2018, 7, 1000191.
56. Amin, M.K.; Boateng, J.S. Enhancing stability and mucoadhesive properties of chitosan nanoparticles by surface modification
with sodium alginate and polyethylene glycol for potential oral mucosa vaccine delivery. Mar. Drugs 2022, 20, 156. [CrossRef]
57. Yuan, Y.; Zhang, S.; Ma, M.; Wang, D.; Xu, Y. Encapsulation and delivery of curcumin in cellulose nanocrystals nanoparticles
using pH-driven method. LWT 2022, 155, 112863. [CrossRef]
58. Chang, S.-H.; Lin, H.-T.V.; Wu, G.-J.; Tsai, G.J. pH Effects on solubility, zeta potential, and correlation between antibacterial
activity and molecular weight of chitosan. Carbohydr. Polym. 2015, 134, 74–81. [CrossRef]
59. Shah, R.M.; Eldridge, D.S.; Palombo, E.A.; Harding, I.H. Stability mechanisms for microwave-produced solid lipid nanoparticles.
Colloids Surf. A Physicochem. Eng. Asp. 2022, 643, 128774. [CrossRef]
60. Garg, N.K.; Sharma, G.; Singh, B.; Nirbhavane, P.; Tyagi, R.K.; Shukla, R.; Katare, O. Quality by Design (QbD)-enabled
development of aceclofenac loaded-nano structured lipid carriers (NLCs): An improved dermatokinetic profile for inflammatory
disorder (s). Int. J. Pharm. 2017, 517, 413–431. [CrossRef]
61. Safarpour, F.; Kharaziha, M.; Emadi, R. Inspiring biomimetic system based on red blood cell membrane vesicles for effective
curcumin loading and release. Int. J. Pharm. 2022, 613, 121419. [CrossRef]
62. Aditya, N.; Shim, M.; Lee, I.; Lee, Y.; Im, M.-H.; Ko, S. Curcumin and genistein coloaded nanostructured lipid carriers: In vitro
digestion and antiprostate cancer activity. J. Agric. Food Chem. 2013, 61, 1878–1883. [CrossRef]
63. Ganassin, R.; da Silva, V.C.M.; Araujo, V.H.S.; Tavares, G.R.; da Silva, P.B.; Cáceres-Vélez, P.R.; Porcel, J.E.M.; Rodrigues, M.C.;
Andreozzi, P.; Fernandes, R.P. Solid lipid nanoparticles loaded with curcumin: Development and in vitro toxicity against CT26
cells. Nanomedicine 2022, 17, 167–179. [CrossRef]
64. Zhang, J.; He, Y.; Jiang, J.; Li, M.; Jin, C.; Wang, L.; Wang, D. In vitro and in vivo evaluation of folate-mediated PEGylated
nanostructured lipid carriers for the efficient delivery of furanodiene. Drug Dev. Ind. Pharm. 2017, 43, 1610–1618. [CrossRef]
65. Al-Hmoud, H.A.; Ibrahim, N.E.; El-Hallous, E.I. Surfactants solubility, concentration and the other formulations effects on the
drug release rate from a controlled-release matrix. Afr. J. Pharm. Pharmacol. 2014, 8, 364–371.
66. Katona, G.; Sipos, B.; Ambrus, R.; Csóka, I.; Szabó-Révész, P. Characterizing the Drug-Release Enhancement Effect of Surfactants
on Megestrol-Acetate-Loaded Granules. Pharmaceuticals 2022, 15, 113. [CrossRef]
67. Kontomaris, S.V.; Stylianou, A.; Malamou, A. Atomic Force Microscopy Nanoindentation Method on Collagen Fibrils. Materials
2022, 15, 2477. [CrossRef] [PubMed]
68. Maw, P.D.; Pienpinijtham, P.; Pruksakorn, P.; Jansook, P. Cyclodextrin-based Pickering nanoemulsions containing amphotericin B:
Part II. Formulation, antifungal activity, and chemical stability. J. Drug Deliv. Sci. Technol. 2022, 69, 103174. [CrossRef]
69. Barman, D.; Narang, K.; Parui, R.; Zehra, N.; Khatun, M.N.; Adil, L.R.; Iyer, P.K. Review on recent trends and prospects in
π-conjugated luminescent aggregates for biomedical applications. Aggregate 2022, 3, e172. [CrossRef]
70. Castelli, R.; Ibarra, M.; Faccio, R.; Miraballes, I.; Fernández, M.; Moglioni, A.; Cabral, P.; Cerecetto, H.; Glisoni, R.J.; Calzada,
V. T908 Polymeric Micelles Improved the Uptake of Sgc8-c Aptamer Probe in Tumor-Bearing Mice: A Co-Association Study
between the Probe and Preformed Nanostructures. Pharmaceuticals 2021, 15, 15. [CrossRef]
71. Rubab, S.; Naeem, K.; Rana, I.; Khan, N.; Afridi, M.; Ullah, I.; Shah, F.A.; Sarwar, S.; ud Din, F.; Choi, H.-I. Enhanced
neuroprotective and antidepressant activity of curcumin-loaded nanostructured lipid carriers in lipopolysaccharide-induced
depression and anxiety rat model. Int. J. Pharm. 2021, 603, 120670. [CrossRef]
72. Parhi, R. The Role of Phytocompounds in Cosmeceutical Applications. In Medicinal Plants: Chemistry, Pharmacology, and Therapeutic
Applications; CRC Press: Boca Raton, FL, USA, 2019; pp. 139–160.
73. Ahmad, M.Z.; Ahmad, J.; Aslam, M.; Khan, M.A.; Alasmary, M.Y.; Abdel-Wahab, B.A. Repurposed drug against COVID-19:
Nanomedicine as an approach for finding new hope in old medicines. Nano Express 2021, 2, 022007. [CrossRef]
74. Pawar, R.P.; Rohane, S.H. Role of Autodock vina in PyRx Molecular Docking. Asian J. Res. Chem. 2021, 14, 132–134.
Molecules 2023, 28, 1833 23 of 23
75. Pathak, R.K.; Seo, Y.-J.; Kim, J.-M. Structural insights into inhibition of PRRSV Nsp4 revealed by structure-based virtual screening,
molecular dynamics, and MM-PBSA studies. J. Biol. Eng. 2022, 16, 1–11. [CrossRef]
76. Ram, H.; Sharma, R.; Dewan, D.; Singh, A. Insight view on pandemic COVID-19: Worldwide perspective with homoeopathic
management through repertorial analysis. Homœopathic Links 2020, 33, 180–195. [CrossRef]
77. Pawar, S.S.; Rohane, S.H. Review on discovery studio: An important tool for molecular docking. Asian J. Res. Chem 2021, 14,
86–88. [CrossRef]
78. Xanthopoulou, E.; Zamboulis, A.; Terzopoulou, Z.; Bikiaris, D.N.; Kourtidou, D.; Tarani, E.; Chrissafis, K.; Papageorgiou, G.Z.
Towards novel lignin-based aromatic polyesters: In-depth study of the thermal degradation and crystallization of poly (propylene
vanillate). Thermochim. Acta 2022, 709, 179145. [CrossRef]
79. Xing, R.; Mustapha, O.; Ali, T.; Rehman, M.; Zaidi, S.; Baseer, A.; Batool, S.; Mukhtiar, M.; Shafique, S.; Malik, M. Development,
characterization, and evaluation of SLN-loaded thermoresponsive hydrogel system of topotecan as biological macromolecule for
colorectal delivery. BioMed Res. Int. 2021, 2021. [CrossRef]
80. Gordillo-Galeano, A.; Mora-Huertas, C.E. Hydrodynamic diameter and zeta potential of nanostructured lipid carriers: Emphasiz-
ing some parameters for correct measurements. Colloids Surf. A Physicochem. Eng. Asp. 2021, 620, 126610. [CrossRef]
81. Reddy, K.B. In Vitro-In Vivo Characterization of Oleuropein loaded Nanostructured Lipid Carriers in the Treatment of Streptococ-
cus pneumoniae induced Meningitis. Asian J. Pharm. 2019, 13. [CrossRef]
82. Onugwu, A.L.; Attama, A.A.; Nnamani, P.O.; Onugwu, S.O.; Onuigbo, E.B.; Khutoryanskiy, V.V. Development and optimization
of solid lipid nanoparticles coated with chitosan and poly (2-ethyl-2-oxazoline) for ocular drug delivery of ciprofloxacin. J. Drug
Deliv. Sci. Technol. 2022, 74, 103527. [CrossRef]
83. Rahamathulla, M.; Saisivam, S.; Alshetaili, A.; Hani, U.; Gangadharappa, H.V.; Alshehri, S.; Ghoneim, M.M.; Shakeel, F. Design
and Evaluation of Losartan Potassium Effervescent Floating Matrix Tablets: In Vivo X-ray Imaging and Pharmacokinetic Studies
in Albino Rabbits. Polymers 2021, 13, 3476. [CrossRef]
84. Ulla, M.R.; Saisivam, S. Floating matrix tablet of losartan potassium by using hydrophilic swelling polymer and natural gum.
Turk. J. Pharm. Sci 2013, 10, 435–446.
85. Khan, K.A.; Khan, G.M.; Muzammal, M.; Al Mohaini, M.; Alsalman, A.J.; Al Hawaj, M.A.; Ahmad, A.; Niazi, Z.R.; Shah, K.U.;
Farid, A. Preparation of Losartan Potassium Controlled Release Matrices and In-Vitro Investigation Using Rate Controlling
Agents. Molecules 2022, 27, 864. [CrossRef]
86. Homa, Z.; Ahsan, M.Q.; Rahman, M.M.; Ibrahim, M.; Sultana, N.A.; Haque, S.; Chowdhury, M. Design, Development and In-vitro
Evaluation of Amlodipine besylate Effervescent floating tablets by using different polymers. Pharm. Biol. Eval. 2015, 2, 179–187.
87. Sapavatu, S.N.; Jadi, R.K. Formulation development and characterization of gastroretentive drug delivery systems of loratadine.
Int. J. Appl. Pharm. 2019, 11, 91–99. [CrossRef]
88. Saraogi, G.K.; Tholiya, S.; Mishra, Y.; Mishra, V.; Albutti, A.; Nayak, P.; Tambuwala, M.M. Formulation development and
evaluation of pravastatin-loaded nanogel for hyperlipidemia management. Gels 2022, 8, 81. [CrossRef] [PubMed]
89. Pujals, S.; Feiner-Gracia, N.; Delcanale, P.; Voets, I.; Albertazzi, L. Super-resolution microscopy as a powerful tool to study
complex synthetic materials. Nat. Rev. Chem. 2019, 3, 68–84. [CrossRef]
90. Kurniawan, J.; Ventrici de Souza, J.F.; Dang, A.T.; Liu, G.-Y.; Kuhl, T.L. Preparation and characterization of solid-supported lipid
bilayers formed by Langmuir–Blodgett deposition: A tutorial. Langmuir 2018, 34, 15622–15639. [CrossRef] [PubMed]
91. Presas, E.; McCartney, F.; Sultan, E.; Hunger, C.; Nellen, S.; Alvarez, C.V.; Werner, U.; Bazile, D.; Brayden, D.J.; O’Driscoll, C.M.
Physicochemical, pharmacokinetic and pharmacodynamic analyses of amphiphilic cyclodextrin-based nanoparticles designed to
enhance intestinal delivery of insulin. J. Control. Release 2018, 286, 402–414. [CrossRef]
92. Malik, Y.A. Properties of coronavirus and SARS-CoV-2. Malays. J. Pathol. 2020, 42, 3–11.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.