You are on page 1of 15

International Journal of Pharmaceutics 640 (2023) 123035

Contents lists available at ScienceDirect

International Journal of Pharmaceutics


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

Aqueous cannabidiol β-cyclodextrin complexed polymeric micelle nasal


spray to attenuate in vitro and ex vivo SARS-CoV-2-induced cytokine storms
Narumon Changsan a, Somchai Sawatdee b, Roongnapa Suedee c,
Charisopon Chunhachaichana d, Teerapol Srichana d, *
a
College of Pharmacy, Rangsit University, Pathumtani 12000, Thailand
b
Drug and Cosmetics Excellence Center and School of Pharmacy, Walailak University, Thasala, Nakhon Si Thammarat 80161, Thailand
c
Molecular Recognition Materials Research Unit, Nanotec-PSU Center of Excellence on Drug Delivery System Department of Pharmaceutical Chemistry, Faculty of
Pharmaceutical Sciences, Prince of Songkla University Hat Yai, Songkhla 90112, Thailand
d
Drug Delivery System Excellence Center, Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai,
Songkhla 90112, Thailand

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

Keywords: Cannabidiol (CBD) has a number of biological effects by acting on the cannabinoid receptors CB1 and CB2. CBD
Cannabidiol may be involved in anti-inflammatory processes via CB1 and CB2 receptors, resulting in a decrease of pro-
β-Cyclodextrin inflammatory cytokines. However, CBD’s poor aqueous solubility is a major issue in pharmaceutical applica­
Polymeric micelles
tions. The aim of the present study was to develop and evaluate a CBD nasal spray solution. A water-soluble CBD
Nasal spray solution
SARS-CoV-2
was prepared by complexation with β-cyclodextrin (β-CD) at a stoichiometric ratio of 1:1 and forming polymeric
Pro-inflammatory cytokine micelles using poloxamer 407. The mixture was then lyophilized and characterized using FT-IR, DSC, and TGA.
CBD-β-CD complex-polymeric micelles were formulated for nasal spray drug delivery. The physicochemical
properties of the CBD-β-CD complex-polymeric micelle nasal spray solution (CBD-β-CDPM-NS) were assessed.
The results showed that the CBD content in the CBD-β-CD complex polymeric micelle powder was 102.1 ± 0.5%
labeled claim. The CBD-β-CDPM-NS was a clear colorless isotonic solution. The particle size, zeta potential, pH
value, and viscosity were 111.9 ± 0.7 nm, 0.8 ± 0.1 mV, 6.02 ± 0.02, and 12.04 ± 2.64 cP, respectively. This
formulation was stable over six months at ambient temperature. The CBD from CBD-β-CDPM-NS rapidly released
to 100% within 1 min. Ex vivo permeation studies of CBD-β-CDPM-NS through porcine nasal mucosa revealed a
permeation rate of 4.8 μg/cm2/min, which indicated that CBD was effective in penetrating nasal epithelial cells.
CBD-β-CDPM-NS was tested for its efficacy and safety in terms of cytokine production from nasal immune cells
and toxicity to nasal epithelial cells. The CBD-β-CDPM-NS was not toxic to nasal epithelial at the concentration of
CBD equivalent to 3.125–50 μg/mL. When the formulation was subjected to bioactivity testing against monocyte-
like macrophage cells, it proved that the CBD-β-CDPM-NS has the potential to inhibit inflammatory cytokines.
CBD-β-CDPM-NS demonstrated the formulation’s ability to reduce the cytokine produced by S-RBD stimulation
in ex vivo porcine nasal mucosa in both preventative and therapeutic modes.

Abbreviations: β-CD, beta-cyclodextrin; γ-CD, gamma-cyclodextrin; ACE2, angiotensin-converting enzyme 2; CB1, endo-cannabinoid receptors 1; CB2, endo-
cannabinoid receptors 2; CBD, cannabidiol; CBD-β-CD, cannabidiol and beta cyclodextrin complex; CBD-β-CDPM, cannabidiol and beta cyclodextrin complex
polymeric micelles; CBD-β-CDPM-NS, cannabidiol and beta cyclodextrin complex polymeric micelle nasal spray solution; CD, cyclodextrin; COVID-19, Corona virus
disease-19; DM-β-CD, dimethyl beta cyclodextrin; DMEM, Dulbecco’s Modified Eagle Medium; DSC, differential scanning calorimetry; ELISA, enzyme-linked
immunosorbent assay; FBS, fetal bovine serum; FT-IR, Fourier transform infrared spectroscopy; HEC, hydroxyethyl cellulose; HP-β-CD, hydroxypropyl beta cyclo­
dextrin; HPLC, high-performance liquid chromatography; IL-1β, interleukin-1β; IL-18, interleukin-18; IL-6, interleukin-6; MTT, 3-(4, 5-dimethylthiazol-2-yl)-2,5-
diphenyl-tetrazolium bromide; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; S-RBD, recombinant spike receptor binding domain of SARS-CoV-
2; TGA, thermogravimetric analysis; TNF-α, tumor necrosis factor-α.
* Corresponding author.
E-mail address: teerapol.s@psu.ac.th (T. Srichana).

https://doi.org/10.1016/j.ijpharm.2023.123035
Received 11 December 2022; Received in revised form 23 April 2023; Accepted 5 May 2023
Available online 12 May 2023
0378-5173/© 2023 Elsevier B.V. All rights reserved.
N. Changsan et al. International Journal of Pharmaceutics 640 (2023) 123035

1. Introduction Improved water solubility using inclusion complexes with cyclo­


dextrins (CDs) has been widely reported (Carneiro et al., 2019;
Coronavirus disease 2019 (COVID-19) is an acute respiratory disease Hădărugă et al., 2019; Fenyvesi et al., 2020 Păduraru et al., 2022).
caused by the severe acute respiratory syndrome coronavirus-2 (SARS- Several research studies reported CBD complexed with α-CD, γ-CD,
CoV-2) (Anil et al., 2021). Clinical manifestation might range from mild β-CD, HP-β-CD, and DM-β-CD (Lv et al., 2019; Li et al., 2021; Nivor­
to severe, or it can be asymptomatic. COVID-19 can cause pneumonia, ozhkin, 2019; Ramalho et al., 2021; Mannila et al., 2007; Hatziagapiou
acute respiratory distress syndrome, and multiorgan failure in severe et al., 2022). CBD/DM-β-CD and CBD/β-CD displayed strong in­
cases. teractions (Li et al., 2021). The water solubility and dissolution rates of
Respiratory distress syndrome, the major cause of COVID-19 death, the CBD/DM-β-CD and CBD/β-CD complexes were 614-fold and 17-fold,
is characterized by the release of pro-inflammatory cytokine storms, respectively (Li et al., 2021). The bioavailability and pharmacokinetic
such as interleukin 6 (IL-6) and interleukin 1β (IL-1β) as well as inter­ parameters of CBD/β-CD inclusion complexes showed that the CBD
leukin 18 (IL-18). In critically ill patients, antiviral medications may be plasma concentration was higher than orally administered ethanolic
insufficient to control the cytokine storm and respiratory distress. It is CBD (Ramalho et al., 2021).
consequently critical to develop novel therapeutic alternatives that can In this study, we developed a nasal aqueous solution of CBD by
decrease the cytokine storm to reduce the adverse outcome (Esposito complexing with β-CD to increase water solubility and then formed
et al., 2020; Ramasamy and Subbian, 2019). polymeric micelles with poloxamer. Poloxamer refers to nonionic tri­
Cannabidiol (CBD) is a non-psychoactive phytocannabinoid synthe­ block copolymers formed by polar (polyethylene oxide) and non-polar
sized by Cannabis sativa (Vallée, 2022; Esposito et al., 2020). CBD is (polypropylene oxide) blocks (Kabanov, et al., 2002). Poloxamer 407
regarded as one of the most fascinating emerging molecules in the has a molecular weight of approximately 12,600 and is highly soluble in
discipline of pharmacology since it has many therapeutic effects related water (Nagy et al., 2020; Kabanov, et al., 2002). Poloxamer 407 is
to its anticonvulsant, sedative, hypnotic, antipsychotic, anticancer, anti- widely used in pharmaceuticals as a solubilizer, release modifier, and
inflammatory, and neuroprotective properties (Esposito et al., 2020). suspending agent (Rowe et al., 2009). Furthermore, poloxamer 407 has
CB1 and CB2 endocannabinoid receptors are involved in anti- thermo-reversible and bioadhesive properties (Dumortier et al., 2006).
inflammatory activities. CBD, as an agonist of the CB2 receptor, has a Poloxamer in aqueous solutions has an amphiphilic nature that leads to
wide spectrum of immunomodulatory and anti-inflammatory proper­ aggregation of their molecules to form polymeric micelles with a hy­
ties, and it can reduce the unregulated cytokine production that causes drophobic interior composed of polypropylene oxide blocks and a hy­
acute lung injury. CBD has recently been considered as a possible drophilic exterior composed of polyethylene oxide blocks (Kabanov,
medication in the treatment of SARS-CoV-2 because it possesses anti- et al., 2002). Poloxamer forms polymeric micelles by self-assembly of
inflammatory and immune-suppressive properties in pre-clinical amphiphilic block copolymers in aqueous solutions (Ghezzi et al., 2021).
COVID-19 models. This compound has the potential to reduce the In a diluted aqueous solution, amphiphilic molecules exist as amphi­
release of pro-inflammatory cytokines, which are responsible for philes. The critical micelle concentration of poloxamer 407 is 2.8 × 10− 6
inflammation during SARS-CoV-2 infection (Vallée, 2022). In addition M (~0.0342 mg/mL) and the hydrophilic-lipophilic balance value is 22
to anti-inflammatory effects, CBD has the ability to reduce the severity (Suksiriworapong et al., 2015; Nagy et al., 2020). Drugs can be encap­
and progression of the disease by down-regulating the expression of sulated in the polymeric micelles during their formation depending on
angiotensin-converting enzyme 2 (ACE2) and transmembrane serine the method used for the preparation and physicochemical characteris­
protease 2, which are crucial viral gateways for SARS-CoV-2 cellular tics of the drug (Ghezzi et al., 2021).
invasion, thereby limiting SARS-CoV-2 entry into vulnerable hosts. The developed CBD-β-cyclodextrin complexed polymeric micelle
Furthermore, CBD, as a peroxisome proliferator-activated receptor-γ nasal spray formulation was tested for nasal cell cytotoxicity and the
agonist, has a direct antiviral effect and is a regulator of fibroblast/ ability to suppress pro-inflammatory cytokines produced from recom­
myofibroblast activation and can limit the formation of pulmonary binant spike receptor binding domain (S-RBD) of SARS-CoV-2 induced
fibrosis, thereby improving lung function in recovered patients (Holst monocyte-like macrophages and ex vivo porcine mucosa explant tissue.
et al., 2022; Esposito et al., 2020; Janecki et al., 2022).
The nasal route can be employed for local or systemic treatment. The 2. Material and methods
nasal cavity is advantageous for systemic absorption due to its high
vascularization, wide surface area-to-volume ratio, and additional 2.1. Materials
benefits over alternative routes (Pires, et al., 2022). However, intranasal
delivery also has its limitations since only a small volume (maximum Cannabidiol was provided gratis by Quantum Biotech Co., Ltd.
150–200 μL in humans) can be applied, therefore potent drugs are (Pathum Thani, Thailand) with 99% purity. β-cyclodextrin (Cavamax
required. In addition, it is complicated developing drugs with poor water W7 FG) was purchased from Wacker Chemical Corporation, Michigan,
solubility at high potency without using significant amounts of poten­ USA. Poloxamer 407 (Kolliphor P407) was purchased from BASF Can­
tially harmful cosolvents or surfactants. CBD is classified as bio­ ada Inc., Mississauga, Canada. Hydroxyethyl cellulose (HEC) was ob­
pharmaceutics classification system Class II with very poor water tained from Chemipan Corporation Co., Ltd., Bangkok, Thailand).
solubility with a solubility of 0.1 μg/mL in water (Amanat et al., 2020), Glycerin was procured from Chanjao Longevity Co., Ltd, Bangkok,
35 mg/mL in ethanol (Cayman Chemical, 2015), and a bioavailability of Thailand. Sodium citrate was purchased from Krungthepchemi Co., Ltd.,
around 6% (Koch et al., 2020). This restricts its application as a nasal Bangkok, Thailand. Citric acid monohydrate was obtained from RFCL
solution (Li et al., 2021). Limited, New Delhi, India. Benzalkonium chloride was obtained from
CBD must be in a solution form at the site of absorption; conse­ Sigma-Aldrich, Co., Darmstadt, Germany. Absolute ethanol and aceto­
quently, CBD in the form of better aqueous solubility may result in nitrile were analytical grade and purchased from RCI Labscan, Bangkok,
greater CBD absorption. Particularly, a more water-soluble CBD would Thailand.
have a wider range of applications in aqueous formulations like solu­
tions, ocular drops, injections, and aerosols. CBD nasal spray was 2.2. Preparation of solid-state CBD-CD inclusion complexed polymeric
developed in various research projects and employed for various ap­ micelles (CBD-β-CDPM)
plications including anti-inflammatory and epilepsy treatments (Paudel
et al., 2010; Preve Ceutical Medical Inc, 2022; Patent WO2017/ CBD 7.9 g (25 mmol) was dissolved in 78.0 g of the warm ethanol at
208072A2). CBD intranasal administration has rapid absorption 50 ◦ C and stirred with a magnetic stirrer (C-MAG HS 7, IKA® Works Co.
compared to oral delivery (Polidoro et al., 2022). Ltd, Bangkok, Thailand.) until a clear solution was obtained that

2
N. Changsan et al. International Journal of Pharmaceutics 640 (2023) 123035

Table 1 2.4. Assay of CBD in the CBD-β-CDPM-NS formulation


Composition of the CBD-β-CD complexed polymeric micelle (CBD-β-CDPM)
formulation. High-performance liquid chromatography (HPLC) was used to
Ingredients Amount (g) Function analyze the CBD content in the CBD-β-CDPM powder and CBD-β-CDPM-
Cannabidiol 7.9 Active ingredient
NS formulation. The HPLC system included the CBM-20A system
β-Cyclodextrin 28.4 Complexing agent controller (Shimadzu Corporation, Tokyo, Japan), LC-30AD pump, SIL-
Poloxamer 407 9.9 Solubilizing agent 30AC autosampler, SPD-M20A PDA detector, and CTO-20AC column
Ethanol 78.0 Solvent of CBD oven. A Shim-pack GIS C18 (150 mm × 4.6 mm, 3 μm) from Shimadzu
Purified water 875.8 Vehicle
(Shimadzu Corporation, Tokyo, Japan) at 15 ◦ C was used to perform the
Total weight 1000
separations. A degassed mixture of 70% acetonitrile and 30% ultrapure
Note: The molar ratio of cannabidiol, β-cyclodextrin, and poloxamer 407 was water made up the mobile phase. The sample or standard CBD substance
approximately 1:1:0.03. was dissolved in the mobile phase. The injection volume and flow rate
were 10 μL and 1 mL/min, respectively. A spectrophotometer was used
to monitor the separation at 207 nm. LC Solution software (Shimadzu
Table 2
Composition of the CBD-β-CDPM nasal spray solution.
Corporation, Tokyo, Japan) was used to process the acquired data. The
validation parameters included precision, accuracy, specificity, limit of
Ingredients Amount (g) Function
quantitation, limit of detection, linearity, and robustness. There was no
CBD-β-CDPM powder 0.30* Active ingredient matrix interference of either CBD-β-CDPM powder or CBD-β-CDPM-NS
Hydroxyethyl cellulose (HEC) 0.05 Mucoadhesive polymer in the chromatographic analysis.
Citric acid monohydrate 0.20 Buffering agent
Sodium citrate 0.28 Buffering agent
Glycerin 2.15 Humectant 2.5. Characterization of dried CBD-β-CDPM powder
Ethanol (99.8%) 2.00 Permeation enhancer
Benzalkonium chloride 0.02 Preservative The moisture content of the dried CBD-β-CDPM powder was
Sterile water for injection 95.00 Vehicle
analyzed using thermogravimetric analysis (TGA). Samples were accu­
*
Note: The 0.30 g of CBD-β-CDPM powder is equivalent to 0.05 g of pure CBD. rately weighed and subjected to heating at a constant rate of 10 ◦ C/min
from 30 to 150 ◦ C under a nitrogen purge using a thermogravimetric
resulted in an alcoholic CBD solution. Poloxamer 407, 9.9 g (0.79 mmol) analyzer (Model TGA 7 PerkinElmer, Inc., USA).
and β-CD 28.4 g (25 mmol) were dispersed in separate vessels in 400 mL All samples including CBD, β-CD, poloxamer 407, CBD-β-CD, CBD-
and 476 mL, respectively, of water at 70 ◦ C by slow addition and stirring β-CDPM powder, and the physical mixing of CBD, β-CD, and poloxamer
with a high shear mixer (IKA RW 20, Co. Ltd, Bangkok, Thailand) at 407 were subjected to differential scanning calorimetry (DSC). DSC
1000 rpm for at least 60 min or until a clear solution was formed. The analysis was carried out on a differential scanning calorimeter (DSC 800,
aqueous β-CD solution and aqueous poloxamer 407 solution were then Perkin Elmer Inc., USA). Each sample (5–10 mg) was heated at a rate of
combined to create a homogenous mixture. The aqueous mixture of 10 ◦ C/min from 30 to 300 ◦ C under a flowing argon atmosphere (flow
β-CD and poloxamer 407 was then mixed with CBD ethanolic solution rate: 20 mL/min) in an aluminum pan.
and the mixture was stirred at a low shear rate (500 rpm) for at least 30 All samples including CBD, β-CD, poloxamer 407, CBD-β-CD, and
min or until a uniform dispersion was created. As a consequence, 1000 g CBD-β-CDPM powder were determined by Fourier transform infrared
of the combination were produced. (FT-IR) spectroscopy. The sample powder (1 mg) was mixed with KBr in
The mixture solution was freeze-dried to produce a water-soluble a 1:100 ratio following by compression to obtain 2 mm transparent disc.
CBD-β-CDPM using the Martin Christ Model Delta 2–24 LSC plus FT-IR spectra between 4000 and 400 cm− 1 were determined after an
freeze-dryer (Osterode am Harz, Germany) using the following condi­ accumulation of 16 scans using a Spectrum One (Perkin Elmer, Inc.,
tions: pre-freeze temperature at − 40 ◦ C for 24 h; primary drying at USA).
− 10 ◦ C for 24 h at a chamber pressure of 0.633 mbar; and secondary
drying at 10 ◦ C for 4 h and 20 ◦ C for 20 h. The schematic diagram of 2.6. Characterization of CBD-β-CDPM-NS
CBD-β-CDPM preparation is illustrated in Supplementary Fig. S1, and
the ingredients of the CBD-β-CDPM are listed in Table 1. CBD-β-CDPM was dispersed in a purified water at a concentration of
0.3% w/v which was equal to the concentration of CBD-β-CDPM in the
2.3. Preparation of the nasal spray solution containing CBD-β-CD CBD-β-CDPM-NS formulation. The particle size distribution and zeta
complexed polymeric micelles (CBD-β-CDPM-NS) potential were then determined by a Zetasizer (Nano ZS ZEN3600,
Malvern Instruments Ltd., Worcestershire, UK). CBD-β-CDPM-NS was
All ingredients for the nasal spray formulation containing (CBD- also analyzed without dilution. The sample measurements were per­
β-CDPM) are provided in Table 2. For each 100 mL of nasal solution, 20 formed at 25 ◦ C. The average of ten measurements at an angle of 90◦ was
mL of sterile water for injection and 2 g of ethanol were combined. Then, used to calculate the particle size and polydispersity index of the
0.3 g of the dried CBD-β-CDPM powder (equivalent to CBD 50 mg) was investigated formulations.
gradually added during constant stirring. After all excipients were dis­ The viscosity of CBD-β-CDPM-NS was analyzed using a Modular
solved, sodium citrate, benzalkonium chloride, and citric acid mono­ Advanced Rheometer System (HAAKE MARS 60; Thermo Fisher Scien­
hydrate were added. Glycerin was then added in the last step. HEC was tific, Bremen, Germany). The rheometer used a 60 mm diameter parallel
disseminated in sterile water for injection and then homogeneously plate at a gap of 0.5 mm with a Peltier temperature control system. After
combined with the CBD-β-CDPM solution to obtain the nasal spray so­ loading a 3 g sample onto the bottom plate, the upper plate was moved
lution The resultant nasal spray formulation was kept at controlled room to the designed gap. The flow experiments were conducted at a constant
temperature for subsequent analysis. As a control, a formulation without 25 ◦ C with a shear rate of 1 to 1000 s− 1. A frequency sweep was con­
CBD-β-CDPM was prepared. Also, CBD-β-CD was prepared similarly to ducted in a range of 0.1 Hz to 100 kHz. Each batch of tests was per­
the above method without adding poloxamer 407 to compare the formed in triplicate.
interaction or characteristics of CBD and β-CD without surfactant The surface tension of CBD-β-CDPM-NS formulation was measured
interference. by the Du Noüy ring method. The Du Noüy ring is an automated oper­
ation to determine the surface tension. The tensiometer used in this
experiment was a model DY-300 (Kyowa Interface Science Co., Ltd.,

3
N. Changsan et al. International Journal of Pharmaceutics 640 (2023) 123035

Tokyo, Japan). Approximately 200 mL of CBD-β-CDPM-NS was placed in Biohit®, Helsinki, Finland) at 570 nm. The percentage of cell prolifer­
a glass surface tension cup and loaded on the equipment. The ring was ation was calculated and compared to the negative control.
cleaned after each measurement to ensure the residue was completely
removed. All measurements were performed in triplicate. 2.11. In vitro CBD-β-CDPM-NS efficacy to reduce S-RBD-induced pro-
The osmolality of CBD-β-CDPM-NS was determined by an automatic inflammatory cytokine production
cryoscopic osmometer (Osmomat® 030, Gonotec, ELITechGroup Inc.,
USA). All measurements were performed in triplicate. This experiment was performed according to previous studies (Sri­
chana et al., 2022). The coding sequence for the S-RBD of SARS-CoV-2
2.7. Stability study (S-RBD, residue 317–539) was optimized for expression in mammalian
cells from the sequence obtained from the reference strain (Wuhan-Hu-
To determine the stability under actual storage conditions in 1) and cloned into pSecTag using restriction enzyme cloning (BsiWI and
Thailand, which is in a hot and humid zone, 20 mL of the CBD-β-CDPM- XhoI) in-frame with the Igκ leader sequence at the N-terminus and the
NS formulation was placed in a polyethylene nasal spray bottle and kept Myc-His tag at the C-terminus. Recombinant tagged S-RBD protein was
for six months at 30 ± 0.5 ◦ C and at a relative humidity of 75 ± 5% produced according to a previously published protocol (Amanat et al.,
(ASEAN Guideline on Stability of Drug Products, 2018). The content of 2020). Briefly, human embryonic kidney 293 T cells were maintained in
CBD in CBD-β-CDPM-NS and physical properties of the formulation OptiMEM with 10% fetal bovine serum supplement and transfected with
including appearance, pH, particle size, and zeta potential were pSecTag-SRBD using FuGENE transfection reagent according to the
recorded. manufacturer’s protocol. The cell media were then changed to OptiMEM
with no supplement at 6 h post-transfection. At 72 h post-transfection,
2.8. In vitro drug release studies the supplement was conditioned with binding buffer (4X) (1.2 mM
NaCl, 200 mM NaH2PO4, 40 mM imidazole). The conditioned super­
In vitro dissolution of the nasal formulation in a nasal cavity was natant was then mixed with Ni-NTA agarose resin (Qiagen) for 1 h. The
developed according to the previous study (Inoure et al., 2022). The CBD unbound proteins were washed off with a wash buffer (300 mM NaCl,
dissolution of CBD-β-CDPM-NS was evaluated. Phosphate-buffered sa­ 50 mM NaH2PO4, 20 mM imidazole). Recombinant S-RBD was then
line (PBS) pH 6.4 was used as an artificial nasal fluid (Washington et al., eluted with an elution buffer (300 mM NaCl, 50 mM NaH2PO4, 250 mM
2000). The temperature of the medium was kept at 35 ◦ C. A quantity of imidazole). The eluted fractions were pooled, concentrated and buffer-
500 μL of CBD-β-CDPM-NS (equivalent to 50 μg × 5 doses of CBD) was exchanged into PBS using a 10 molecular weight cut-off centrifugal fil­
loaded into the simulated chamber with 10 mL dissolution medium. ter unit. The protein concentration was measured using the Bradford
After dosing, a 100 μL aliquot of medium was collected from the bottom method (Bradford, 1976). Recombinant S-RBD was stored at − 80 ◦ C in
of the chamber at intervals of 0, 1, 2, 3, 4, 5, 7, 10, and 15 min and small aliquots prior to use.
replaced with 100 μL fresh medium. The amount of CBD dissolved was RAW 264.7 cells (100 μL) at a cell density of 105 cells/mL in com­
analyzed by HPLC as described in Section 2.4. plete media were seeded into each well of a 96-well plate. The cells were
allowed to grow until 70–80% confluence for 2 h. Following incubation,
2.9. Cell culture conditions for cell toxicity studies the cells were treated with S-RBD and CBD-β-CDPM-NS in different or­
ders as follows:
The RAW 264.7 mouse monocyte/macrophage cell line (ATCC TIB- Treatment mode: Cells were stimulated with S-RBD protein (10 μg/
71, USA) was grown in Dulbecco’s Modified Eagle Medium (DMEM, mL) and incubated at 37 ◦ C under 5% CO2 for 24 h before treatment with
Gibco®, USA) containing 10% fetal bovine serum (FBS, Gibco®, USA) CBD-β-CDPM-NS containing CBD equivalent to 50 μg/mL and incubated
and 100 U/mL penicillin/streptomycin (Gibco®, USA). The media was at 37 ◦ C under 5% CO2 for 24 h.
replenished every two days while the cells were cultured at 37 ◦ C in a 5% Co-administration mode: Cells were stimulated with S-RBD protein
CO2 incubator. The cells were collected by gentle rocking. A new cell (10 μg/mL) and concurrently treated with CBD-β-CDPM-NS containing
suspension was then prepared by adding fresh culture media, which was CBD equivalent to 50 μg/mL for 24 h at 37 ◦ C in 5% CO2.
then used for further incubation. After 24 h of incubation, the cell supernatant of each experimental
The RPMI 2650 human nasal septum epithelial cell line (ATCC: CCL- mode was assayed for pro-inflammatory cytokine levels that included
30, Rockville, MD, USA) was cultured in Eagle’s Minimum Essential TNF-α, IL-1β, or IL-6, which were generated in the cell supernatants
Medium (EMEM, Gibco®, USA) supplemented with 10% fetal bovine obtained from the experiment explained earlier. Measurements used a
serum (FBS, Gibco®, USA), 100 U/mL penicillin/streptomycin (Gibco®, rat TNF-α, IL-1β, or IL-6 Quantikine enzyme-linked immunosorbent
USA). The cells were incubated at 37 ◦ C in a 5% CO2 incubator and the assay kit (R&D Systems, Inc., Minneapolis, MN, USA). Using the 96-well
media were changed every two days. They were harvested by gentle plates included in the ELISA kit, 50 μL of TNF-α, IL-1β, or IL-6 diluent
rocking, followed by the addition of fresh culture medium to create a was added to each well. The experimental cell supernatant in the
new cell suspension for further incubation. amount of 50 μL was added and incubated for 2 h at room temperature.
Each well was washed (5X) with a buffer solution followed by adding the
2.10. Cell proliferation and viability assay conjugate solution (100 μL) of either TNF-α, IL-1β, or IL-6 to each well
and incubated for another 2 h. Each well was then washed with a buffer
A quantity of 100 μL of RAW 264.7 cells or RPMI 2650 at a con­ (5X) and 100 μL of substrate solution was added. The plates were
centration of 1 × 105 cell/mL was seeded in a 96-well plate with com­ incubated for 30 min at room temperature followed by adding a stop
plete medium. After 24 h of incubation, the culture plates were treated solution (100 μL). The absorbance of a consequent reaction was
with CBD-β-CDPM-NS at CBD concentrations equivalent to 3.12–50 µg/ measured at 450 nm with a microplate reader (Biohit 830, Biohit®,
mL in fresh media. Untreated cells were used as a negative control. After Helsinki, Finland). The absorbance results were quantified using TNF-α,
the cells were exposed to the test samples for 24 h, cell viability was IL-1β, and IL-6 standard curves.
measured using a solution (5 mg/mL) of 3-(4, 5-dimethylthiazol-2-yl)-
2,5-diphenyl-tetrazolium bromide (MTT). Briefly, the cells were 2.12. Ex vivo studies on the activity of CBD-β-CDPM-NS on porcine nasal
treated with 80 µL of fresh media and the MTT solution (20 µL), which epithelia mucosa
was then incubated at 37 ◦ C under 5% CO2 for 4 h. The media containing
MTT were then removed and dimethyl sulfoxide (100 µL) was added. Pigs (22–23 weeks old, 80 kg) were dissected by the veterinary staff
Absorbance was recorded using a microplate reader (Biohit 830, of Charoen Pokphand (CP) Foods Public Company, Ltd, Thailand. To

4
N. Changsan et al. International Journal of Pharmaceutics 640 (2023) 123035

Fig. 1. Schematic diagrams represent an overview of ex vivo experimental design.

maintain the tissue integrity, the nasal mucosa tissue was immediately prevention study, nasal mucosa explants were treated with 25 μL of
removed from the pig snout. The mucosa explants were prepared using CBD-β-CDPM-NS for 30 min before S-RBD protein challenge. Following
the procedure described by Tulinski et al. (2013), with slight modifi­ 6 h of S-RBD incubation in the treatment study, 25 μL of CBD-β-CDPM-
cations. In brief, isolated nasal mucosa explants were placed on sterile NS was applied to the explant surface and incubated for 30 min. After
gauze soaked in DMEM supplemented with 1 mg/mL streptomycin and the incubation time ended, the tissue was diced into small fractions
1000 U/mL penicillin. The air–liquid interface was kept open and the suspended in culture medium (DMEM) and transferred to a micro­
cilia membrane exposed to the air. The process of explant preparation is centrifuge tube, placed in an ice bath, and then subjected to a 10 min
depicted in Fig. 1. sonication process. The generated cytokines (TNF-α, IL-1β, or IL-6) were
Porcine nasal mucosa explants were divided into pieces approxi­ detected in the supernatant after centrifuging the tissue suspension at
mately 1 cm2 and placed into a well of a 24-well plate filled with DMEM 5000 rpm for 5 min. The Quantikine® ELISA Kit (R&D Systems, Inc.,
to the half-height of the explants. The ability of CBD-β-CDPM-NS to Minneapolis, MN, USA) was used to measure the porcine TNF-α, IL-1β,
suppress the cytokine provoked by the S-RBD protein was examined in or IL-6 cytokine levels following the manufacturer’s instructions.
two experimental studies: prevention and treatment. The surface of a
porcine nasal mucosa explant was exposed to S-RBD protein 50 µL (10
µg/mL), which was then incubated for 6 h in a CO2 incubator to activate
the immune system and generate pro-inflammatory cytokines. In the

5
N. Changsan et al. International Journal of Pharmaceutics 640 (2023) 123035

Table 3 epithelium explant membrane. The diffusion cells were maintained in a


Initial physical properties and CBD content of CBD-β-CDPM powder and after circulating water bath at 100 rpm. Samples of 500 μL were collected at
reconstitution to CBD-β-CDPM-NS with stability results (n = 3–6). each time point of 0, 5, 10, 15, 20, 25, 30, 45, and 60 min and fresh PBS
Test Initial data Data after 6 months pH 6.4 was used to replace the equal amounts of sampling. The 500 μL
storage collected samples were diluted with 300 μL of mobile phase. The diluted
CBD-β-CDPM powder samples were filtered with a 0.22 μm nylon membrane filter before the
CBD content (%) 102.1 ± 0.5 100.7 ± 0.6 CBD analysis using HPLC described in Section 2.4. Following the
Appearance White to off-white off-white powder experiment, the leftover porcine nasal mucosa membranes were placed
powder
in centrifuge tubes with an additional 800 μL of mobile phase. The
Water content (%) 4.79 ± 0.3 4.40 ± 0.2
Particle size (Z-average, 101.4 ± 6.9 102.5 ± 7.3 samples were digested with a cell digestion machine (IKA, Bangkok,
nm) * Thailand) and filtered through a 0.22 μm nylon membrane. The super­
PDI 0.25 ± 0.01 0.31 ± 0.02 natant was then subjected to HPLC analysis to determine the CBD con­
Zeta potential (mV)* − 8.9 ± 0.4 − 10.5 ± 0.2 tent retained in the membrane. The experiment was done in quadruplet.
CBD-β-CDPM-NS
CBD content (%) 99.80 ± 1.3 98.80 ± 0.8
From the in vitro membrane permeation data, steady-state flux (Jss) (μg/
Appearance Clear colorless solution Clear colorless solution cm2/min) and the cumulative amount of CBD through the mucosa
pH 6.02 ± 0.02 6.12 ± 0.01 membranes (μg/cm2) were determined.
Osmolality (mOs/kg) 300 ± 0.8 302 ± 0.6
Particle size (Z-average, 111.9 ± 0.7 121.4 ± 1.1
2.14. Statistical analysis
nm)
PDI 0.15 ± 0.01 0.22 ± 0.02
Zeta potential (mV) 0.8 ± 0.1 1.0 ± 0.2 All results were reported as mean and standard deviations. Signifi­
Viscosity (mN s/m2 [cP]) 12.04 ± 2.64 15.20 ± 1.03 cant differences in the mean parameters were analyzed using one-way
All values are presented as mean ± SD. analysis of variance (ANOVA). The significance level was set at 0.05
*
After reconstitution with sterile water for injection. for all tests.

2.13. Ex vivo nasal epithelium permeation studies using Franz diffusion 3. Results and discussion
cells
3.1. Validation results of CBD in the CBD-β-CDPM and CBD-β-CDPM-NS
Porcine nasal epithelium explants were cut into 2 × 2 cm2 pieces and
CBD transport was determined through the membrane. The explants The HPLC analysis was very specific to CBD. The retention time, limit
were mounted between the donor and receptor chamber of the Franz of CBD detection, and limit of quantitation were 3.7 min, 0.25 μg/mL,
diffusion cells (Hanson Technology, Chatsworth, CA USA) with an and 1 μg/mL, respectively. The accuracy and precision of the analysis
effective diffusion area of 1.76 cm2 and cell volume of 8 mL filled with were both 99% with a 1.2% relative standard deviation. The results of
freshly prepared PBS pH 6.4. After an equilibrium time of 15 min at 35 ◦ C the HPLC system were linear over a range of 10–50 μg/mL, and the
± 0.5 ◦ C, 500 μL of CBD-β-CDPM-NS (equivalent to 250 μg CBD) were system was able to give consistent results for different operators.
loaded into the donor compartment through direct contact with nasal

Fig. 2. CBD release from CBD-β-CDPM powder (blue line) and CBD-β-CDPM-NS (black line). Drug release studies were performed by dialysis simulated condition in
the nasal cavity (PBS pH 6.4, 8 mL) under sink conditions while maintaining the temperature at 35 ± 0.5 ◦ C. The data presented is mean of drug release with
standard deviation of 3 replicates. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

6
N. Changsan et al. International Journal of Pharmaceutics 640 (2023) 123035

Fig. 3. CBD from the CBD-β-CDPM-NS permeability through nasal porcine mucosa determined by Franz diffusion cells in PBS. (A) Steady-state flux across nasal
porcine mucosa. (B) Cumulative amount of CBD over time through nasal porcine mucosa. Data are expressed as mean ± SD (n = 4).

3.2. Development of CBD-β-CDPM with dimensions of a = 10.617 Å, b = 10.649 Å, and c = 17.266 Å and β
= 95.30(4)◦ (Jones et al., 1977), and another study reported similar
CBD complexation with CD was developed to improve water solu­ dimensions of a = 10.4395 Å, b = 10.8739 Å, and c = 16.7853 Å and β =
bility. β-CD has undergone extensive safety testing and has been 95.448(1)◦ (Mayr et al., 2017). We confirmed the results of those studies
approved for use in pharmaceutical products (Loftsson and Brewster, from our CBD crystallography as shown in the supplementary data files.
2010; Lv et al., 2019; Haimhoffer et al., 2019). Also, β-CD is safe for These findings indicated that a portion of the CBD molecule could be
human ingestion and application (Pereira et al., 2021; Poulson, et al., inserted into the β-CD cavity of 6.0–6.4 Å. In addition, another group
2022; European Medicines Agency, 2014). Furthermore, β-CD was reported that the β-CD cavity was 7.8 Å (Păduraru et al., 2022). β-CD has
chosen in this study due to the similarity of its internal cavity size of hydrophilic properties with a solubility in 25 ◦ C water of 18 mg/mL
6.0–6.4 Å (Poulson, et al., 2022) and the size of the CBD molecule. From (Chatjigakis et al., 1992). Furthermore, as the temperature is increased,
the literature, the crystal structure of CBD was reported to be monoclinic β-CD becomes more soluble (Poulson, et al., 2022; Yong et al., 2008). For

7
N. Changsan et al. International Journal of Pharmaceutics 640 (2023) 123035

Fig. 4. The cytotoxicity profiles of the CBD-β-CDPM, CBD-β-CDPM-NS with CBD concentrations equivalent to 3.125–50 μg/mL, and nasal spray blank formulation
against the human nasal septum epithelial cell line (RPMI 2650) (A), and mouse monocyte/macrophage cell line (RAW264.7) (B). Data are expressed as mean ± SD
(n = 4).

this reason, the manufacturing process in this study was performed at formulation. The developed water-soluble CBD-β-CDPM is white to an
70 ◦ C. After preparation, CBD-β-CDPM was lyophilized to produce a dry off-white amorphous powder at room temperature after the lyophiliza­
powder. This technique provided the amorphous formulation of CBD as tion process, and the powder is free flowing with a high bulk volume.
reported in a previous study (Koch, et al., 2020). This current method is The yield of water-soluble CBD-β-CDPM was 90.9% (42.0 g) of the total
similar to the method by Koch et al., (2020) except CBD was dissolved in weight of the solid ingredients (46.2 g) in the formulation. The TGA
ethanol before adding it to the CD solution, which resulted in a higher analysis showed moisture content of the water-soluble CBD-β-CDPM was
amount of CBD loaded into the CD molecules. 4.8 ± 0.3% w/w. After reconstituting the CBD-β-CDPM with water for
The CBD encapsulated within the cavity of β-CD to form a 1:2 injection, a clear colorless solution that easily dissolved was obtained.
host–guest inclusion complex is described in the literature (Lv et al., The solubility of CBD-β-CD was 0.4 μg/mL, which is 4 times higher than
2019). Another consistent study prepared CBD as a β-CD inclusion pure CBD. When CBD-β-CD was formulated with poloxamer 407, its
complex. The molar stoichiometry of the CBD and β-CD complex was 1:2 solubility at 25 ◦ C was 2.5 mg/mL, which is equal to 427.5 μg/mL of
by the precipitation method in which only 7 mg of β-CD was required to CBD (theoretically, 46.2 g of CBD-β-CDPM powder contains 7.9 g of
complex 1 mg of CBD (Mannila et al., 2007). However, Li et al. (2022) CBD). Thus, the aqueous solubility of CBD-β-CDPM was 4275 times
reported CBD was inclusion complexed with hydroxypropyl-β-CD at a higher than the solubility of pure CBD at 0.1 μg/mL (Amanat et al.,
ratio of 1:1. 2020), which is beneficial for developing aqueous nasal spray solutions.
In this study, CBD was formulated with β-CD in a 1:1 ratio and
poloxamer to improve the aqueous solubility and bioavailability of the
3.3. Characterization of CBD-β-CDPM
poorly soluble CBD. The CBD-β-CDPM solution was freeze-dried into
powder and reconstituted as an active ingredient in a nasal spray
A previously published thermal analysis reported that poloxamer

8
N. Changsan et al. International Journal of Pharmaceutics 640 (2023) 123035

Fig. 5. In vitro results of cytokine levels: (A) TNF-α, (B) IL-1β, and (C) IL-6 produced from macrophage-like cells (RAW264.7) after exposure to S-RBD, culture
medium (negative control), CBD-β-CDPM-NS, S-RBD following with CBD-β-CDPM-NS 24 h later (treatment), and co-administration of S-RBD and CBD-β-CDPM-NS.
Data are expressed as mean ± SD (n = 3).

407 had the lowest melting point of 59.49 ◦ C (Emam et al., 2021) fol­ absence of the poloxamer 407 peak. This indicated that the three mol­
lowed by CBD at 67.5 ◦ C (Stinchcomb et al., 2004), whereas β-CD melted ecules could interact even in the physical mixture during the heating
at 267–269 ◦ C (Thurein et al., 2018). However, the broad endothermic stage, and that the lowest melting components of poloxamer 407 and
profile of β-CD showed a peak at around 100–150 ◦ C, which corresponds CBD could act as a cosolvent system while forming a complex structure
to the loss of crystalized water molecules from the β-CD cavity. Several of those three components.
other researchers reported results such as a centered peak at 145 ◦ C When the CBD complexed with β-CD (CBD-β-CD), it was found that
(Marangoci et al., 2019), a range of 34–155 ◦ C (peak ~ 125 ◦ C) (Júnior part of the CBD structure was possibly inserted into the CD cavity. The
et al., 2019), and a peak at 113.2 ◦ C (David et al., 2019). In this study, small CBD peak at 60 ◦ C was found to have a much smaller peak area
the thermograms of the raw materials of pure compound poloxamer together with an amorphous halo from 100 to 120 ◦ C that was possibly
407, CBD, and β-CD had melting peaks at 53.9, 64.2, and 122.9 ◦ C, caused from free CBD that did not form the inclusion complex with β-CD
respectively. The thermogram of the physical mixture showed an (Fig. S2).
absence of the characteristic melting peaks of poloxamer 407 and β-CD The DSC thermogram showed that the CBD-β-CDPM is an amorphous
(Supplementary Fig. S2). The melting point of CBD shifted to a lower solid, while the DSC thermogram did not show the response peak of any
melting point temperature of 60 ◦ C. This indicated that the physical compounds. Hence the mixture was completely amorphous. It can be
mixture formed a complex with β-CD during the heating phase that postulated that the mixture is readily soluble in water without free CBD
resulted in a much lower melting point than the original melting point of remaining (Fig. S2). When CBD and poloxamer 407 were prepared as a
pure β-CD caused by eutectic phenomena. Also, the melting point of CBD mixture, there was no crystallinity observed in the thermogram, which
shifted to a lower value in the mixture of the three components with an indicated only an amorphous phase in the mixture.

9
N. Changsan et al. International Journal of Pharmaceutics 640 (2023) 123035

Fig. 6. Ex vivo results of cytokine levels: (A) TNF-α, (B) IL-1β, and (C) IL-6 produced from porcine nasal mucosa explant after exposure to S-RBD, culture medium
(negative control), CBD-β-CDPM-NS, S-RBD 6 h following with CBD-β-CDPM-NS for 30 min (treatment), and CBD-β-CDPM-NS for 30 min following with S-RBD for 6 h
(prevention). Data are expressed as mean ± SD (n = 3).

The FT-IR spectra of CBD, poloxamer 407, β-CD, CBD-β-CD and CBD- 2001). The characteristic IR bands of β-CD and CBD were found in the
β-CDPM are shown in Supplementary Fig. S3. The potential interactions complex of CBD-β-CD and peak intensity decreased compared to raw
between CBD and β-CD were investigated by FT-IR analysis by compounds, which confirmed that molecular interaction between CBD
comparing the respective absorption spectra, considering specific func­ and β-CD occurred.
tional groups of the CBD, poloxamer 407 and β-CD alone, and complexes From the FT-IR results, CBD, β-CD and poloxamer did not chemically
of CBD-β-CD. The potential interactions of poloxamer 407 in the com­ interact with one another as all fingerprints of each component appeared
plexes system were also investigated. If CBD forms the inclusion com­ in the spectra. Possible structures of CBD-β-CDPM could be a mixture of:
plexes with β-CD, the characteristic peaks of CBD probably shift, (A) the CBD inclusion complex with β-CD at a ratio of 1:1; (B) CBD
decrease, or disappear (Li et al., 2021). The characteristic bands at without complex encapsulation in the core of poloxamer; (C) CBD-β-CD
2925–2856 cm− 1 (C–H stretching) and at ~ 3408 cm− 1 (O–H stretching) complex may encapsulate in the core of the poloxamer but might be
were found in CBD and β-CD alone and also in the complex of CBC-β-CD difficult to occur due to hydrophilicity molecules; and (D) CBD-β-CD
and CBD-β-CDPM. The CBD spectrum showed characteristic bands at complex inserted between hydrophilic parts of poloxamer micelles
1623–1583 cm− 1 (cyclic alkene C = C stretching) that were not found in (Supplementary Fig. S4).
the β-CD and poloxamer 407. Only C–O stretching of aliphatic ether was
found in β-CD at the wave number 1158 cm− 1 and also in the poloxamer
407 that usually occurs in the range 1260–1000 cm− 1 (Pavia et al.,

10
N. Changsan et al. International Journal of Pharmaceutics 640 (2023) 123035

Fig. 7. Schematic diagram of CBD-β-CDPM-NS as a potential agent to reduce cytokine storm caused by SARS-CoV-2 infection.

3.4. Development and stability of CBD-β-CDPM-NS equilibrium moisture during storage.


The particle size of CBD-β-CDPM-NS initially was 111.9 ± 0.7 d.nm
CBD-β-CDPM was used as an active ingredient along with a buffering and the zeta potential was 0.8 ± 0.1 mV. The particle size of CBD-
agent, preservative, and ethanol as a permeation enhancer (Bhise et al., β-CDPM-NS after storage at room temperature for six months increased
2008) to formulate a nasal spray solution. The formulation for CBD- to 121.4 ± 1.1 d.nm with a small change in the surface charge. The CBD-
β-CDPM nasal spray (CBD-β-CDPM-NS) was a clear solution. β-CD complex polymer micelle enlargement indicated some degree of
HEC (0.05 %w/w) was used as a mucoadhesive polymer in this instability although no precipitation or aggregation occurred, and the
experiment due to its adhesiveness to the nasal epithelial cells (Cho surface charge increased to nearly zero. This result was possibly caused
et al., 2021). In addition to the good mucoadhesive properties, Hansen by the buffering agent and preservative in the nasal spray formulation
et al. reported that HEC enhanced the transport of drug molecules across that affected the counter ions of the colloidal system. In addition, the
the nasal epithelium (Hansen et al., 2015). A preliminary study of HEC environmental change of the colloidal system may lead to slightly larger
concentration at varying concentrations of 0.05, 0.1, 0.15, 0.2, and sizes and structures of the micelles (Perumal et al., 2022).
0.25% w/w had no effect on surface tension but had a significant impact The osmolarity of this formulation was 300 ± 0.8 mOsm/kg in the
on viscosity. The viscosity of HEC ranged from 6.2 cP at 0.05% (w/v) to range of 285–310, which indicated an isotonic solution suitable for nasal
82.1 cP at 0.25% (w/v) (Supplementary Fig. S5). On the basis of these preparation (USP43; Ehrick et al., 2013; Barros et al., 2022). The vis­
results, the lowest concentration of HEC with the lowest viscosity was cosity of the nasal spray formulation was 12.04 ± 2.64 cP at the shear
chosen for the formulation for easy release from the container. rate of 50 s− 1. The rheogram of the CBD-β-CDPM-NS showed a shear
The average size of the CBD-β-CDPM before freeze-drying was 101.4 thinning system, which was typical of pseudoplastic behavior (Supple­
± 6.9 d.nm and its polydispersity index was 0.249 ± 0.013 (n = 6) mentary Fig. S6). As the shear rate increased, the viscosity decreased.
(Table 3). The literature reported that polymeric micelles of pure The local pH value inside the nasal cavity directly influences the rate and
poloxamer 407 showed a particle size under 100 nm (Bahman et al., extent of drug absorption. It has been suggested that the optimum pH
2019). The particle size of CBD-β-CDPM was larger than empty polox­ value of nasal spray should range from 4.5 to 6.5 (Dhakar et al., 2011) or
amer micelles, which was likely due to the insertion of CBD or CBD-β-CD 5.5 to 6.5, which is close to the physiological pH (Barros et al., 2022).
in the core of the micelles. The apparent zeta potential of the CBD- The pH value of the nasal spray formulation in this current study was
β-CDPM before freeze-drying was − 8.9 ± 0.4 mV. This result indicated 6.02 ± 0.02, which is suitable for nasal epithelial cells. The surface
the micelles were negatively charged, which was similar to the results of tension was 34.5 mN/m, which was lower than sterile water (72.8 mN/
− 13.57 mV in a previous report on polymeric micelles of gambogic acid m at 20 ◦ C) (Sinko, 2011). This result indicated that poloxamer 407
(Saxena and Hussain, 2012). Both the polypropylene oxide and poly­ reduced the surface tension and formed complex polymeric micelles in
ethylene oxide segments in poloxamer 407 are nonionic; therefore, the the bulk solution.
addition of CBD and β-CD affected the surface charge of the polymeric The assay results of CBD are shown in Table 3. The CBD content in
micelles. the CBD-β-CDPM was 102.1 ± 0.5% of the theoretical value (found 8.06
After storage of CBD-β-CDPM for six months, reconstitution of the g in the theoretical CBD content of 7.9 g in a total solid content of 46.2
powder with sterile water for injection resulted in a particle size of CBD- g). When CBD-β-CDPM powder was formulated as a nasal spray solution
β-CDPM of 102.5 ± 7.3 d.nm and a zeta potential of − 10.5 ± 0.2 mV. (CBD-β-CDPM-NS), the assay content was 99.8 ± 1.3% of the theoretical
The size of the CBD-β-CDPM was reduced with a lower negative charge. content.
This was possibly due to stabilization of the size and charge by The chemical and physical properties, which included CBD content,

11
N. Changsan et al. International Journal of Pharmaceutics 640 (2023) 123035

appearance, pH, osmolality, particle size, and zeta potential, of the CBD- capacity of the CBD-β-CDPM-NS formulation to significantly lower the
β-CDPM powder and CBD-β-CDPM-NS did not significantly change (p- levels of TNF-α, IL-1β, and IL-6. However, it is apparent from comparing
value < 0.5). These results confirmed that the product was stable in the treatment mode and co-administration mode that the co-
storage up to six months at room temperature. administration mode demonstrated significantly greater efficacy than
the treatment mode. All measured cytokine levels generated when S-
3.5. CBD release profile from CBD-β-CDPM-NS and permeation through RBD and CBD-β-CDPM-NS formulation were administered together were
nasal epithelium extremely low and similar to the untreated control levels as depicted in
Fig. 5, A–C. These findings are in agreement with our previous reports
The in vitro CBD release from polymeric micelles of CBD-β-CDPM-NS (Srichana et al., 2022), which showed that co-administration of a CBD
in simulated nasal cavity conditions through the dialysis membrane is formulation and allergens had the greatest impact on reducing the level
shown in Fig. 2. CBD release from CBD-β-CDPM-NS was 104 ± 0.26%, of macrophage-produced cytokines. CBD binding to the CB2 receptor on
while CBD release from CBD-β-CDPM was 91.94 ± 0.15% in 15 min. The macrophages/monocyte resulted in a decrease in cytokine production in
CBD in the nasal spray (CBD-β-CDPM-NS) was 100% released within the RAW264.7 cells exposed to S-RBD (Perwee, 2008). Co-administration of
first minute because the CBD molecules were complexed with β-CD in the CBD-β-CDPM-NS formulation and S-RBD would also function
the form of micelle and colloidal systems that were easily released from through additional mechanisms in addition to the CBD-CB2 interaction
the complexed state. In contrast to the CBD-β-CDPM powder, CBD to lessen the cytokine release induced by S-RBD. This was suggested to
release was slower than CBD-β-CDPM-NS. be the result of competing interactions between S-RBD and CBD with
The CBD permeation rate and amount of CBD per area of the mem­ macrophages/monocytes; however, CBD interactions with macro­
brane from CBD-β-CDPM-NS through the porcine nasal mucosa are phages/monocytes did not lead the immune cells to release response
shown in Fig. 3 (A) and (B). The steady-state flux (Jss) of CBD over cytokines like S-RBD did. In consequence of this, macrophage/monocyte
excised porcine nasal mucosa was 4.8 μg/cm2/min, and the rapid ab­ production of cytokines decreased when CBD was used instead of S-RBD
sorption of CBD from CBD-β-CDPM-NS increased after CBD-β-CDPM-NS in the interaction between macrophages/monocytes. According to the
was administered to the diffusion cells. After 15 min the CBD permeated findings, the combined effects of CBD-CB2 interaction and CBD
the receptor cells at a lower permeation rate. The results showed that the competitive contact with macrophages resulted in a very low cytokine
permeation rate of CBD was enhanced by increased aqueous solubility of level that was identical to the untreated control.
CBD (Ghezzi et al., 2021). However, CBD remained in the nasal
epithelium and in the donor phase. The CBD retained in the nasal 3.7. Ex vivo efficacy of CBD-β-CDPM-NS with porcine nasal mucosa
epithelial explants and the permeation across the mucosa correlated explants
with the ex vivo CBD-β-CDPM-NS as described in Section 3.6.
CBD-β-CDPM-NS efficacy was also evaluated in ex vivo porcine nasal
3.6. Cell viability and in vitro efficacy of CBD-β-CDPM-NS mucosa to demonstrate the formulation’s activity in living tissue. Since
the nasal cavity is the primary site of infection for the SARS-CoV-2 virus,
The cytotoxicity profiles of the CBD-β-CDPM, CBD-β-CDPM-NS, and it stands to reason that the nasal cavity would be the target of preven­
nasal spray blank formulation against the human nasal septum epithelial tative and therapeutic treatments. Thus, the CBD-β-CDPM-NS adminis­
cell line (RPMI 2650) and mouse monocyte/macrophage cell line tered to porcine nasal mucosa explants was investigated in both
(RAW264.7) are presented in Fig. 4A and Fig. 4B, respectively. CBD- preventive and therapeutic models. In the preventative model, CBD-
β-CDPM and CBD-β-CDPM-NS with CBD concentrations equivalent to β-CDPM-NS was administered 30 min before S-RBD protein exposure. In
3.125–50 μg/mL were used to challenge the cell line. The results showed the treatment model, explants were exposed to S-RBD for 6 h to induce
that all concentrations of CBD-β-CDPM, CBD-β-CDPM-NS, and the nasal an immunological response, followed by treatment with CBD-β-CDPM-
spray blank formulation were safe for both RPMI 2650 and RAW264.7 NS to reduce cytokine production. Co-administration of CBD-β-CDPM-
cells with nearly 100% viability. NS and S-RBD was omitted from the experiments because it does not
The SARS-CoV-2 virus’s recombinant S-RBD is a viral protein that reflect the actual use of CBD-β-CDPM-NS.
binds to the host cell’s ACE2 receptor and enters the cell, causing Ex vivo effectiveness in reducing cytokine production induced by S-
pathological changes in lung injury and inflammatory responses that RBD correlated with the in vitro cell culture model (Section 3.6). In terms
lead to excessive production of pro-inflammatory cytokines, inflamma­ of TNF-α, IL-1β, and IL-6 levels (Fig. 6) CBD-β-CDPM-NS did not cau­
tion, and tissue injury (Montazersaheb et al., 2022). These immune re­ se explant tissue to produce all the tested cytokines because the cytokine
actions might indicate the onset of a cytokine storm with severe levels found in the CBD-β-CDPM-NS exposed tissue were not signifi­
clinicopathological consequences and severe COVID-19 disease (Mah­ cantly different from the negative control tissue (p-value > 0.05). The
mudpour et al., 2020). Stimulation of RAW264.7 macrophage-like cells tissue stimulated by S-RBD produced a significant quantity of response
by S-RBD as a COVID-19 surrogate was employed to determine the ef­ cytokines more than the negative control (p-value < 0.05). CBD-
ficacy of CBD-β-CDPM-NS formulation to inhibit excessive pro- β-CDPM-NS administration both before (prevention) and after (treat­
inflammatory cytokines, such as TNF-α, IL-1β, and IL-6. Stimulation ment) S-RBD exposure significantly decreased the cytokine level relative
resulted in the release of extraordinarily high levels of TNF-α, IL-1β, and to the S-RBD exposure control tissue. The prevention condition showed
IL-6, which were 539.18 ± 5.22 pg/mL (Fig. 5A), 273.38 ± 4.61 pg/mL TNF-α values decreased from 44.59 ± 8.17 pg/g to 11.37 ± 6.34 pg/g,
(Fig. 5B), and 1666.36 ± 5.96 pg/mL (Fig. 5C), respectively. The CBD- IL-1β values decreased from 610.45 ± 37.65 pg/g to 405.05 ± 48.83 pg/
β-CDPM-NS formulation, on the other hand, did not trigger RAW264.7 g, and IL-6 values decreased from 1655.30 ± 53.21 pg/g to 747.97 ±
cells to release any pro-inflammatory cytokines because the levels 143.60 pg/g, p-value < 0.05). In addition, the treatment condition can
detected were very low and equivalent to the levels detected in the decrease the cytokine to 15.38 ± 2.22 pg/g, 418.78 ± 53.78 pg/g and
untreated control group (3rd column in Fig. 5 A–C). This indicated that 843.34 ± 206.42 pg/g of TNF-α, IL-1β, and IL-6 respectively, p-value <
the CBD-β-CDPM-NS formulation did not cause injury to the cells. 0.05). There were no significant differences in the reduction of produced
The capacity of the CBD-β-CDPM-NS formulation to suppress the cytokines from explant tissue exposed to S-RBD between the prevention
production of pro-inflammatory cytokines as a result of S-RBD stimu­ and treatment models (p-value > 0.05). This demonstrated that CBD-
lation was evaluated when CBD-β-CDPM-NS formulation was applied β-CDPM-NS was effective in alleviating the effect of S-RBD in stimu­
after S-RBD stimulation (treatment mode) or concurrently with S-RBD lating living tissue to produce response cytokines in both prevention and
stimulation (co-administration mode). Upon comparison with the S-RBD treatment models, which thereby relieved the viral-induced inflamma­
stimulation control group, all experiment modes demonstrated the tory reaction.

12
N. Changsan et al. International Journal of Pharmaceutics 640 (2023) 123035

Furthermore, β-CD has been demonstrated to have antiviral activities Innovation, Thailand for providing the spike protein of SARS-CoV-2, as
owing to its high cholesterol sequestering capacity. The depletion of well as Ms. Titpawan Nakpheng for technical support. The authors
cholesterol and destruction of lipid rafts, the virus’s cholesterol-rich would like to thank the veterinary staff of Charoen Pokphand, Foods
membrane regions, occurs as a result of structural deformation of the Public Company, Ltd, Thailand for nasal tissue preparation.
viral envelope (Braga, 2019). In addition, Lu et al. (2008) reported that
lipid rafts are crucial for SARS-CoV entry into cells (Lu et al., 2008). Appendix A. Supplementary data
Cyclodextrins can also reduce the amount of cholesterol in host cell
membranes and make them less susceptible to viral infection (Braga, Supplementary data to this article can be found online at https://doi.
2019). org/10.1016/j.ijpharm.2023.123035.
Furthermore, a nasal spray with a polysaccharide base, such as HEC,
can establish a physical barrier to limit virus-cell surface interaction by References
trapping or binding to the virus for removal by mucociliary action,
thereby preventing viral proliferation and distribution in the airways Amanat, F., Stadlbauer, D., Strohmeier, S., Nguyen, T.H.O., Chromikova, V., McMahon,
M., Jiang, K., Arunkumar, G.A., Jurczyszak, D., Polanco, J., Bermudez- Gonzalez, M.,
(Fais et al., 2022; Moakes et al., 2021). Kleiner, G., Aydillo, T., Miorin, L., Fierer, D.S., Lugo, L.A., E.M. Kojic, E.M., Stoever,
In summary, CBD, β-CD, and poloxamer were successfully formu­ J., Liu, S.T.H., Cunningham-Rundles, C., Felgner, P.L., Moran, T., García- Sastre, A.,
lated as water-soluble CBD-β-CD complexed-poloxamer micelles in a Caplivski, D., Cheng, A.C., Kedzierska, K., Vapalahti, O., Hepojoki, J.M., Simon, V.,
Krammer, F., 2020. A serological assay to detect SARS-CoV-2 seroconversion in
molar ratio of approximately 1:1:0.03 as proposed in Fig. 7 and can be humans, Nat. Med. 26(7), 1033–1036. https://doi.org/10.1038/s41591-020-0913-
used as an active ingredient in developing nasal spray formulations. 5.
Anil, S.M., Shalev, N., Vinayaka, A.C., Nadarajan, S., Namdar, D., Belausov, E., Shoval, I.,
Mani, K.A., Mechrez, G., Koltai, H., 2021. Cannabis compounds exhibit anti-
4. Conclusions inflammatory activity in vitro in COVID-19-related inflammation in lung epithelial
cells and pro-inflammatory activity in macrophages. Sci Rep. 11 (1), 1462. https://
The CBD-β-CD complex-polymeric micelle nasal spray solution (CBD- doi.org/10.1038/s41598-021-81049-2.
Asean guideline on stability of drug product (R1), Available online at https://asean.
β-CDPM-NS) showed physicochemical characteristics suitable for
org/wp-content/uploads/2018/01/25PPWG-ANNEX-7-iv-Final-ASEAN-Guideline-
application as a nasal spray. CBD-β-CDPM-NS demonstrated its efficacy on-Stability-Study-Drug-Product-R2.pdf. Access on 26 September 2022.
and safety due to very low cytokine production from nasal immune cells Bahman, F., Elkaissi, S., Greish, K., Taurin, S., Chapter 8 Polymeric micelles in
and very low toxicity to nasal epithelial cells. The formulations were management of lung cancer. Kesharwani, P. (editor), Nanotechnology-Based
Targeted Drug Delivery Systems for Lung Cancer. Academic Press, USA. 2019. page
successful in reducing COVID-19/SARS-CoV-2-related inflammatory 193-216.
reactions both in vitro and ex vivo. The findings of this study indicate that Barros, C., Portugal, I., Batain, F., Portella, D., Severino, P., Cardoso, J., Arcuri, P.,
CBD-β-CDPM-NS strongly inhibits the immunological response of Chaud, M., Alves, T., 2022. Formulation, design and strategies for efficient
nanotechnology-based nasal delivery systems. RPS Pharmacy and Pharmacology
macrophage/monocytes induced by the SARS-CoV-2 spike protein RBD, Reports. 1, 1–23. https://doi.org/10.1093/rpsppr/rqac003.
which may eventually lead to asymptomatic infection or symptom relief Bhise, S.B., Yadev, A.V., Avachat, A.M., Malayandi, R., 2008. Bioavailability of intranasal
once infection has commenced. The developed CBD-β-CDPM-NS may be drug delivery system. Asian J. Pharm. 201–215. https://doi.org/10.22377/ajp.
v2i4.203.
recommended as an early-stage prophylactic in SARS-CoV-2 infection Bradford, M.M., 1976. A rapid and sensitive method for the quantitation of microgram
since it may limit viral proliferation and viral loads in the nasal cavity. quantities of protein utilizing the principle of protein-dye binding. Anal. Biochem.
However, for an accurate conclusion, animal studies should be 72 (1–2), 248–254. https://doi.org/10.1006/abio.1976.9999.
Braga, S.S., 2019. Cyclodextrins: Emerging medicines of the new millennium.
performed. Biomolecules. 9 (12), 801. https://doi.org/10.3390/biom9120801.
Carneiro, S.B., Duarte, F.Í.C., Heimfarth, L., Quintans, J.S.S., Quintans-Júnior, L.J.,
CRediT authorship contribution statement Júnior, V.F., Lima, Á.A.N., 2019. Cyclodextrin-drug inclusion complexes: in vivo and
in vitro approaches. Int. J. Mol. Sci. 20, 642. https://doi.org/10.3390/
ijms20030642.
Narumon Changsan: Methodology, Investigation, Writing – orig­ Cayman Chemical, Canabidiol, Product Information, 2015. Available online at: https://
inal draft, Writing – review & editing, Visualization. Somchai Sawat­ www.caymanchem.com/pdfs/90081.pdf.
Chatjigakis, A.K., Donzé, C., Coleman, A.W., 1992. Solubility behavior of β-cyclodextrin
dee: Methodology, Investigation, Writing – original draft, Writing –
in water/cosolvent mixtures. Anal. Chem. 64, 1632–1634. https://doi.org/10.1021/
review & editing, Visualization. Roongnapa Suedee: Conceptualiza­ ac00038a022.
tion, Writing – original draft, Writing – review & editing. Charisopon Cho, C., Hwang, S., Gu, M., Kim, C., Kim, S., Ju, D., Yun, C., Kim, H., 2021. Mucosal
Chunhachaichana: Methodology, Investigation. Teerapol Srichana: vaccine delivery using mucoadhesive polymer particulate systems. Tissue Eng.
Regen. Med. 18 (5), 693–712. https://doi.org/10.1007/s13770-021-00373-w.
Conceptualization, Methodology, Writing – original draft, Writing – David, I., Orboi, M.D., Simandi, M.D., Chirilă, C.A., Megyesi, C.I., Rădulescu, L.,
review & editing, Project administration, Funding acquisition. Drăghia, L.P., Lukinich-Gruia, A.T., Muntean, C., Hădărugă, D.I., Hădărugă, N.G.,
2019. Fatty acid profile of Romanian’s common bean (Phaseolus vulgaris L.) lipid
fractions and their complexation ability by β-cyclodextrin. Plos One. 14, e0225474.
Declaration of Competing Interest Dhakar, R.C., Maurya, S.D., Tilak, V.K., Gupta, A.K., 2011. A review on factors affecting
the design of nasal drug delivery system. Int. J. Drug Deliv. 3, 194–208. https://doi.
The authors declare that they have no known competing financial org/10.5138/ijdd.v3i2.214.
Dumortier, G., Grossiord, J.L., Agnely, F., Chaumeil, J.C., 2006. A review of poloxamer
interests or personal relationships that could have appeared to influence 407 pharmaceutical and pharmacological characteristics. Pharm. Res. 23 (12),
the work reported in this paper. 2709–2728. https://doi.org/10.1007/s11095-006-9104-4.
Ehrick, J.D., Shah, S.A., Shaw, C., Kulkarni, V.S., Coowanitwong, I., De, S., Suman, J.D.,
2013. Chapter 5 Considerations for the development of nasal dosage forms. In:
Data availability Kolhe, P., Shah, M., Rathore, N. (Eds.), Sterile Product Development, vol 6. Springer,
New York, NY. USA.
Data will be made available on request. Emam, M.F., Taha, N.F., Emara, L.H., 2021. A novel combination of Soluplus® and
poloxamer for meloxicam solid dispersions via hot melt extrusion for rapid onset of
action— part 1: dissolution and stability studies. J. Appl. Pharm. Sci. 11 (02),
Acknowledgements 141–150. https://doi.org/10.7324/JAPS.2021.110218.
Esposito, G., Pesce, M., Seguella, L., Sanseverino, W., Lu, J., Corpetti, C., Sarnelli, G.,
This research was supported by the National Science, Research and 2020. The potential of cannabidiol in the COVID-19 pandemic. Br. J. Pharmacol. 177
(21), 4967–4970. https://doi.org/10.1111/bph.15157.
Innovation Fund and Prince of Songkla University (PHA 64050875). The European Medicines Agency, Background review for cyclodextrins used as excipients.
authors would like to thank the Drug Delivery System Excellence Center, 2014. Available online at: https://www.ema.europa.eu/en/documents/report/ba
Faculty of Pharmaceutical Sciences, Prince of Songkla University for ckground-review-cyclodextrins-used-excipients-context-revision-guideline-excipie
nts-label-package_en.pdf. Accessed on 3 Oct. 2022.
providing the facilities and the National Biotechnology Center (BIO­
TEC), NSTDA, Ministry of Higher Education Science Research and

13
N. Changsan et al. International Journal of Pharmaceutics 640 (2023) 123035

Fais, F., Juskeviciene, R., Francardo, V., Mateos, S., Guyard, M., Viollet, C., Constant, S., SARS-COV-2. Adv. Mater. 33 (26), 2008304. https://doi.org/10.1002/
Borelli, M., Hohenfeld, I.P., 2022. Drug-free nasal spray as a barrier against SARS- adma.202008304.
CoV-2 and its delta variant: in vitro study of safety and efficacy in human nasal Montazersaheb, S., Khatibi, S.M.H., Hejazi, M.S., Tarhriz, V., Farjami, A., Sorbeni, F.G.,
airway epithelia. Int. J. Mol. Sci. 23 (7), 4062. https://doi.org/10.3390/ Raheleh Farahzadi, R., Ghasemnejad, T., 2022. COVID-19 infection: an overview on
ijms23074062. cytokine storm and related interventions. Virol 19, 92. https://doi.org/10.1186/
Fenyvesi, F., Nguyen, T.L.P., Haimhoffer, Á., Rusznyák, Á., Vasvári, G., Bácskay, I., s12985-022-01814-1.
Vecsernyés, M., Ignat, S., Dinescu, S., Costache, M., Ciceu, A., Hermenean, A., Nagy, N.Z., Varga, Z., Mihály, J., Domján, A., Fenyvesi, É., Kiss, É., 2020. Highly
Váradi, J., 2020. Cyclodextrin complexation improves the solubility and caco-2 enhanced curcumin delivery applying association type nanostructures of block
permeability of chrysin. Material. 13, 3618. https://doi.org/10.3390/ma13163618. copolymers, cyclodextrins and polycyclodextrins. Polymers. 12, 2167. https://doi.
Ghezzi, M., Pescina, S., Padula, C., Santi, P., Favero, E.D., Cantù, L., Nicoli, S., 2021. org/10.3390/polym12092167.
Polymeric micelles in drug delivery: An insight of the techniques for their Nivorozhkin, A., 2019. Solubilization of phytocannabinoids using cyclodextrins.
characterization and assessment in biorelevant conditions. J. Control. Release. 332 Cannabis Science and Technology. 2 (4), 58–64.
(312–336), 6. https://doi.org/10.1016/j.jconrel.2021.02.031. Păduraru, D.N., Niculescu, A., Bolocan, A., Andronic, O., Grumezescu, A.M., Bîrlă, R.,
Hădărugă, N., Bandur, G.N., David, I., Hădărugă, D.I., 2019. A review on thermal 2022. An updated overview of cyclodextrin-based drug delivery systems for cancer
analyses of cyclodextrins and cyclodextrin complexes. Environ. Chem. Lett. 17, therapy. Pharmaceutics. 14, 1748. https://doi.org/10.3390/
349–373. https://doi.org/10.1007/s10311-018-0806-8. pharmaceutics14081748.
Haimhoffer, Á., Rusznyák, Á., Réti-Nagy, K., Vasvári, G., Váradi, J., Vecsernyés, M., Paudel, K.S., Hammell, D.C., Agu, R.U., Valiveti, S., Stinchcomb, A.L., 2010. Cannabidiol
Bácskay, I., Fehér, P., Ujhelyi, Z., Fenyvesi, F., 2019. Cyclodextrins in drug delivery bioavailability after nasal and transdermal application: effect of permeation
systems and their effects on biological barriers. Sci. Pharm. 87, 33. https://doi.org/ enhancers. Drug Dev. Ind. Pharm. 39 (9), 1088–1097. https://doi.org/10.3109/
10.3390/scipharm87040033. 03639041003657295.
Hansen, K., Kim, G., Desai, K.H., Patel, H., Olsen, K.F., Curtis-Fisk, J., Tocce, E., Pavia, D.L., Lampman, G.M., Kriz, G.S. 2001. Chapter 2 Infrared Spectroscopy in
Jordan, S., Schwendeman, S.P., 2015. Feasibility investigation of cellulose polymers Introduction to Spectroscopy. A guide for students of organic chemistry. 3rd edition,
for mucoadhesive nasal drug delivery applications. Mol. Pharmaceutics. 12 (8), Thomson Learning, Inc., USA. Page 45.
2732. https://doi.org/10.1021/acs.molpharmaceut.5b00264. Pereira, A.G., Carpena, M., Oliveira, P.G., Mejuto, J.C., Prieto, M.A., Gandara, J.S., 2021.
Hatziagapiou, K., Bethanis, K., Koniari, E., Christoforides, E., Nikola, O., Andreou, A., Main applications of cyclodextrins in the food industry as the compounds of choice
Mantzou, A., Chrousos, G.P., Kanaka-Gantenbein, C., Lambrou, G.I., 2022. to form host–guest complexes. Int. J. Mol. Sci. 22, 1339. https://doi.org/10.3390/
Biophysical studies and in vitro effects of tumor cell lines of cannabidiol and its ijms22031339.
cyclodextrin inclusion complexes. Pharmaceutics. 14, 706. https://doi.org/10.3390/ Perumal, S., Atchudan, R., Lee, W., 2022. A review of polymeric micelles and their
pharmaceutics14040706. applications. Polymers. 14, 2510. https://doi.org/10.3390/polym14122510.
Holst, M., Nowak, D., Hoch, E., 2022. Cannabidiol as a treatment for COVID-19 Perwee, R.G., 2008. The diverse CB1 and CB2 receptor pharmacology of three plant
symptoms? A critical review. Cannabis and Cannabinoid Research. online first. cannabinoids: Δ9-tetrahydrocannabinol, cannabidiol and Δ9-
https://doi.org/10.1089/can.2021.0135. tetrahydrocannabivarin. Br. J. Pharmacol. 153, 199–215. https://doi.org/10.1038/
Inoure, D., Yamashita, A., To, H., 2022. Development of in vitro evaluation system for sj.bjp.0707442.
assessing drug dissolution considering physiological environment in nasal cavity. Pires, P., Rodrigues, M., Alves, G., Santos, A.O., 2022. Strategies to improve drug
Pharmaceutics. 14, 2350. https://doi.org/10.3390/pharmaceutics14112350. strength in nasal preparations for brain delivery of low aqueous solubility drugs.
Janecki, M., Graczyk, M., Lewandowska, A., Pawlak, Ł., 2022. Anti-inflammatory and Pharmaceutics. 14, 588. https://doi.org/10.3390/pharmaceutics14030588.
antiviral effects of cannabinoids in inhibiting and preventing SARS-CoV-2 infection. Polidoro, D., Temmerman, R., Devreese, M., Charalambous, M., Ham, L.V., Cornelis, I.,
Int. J. Mol. Sci. 23, 4170. https://doi.org/10.3390/ijms23084170. Broeckx, B.J.G., Mandigers, P.J.J., Fischer, A., Storch, J., Bhatti, S.F.M., 2022.
Jones, P.G., Falvello, L., Kennard, O., Sheldrick, G.M., 1977. Cannabidiol. Acta Cryst. Pharmacokinetics of cannabidiol following intranasal, intrarectal, and oral
B33, 3211–3214. https://doi.org/10.1107/S0567740877010577. administration in healthy dogs. Front. Vet. Sci. 9, 899940 https://doi.org/10.3389/
Júnior, W.F.S., Menezes, D.L.B., Oliveira, L.C., Koester, L.S., Almeida, P.D.O., Lima, E.S., fvets.2022.899940.
Azevedo, E.P., Júnior, V.F.V., Lima, Á.A.N., 2019. Inclusion complexes of β and HPβ- Poulson, B.G., Alsulami, Q.A., Sharfalddin, A., Agammy, E.F.E., Mouffouk, F., Emwas, A.,
cyclodextrin with α, β amyrin and in vitro anti-inflammatory activity. Biomolecules. Jaremko, L., Jaremko, M., 2022. Cyclodextrins: structural, chemical, and physical
9, 241. https://doi.org/10.3390/biom9060241. properties, and applications. Polysacharides. 3, 1–31. https://doi.org/10.3390/
Kabanov, A., Batrakova, E.V., Alakhov, V.Y., 2002. Pluronic® block copolymers as novel polysaccharides3010001.
polymer therapeutics for drug and gene delivery. J. Control. Release. 82, 189–212. PreveCeutical Medical Inc. Available online: https://www.preveceutical.com/ pipeline/c
https://doi.org/10.1016/s0168-3659(02)00009-3. annabinoid-sol-gel/.
Koch, N., Jennotte, O., Gasparrini, Y., Vandenbroucke, F., Lechanteur, A., Evrard, B., Ramalho, Í.M.M., Pereira, D.T., Galvão, G.B.L., Freire, D.T., Amaral-Machado, L.,
2020. Cannabidiol aqueous solubility enhancement: comparison of three amorphous Alencar, É.N., Egito, E.S.T., 2021. Current trends on cannabidiol delivery systems:
formulations strategies using different type of polymers. Int. J. Pharm. 589, 119812 where are we and where are we going? Expert Opin. Drug Deliv. 18 (11),
https://doi.org/10.1016/j.ijpharm.2020.119812. 1577–1587. https://doi.org/10.1080/17425247.2021.1952978.
Li, H., Chang, S., Chang, T., You, Y., Wang, X., Wang, L., Yuan, X., Tan, M., Wang, P., Ramasamy, S., Subbian, S., 2019. Critical determinants of cytokine storm and type I
Xu, P., Gao, W., Zhao, Q., Zhao, B., 2021. Inclusion complexes of cannabidiol with interfereon response in COVID-19 pathogenesis. Clin. Microbiol. Rev. 34 (3),
β-cyclodextrin and its derivative: physicochemical properties, water solubility, and e00299020.
antioxidant activity. J. Mol. Liq. 334, 116070 https://doi.org/10.1016/j. Rowe, R.C., Sheskey, P.J., Quinn, M.E. (Editors). Handbook of Pharmaceutical
molliq.2021.116070. Excipients. 6th edition. Pharmaceutical Press and American Pharmacists Association,
Li, H., Zhao, Q., Wang, L., Wang, P., Zhao, B., 2022. Cannabidiol/hydroxypropyl- 2009.
β-cyclodextrin inclusion complex: structure analysis, release behavior, permeability, Saxena, V., Hussain, M.D., 2012. Poloxamer 407/TPGS mixed micelles for delivery of
and bioactivity under in vitro digestion. New J. Chem. 46, 4700. https://doi.org/ gambogic acid to breast and multidrug-resistant cancer. Int. J. Nanomedicine. 7,
10.1039/D1NJ05998J. 713–721. https://doi.org/10.2147/IJN.S28745.
Loftsson, T., Brewster, M., 2010. Pharmaceutical applications of cyclodextrins: basic Sinko, P.J. editor, Martin’s physical pharmacy and pharmaceutical sciences: physical
science and product development. J. Pharm. Pharmacol. 62, 1607–1621. https://doi. chemical and biopharmaceutical principles in the pharmaceutical sciences. 6th
org/10.1111/j.2042-7158.2010.01030.x. edition. Chapter 15 Interfacial phenomena. 2011. Lippincott Williams & Wilkins,
Lu, Y., Liu, D.X., Tam, J.P., 2008. Lipid rafts are involved in SARS-CoV entry into Vero E6 Philadelphia, USA. Page 357.
cells. Biochem. Biophys. Res. Commun. 369, 344–349. https://doi.org/10.1016/j. Srichana, T., Chunhachaichana, C., Suedee, R., Sawatdee, S., Changsan, N., 2022. Oral
bbrc.2008.02.023. inhalation of cannabidiol delivered from a metered dose inhaler to alleviate cytokine
Lv, P., Zhang, D., Guo, M., Liu, J., Chen, X., Guo, R., Xu, Y., Zhang, Q., Guo, H., Yang, M., production induced by SARS-CoV-2 and pollutants. J. Drug Deliv. Sci. Technol. 76,
2019. Structural analysis and cytotoxicity of host-guest inclusion complexes of 103805 https://doi.org/10.1016/j.jddst.2022.103805.
cannabidiol with three native cyclodextrins. J. Drug Deliv. Sci. Technol. 51, Stinchcomb, A.L., Valiveti, S., Hammell, D.C., Ramsey, D.R., 2004. Human skin
337–344. https://doi.org/10.1016/j.ddst.2019.03.015. permeation of Δ8-tetrahydrocannabinol, cannabidiol and cannabinol. J. Pharm.
Mahmudpour, M., Roozbeh, J., Keshavarz, M., Farrokhi, S., Nabipour, I., 2020. COVID- Pharmacol. 56, 291–297. https://doi.org/10.1211/0022357022791.
19 Cytokine storm: the anger of inflammation. Cytokine. 133, 155151 https://doi. Suksiriworapong, J., Rungvimolsin, T., A-gomol, A., Junyaprasert, V.B., Chantasart, D.,
org/10.1016/j.cyto.2020.155151. 2015. Development and characterization of lyophilized diazepam-loaded polymeric
Mannila, J., Järvinen, T., Järvinen, K., Jarho, P., 2007. Precipitation complexation micelles. AAPS PharmSciTech. 15, 52–64. https://doi.org/10.1208/s12249-013-
method produces cannabidiol/β-cyclodextrin inclusion complex suitable for 0032-4.
sublingual administration of cannabidiol. J. Pharm. Sci. 96 (2), 312–319. https:// Thurein, S.M., Lertsuphotvanit, N., Phaechamud, T., 2018. Physicochemical properties of
doi.org/10.1002/jps.20766. β-cyclodextrin solutions and precipitates prepared from injectable vehicles. Asian J.
Marangoci, N., Timpu, D., Corciova, A., Mircea, C., Petrovici, A., Nicolescu, A., Ursu, E., Pharm. Sci. 13, 438–449. https://doi.org/10.1016/j.ajps.2018.02.002.
Nastasa, V., Bostanaru, A., Mares, M., Pertea, M., Pinteala, M., 2019. β-Cyclodextrin Tulinski, P., Fluit, A.C., van Putten, J.P.M., Bruin, A., Glorieux, S., Wagenaar, J.A.,
as a functional excipient used for enhancing the diminazene aceturate Duim, B., 2013. An Ex Vivo Porcine Nasal Mucosa Explants Model to Study MRSA
bioavailability. Pharmaceutics. 11, 295. https://doi.org/10.3390/ Colonization. PLOS ONE 8 (1), 1–7. https://doi.org/10.1371/journal.pone.0053783.
pharmaceutics11060295. United States Pharmacopeia and National Formulary (USP 43-NF 38). United States
Mayr, T., Grassl, T., Korber, N., Christoffel, V., Bodensteiner, M., 2017. Cannabidiol Pharmacopeial Convention; 2020. <785> Osmolarity.
revisited. IUCrData. 2, x170276 https://doi.org/10.1107/S2414314617002760. Vallée, A., 2022. Cannabidiol and SARS-CoV-s infection. Front. Immunol. 13, 870787
Moakes, R.J.A., Davies, S.P., Stamataki, Z., Grover, L.M., 2021. Formulation of a https://doi.org/10.3389/fimmu.2022.870787.
Composite Nasal Spray Enabling Enhanced Surface Coverage and Prophylaxis of

14
N. Changsan et al. International Journal of Pharmaceutics 640 (2023) 123035

WO Patent WO2017/208072 A2, Nasal cannabidiol compositions, assigned to Acerus intranasally administered buffers. Int. J. Pharm. 198, 1390146. https://doi.org/
Pharmaceutical Corporation, 2017. 10.1016/s0378-5173(99)0044201.
Washington, N., Steele, R.J.C., Jackson, S.J., Bush, D., Mason, J., Gill, D.A., Pitt, K., Yong, Y., Jiguang, L., Yan, S., Qianqing, L., Xiaoming, P., Yansheng, L., Yufeng, H., 2008.
Rawlins, D.A., 2000. Determination of baseline human nasal pH and the effect of Solubility of β-cyclodextrin in different mixed solvents. Pet. Sci. 5, 263–268. https://
doi.org/10.1007/s12182-008-0044-y.

15

You might also like