Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

International Journal of Pharmaceutics 589 (2020) 119776

Contents lists available at ScienceDirect

International Journal of Pharmaceutics


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

Chitosan-coated PLGA nanoparticles for the nasal delivery of ropinirole T


hydrochloride: In vitro and ex vivo evaluation of efficacy and safety
Aikaterini-Theodora Chatzitakia, Sandra Jesusb, Christina Karavasilia, Dimitrios Andreadisc,
Dimitrios G. Fatourosa, Olga Borgesb,d,

a
Department of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece
b
Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
c
Department of Oral Medicine/Pathology, School of Dentistry, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece
d
Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal

A RT ICLE INFO ABSTRACT

Keywords: Nose-to-brain delivery is an attractive route for direct drug delivery to the central nervous system (CNS),
Nasal delivery avoiding hepatic first-pass metabolism and solving blood–brain barrier passage issues. Therefore, the aim of the
PLGA/chitosan nanoparticles present study was the development of PLGA and PLGA/chitosan (chit) nanoparticles (NPs) with mucoadhesive
Ropinirole properties, able to encapsulate ropinirole hydrochloride (RH), an anti-Parkinsonian dopaminergic agonist, and
Mucoadhesion
suitable to promote RH delivery across the nasal mucosa.
Cytotoxicity
NPs produced by nanoprecipitation showed spherical shape and a mean average size of 98.8 nm and 468.0 nm
Ex vivo permeability
(PLGA and PLGA/chit, respectively). RH loaded PLGA/chit NPs showed a complete release of the drug in si­
mulated nasal electrolyte solution (SNES) over the period of 24 h and increased the permeation of RH through
sheep nasal mucosa by 3.22-fold in comparison to PLGA NPs. None of RH loaded NPs induced hemolysis in
whole blood or the production of reactive oxygen species (ROS) in Raw 264.7 cells. On their turn, PLGA/chit NPs
decreased cell viability of Raw 264.7 cells and Peripheral Blood Mononuclear Cells (PBMCs) in a concentration-
dependent manner.
These results revealed that, particularly PLGA/chit NPs, could be a valuable carrier for the delivery of RH to
the CNS, opening a new path for Parkinson’s disease therapy.

1. Introduction and Belgamwar, 2017). Currently, the most common delivery of neu­
rotherapeutics, including for RH, is via the oral route which is asso­
Neurologic disorders such as Parkinson’s disease require direct ciated with an elimination half-life of approximately 6 h and an ex­
transport of therapeutic substances to brain in order to maximize drug tensive hepatic first-pass metabolism resulting in low bioavailability in
concentration in their action site. Parkinson’s disease is characterized the range of 36–57 % and insufficient drug levels in the CNS (Pardeshi
by neurodegeneration and loss of dopaminergic neurons in the Central and Belgamwar, 2017). Also, a great challenge in CNS delivery is the
Nervous System (CNS) and the treatment approaches include the ad­ blood brain barrier (BBB) limitation of drug penetration (Gänger and
ministration of dopamine agonists. Among them, ropinirole hydro­ Schindowski, 2018), which is controlled by the drug’s physicochemical
chloride (RH) is a non-ergoline agonist that acts on D2- and D3-re­ properties and cerebral defensive mechanisms (Pardeshi and
ceptors in the brain (Barcia et al., 2017). RH is indicated for the Belgamwar, 2013). Therefore, the nose-to-brain route for drug admin­
treatment of Parkinson’s disease, as a monotherapy or in combination istration has attracted much attention considering the plethora of
with levodopa to decrease its on–off variations, and for the manage­ benefits that nasal cavity offers. The relatively large surface area of the
ment of restless leg syndrome (Khan et al., 2010). Its precise mechanism nasal epithelium, high vascularization and direct brain targeting render
of action is still unknown, though it is believed to stimulate the post- nasal cavity as a promising route of administration for molecules tar­
synaptic D2-receptors within the caudate-putamen in the brain. RH is geting the brain (Bourganis et al., 2018). Following this route, the ad­
chemically a 4-[2-(dipropylamino)-ethyl]-1,3-dihydro-2H-indol-2-one ministered drugs avoid the first hepatic metabolism and have direct
and is characterized by high aqueous solubility with logP 2.7 (Pardeshi access to the brain through the olfactory and trigeminal pathways


Corresponding author at: Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
E-mail address: olga@ci.uc.pt (O. Borges).

https://doi.org/10.1016/j.ijpharm.2020.119776
Received 16 April 2020; Received in revised form 11 August 2020; Accepted 13 August 2020
Available online 17 August 2020
0378-5173/ © 2020 Elsevier B.V. All rights reserved.
A.-T. Chatzitaki, et al. International Journal of Pharmaceutics 589 (2020) 119776

(Bourganis et al., 2018) which are localized at the roof of nasal cavity, Table 1
offering the opportunity for direct drug transport to CNS (Pardeshi and Concentrations of the raw materials used for the production of the optimized
Belgamwar, 2013). NPs.
The suitability of nose-to-brain delivery of RH has been previously Formulations Concentrations
investigated by Pardeshi et al. (2017) who formulated a nanoplex of RH
utilizing dextran sulfate sodium that provided increased ex vivo drug PLGA (mg/ PVA (% Chitosan (% w/ RH (%
mL) w/v) v) w/v)
permeability across sheep nasal mucosa (Pardeshi and Belgamwar,
2017). Rao et al. (2017) encapsulated ropinirole in a thermo-reversible PLGA NPs 5 0.8 – –
in situ nasal gel showing an approximately 5-fold bioavailability en­ RH – PLGA NPs 5 0.8 – 0.02
hancement in the brain after in vivo intranasal administration, com­ PLGA/chit NPs 5 0.8 0.1 –
RH – PLGA/chit NPs 5 0.8 0.1 0.02
pared to the intravenously administered drug solution (Rao et al.,
2017). Polymeric NPs have been also evaluated as suitable vectors for
the intranasal delivery of RH, as they prevent drug degradation, allow
analytical grade.
sustained drug release and consequently decrease the administration
frequency (Pardeshi et al., 2013).
However, the presence of mucociliary clearance, an innate clear­ 2.2. Methods
ance mechanism that hinders the efficacy of intranasal drug delivery,
creates the necessity for novel pharmaceutical formulation approaches. 2.2.1. Nanoparticle preparation
Exploitation of mucoadhesive agents and adsorption enhancers may NPs were produced via nanoprecipitation method (Table 1). Briefly,
prolong the time of interaction between drug formulation and mucus PLGA NPs were prepared by adding 2 mL of the organic phase, con­
epithelial layer and open tight junctions, respectively (Gänger and sisting of PLGA (5 mg/mL) dissolved in acetone, dropwise in 6 mL of
Schindowski, 2018). Chitosan and its derivatives have been widely in­ 0.8 % w/v PVA aqueous solution. For PLGA/chit NPs the aqueous phase
vestigated as mucoadhesive agents to enhance the adsorption of nasally contained the same concentration of PVA and 0.1 % w/v chitosan dis­
administered hydrophilic drugs and macromolecules, hampering the solved in acetic acid 2 % v/v. The ratio between the organic and the
mucociliary clearance and increasing membrane permeability (Gänger aqueous phase was set at 1:3. The NPs remained under magnetic stir­
and Schindowski, 2018). Previous work from our group on mu­ ring for 60 min at room temperature to achieve maturation and were
coadhesive PLGA-lipid microparticles containing different concentra­ then centrifuged at 3000 g using Vivaspin 20 centrifugal concentrator
tions of trimethyl chitosan (TMC) have been previously evaluated for tubes (Mw 300 kDa Sigma-Aldrich Corporation, St. Louis, MO, USA).
the nasal administration of RH. Ex vivo results demonstrated a 2.35-fold The NPs were washed with pyrogen free water, centrifuged and finally
enhancement of RH permeability across sheep nasal mucosa, compared resuspended with an appropriate volume of water to the final con­
to the drug solution (Karavasili et al., 2016). centration required in each of the following assays (calculated from
In that context, the main objective of the present study was the weighted freeze-dried NP pellets or according to drug content, when
development of mucoadhesive polymeric NPs combining PLGA and applicable). RH loaded NPs were obtained under the same procedure as
chitosan (PLGA/chit), able to encapsulate RH and enhance RH ex vivo blank NPs with the addition of RH in the aqueous phase (0.02 % w/v)
permeability across sheep nasal mucosa, demonstrating suitability for (Karavasili et al., 2016). BSA-FITC adsorbed NPs were prepared by
RH nose-to-brain delivery. In parallel, to guarantee that the developed incubating the protein (100 μg/mL) with NPs suspension (960 μg/mL)
formulation was not only effective but also safe, several biocompat­ for 2 h, protected from light. Rhodamine-B loaded NPs were prepared
ibility studies were performed with human derived cells. by adding 10 µg/mL of Rhodamine-B to the aqueous phase during NP
preparation. The procedure was performed in the dark.
2. Materials and methods
2.3. Characterization of nanoparticles
2.1. Materials
2.3.1. Size, ζ-potential and morphological analysis
Poly(D,L-lactide-co-glycolide) acid [ester terminated lactide:glyco­ Particle size and ζ-potential analysis were performed by dynamic
lide 75:25, molecular weight (Mw): 76 kDa to 115 kDa], polyvinyl al­ light scattering (DLS) and electrophoretic light scattering (ELS), re­
cohol [Mowiol 4–88, Mw ̴ 31 kDa], 3-[4,5-dimethylthiazol-2-yl]-2,5- spectively, using Zetasizer Nano-ZS (Malvern Instruments Ltd, Malver,
diphenyl tetrazolium bromide (MTT), bovine serum albumin [BSA, United Kingdom) at 25 °C. Size and ζ-potential were determined by
96 % fraction V], myoglobin [95 %-100 %] and lysozyme [≥ 80 %], diluting nanoparticle suspensions in milli-Q water.
BSA-FITC (A9771) and lipopolysaccharide (LPS) derived from Morphological characteristics of unloaded nanoparticles suspended
Salmonella enterica serotype minnesota and Rhodamine-B were ob­ in the original medium were examined using a high-resolution
tained from Sigma-Aldrich Corporation (St.Louis, MO, USA). RH was Transmission Electron Microscope (TEM) (FEI-Tecnai G2 Spirit
kindly donated from Pharmathen SA (Marousi Attica, Greece) and Biotwin). A drop of each sample was placed in copper grids (300 mesh)
chitosan (ChitoClear™ − 95 % DD and 8 cP viscosity measured in 1 % and was dried out before visualization.
solution in 1 % acetic acid) was purchased from Primex Bio-Chemicals
AS (Avaldsnes, Norway). Micro-Bicinchoninic acid (micro-BCA) protein 2.3.2. Determination of drug loading efficiency and production yield
assay kit was purchased from Pierce Chemical Company (Rockford, IL, estimation
USA). Mouse RAW 264.7 macrophage cells (ECACC, Salisbury, UK) Accurate volumes of the blank and RH loaded NP formulations,
were maintained in Dulbecco’s modified Eagle medium (DMEM) sup­ corresponding to 0.5 mg/mL of NPs, were dissolved with 50 μL
plemented with HEPES 10 mM, 3.7 g/L sodium bicarbonate, 1 % chloroform and 3.5 mL water and stirred for 15 min. Samples were
Penicillin/Streptomycin and 10 % of inactivated fetal bovine serum centrifuged at 20800 g for 20 min, followed by drug quantification in
(FBS). Roswell Park Memorial Institute (RPMI 1640) supplemented the supernatants at 250 nm using an High Performance Liquid
with 20 mM HEPES, 2 mM L-Glutamin, 1 % Penicillin/Streptomycin Chromatography (HPLC) method (Karavasili et al., 2016). The cali­
and 10 % of FBS was used to culture PBMCs. The nuclear stain Hoechst bration curve of RH in water was linear in the concentration range of
33342, Alexa Fluor-594 WGA were purchased from Invitrogen 0.098 μg/mL to 0.78 μg/mL (R2 = 0.995). The experiments were re­
(Carlsbad, CA, USA). Pyrogen-free water was kindly given by Labesfal peated five times and results were calculated according to the following
Farma (Viseu, Portugal). All other chemicals and reagents were of equation.

2
A.-T. Chatzitaki, et al. International Journal of Pharmaceutics 589 (2020) 119776

Drug Loading Efficiency (LE%) =


weight of RH in NPs (mg)
× 100 2.8. Hemolysis assay
total weight of RH added (mg)
(1) Hemolysis assay was performed as previously described (Jesus
The production yield was determined as the percentage weight of et al., 2020). Blood collected from healthy volunteers after informed
lyophilized NPs over the total amount of RH and raw materials initially written and signed consent, was stabilized with Lithium heparin and
used to produce NPs via nanoprecipitation method. Eq. (2) was used to diluted to a final total hemoglobin concentration of 10 mg/mL. Dif­
calculate the production yield. ferent concentrations of the formulations with and without RH were
added to 100 μL of the diluted blood and 700 μL of PBS in a final vo­
weight of freeze dried NPs lume of 900 μL. The same procedure was repeated also without blood to
Yield (%) = × 100
weight of raw materials and drug (2) determine if nanoparticles interfere with the hemolysis assay. PBS and
Triton-X-100 were used as a negative (NC) and positive (PC) controls of
the assay. The samples were mixed and incubated for 3 h at 37 °C with a
2.4. In vitro release studies
mild shake every 30 min and after that period, they were centrifuged at
800 g for 15 min. Hemolysis was evaluated by mixing 100 μL of each
In vitro release studies of RH encapsulated in PLGA and PLGA/chit
supernatant with 100 of cyanmethemoglobin (CMH) reagent in to a 96-
NPs were carried out in SNES pH 5.5 at 35 °C ± 0.2 °C, using the
well plate, shaking for 2 min to 3 min and reading the absorbance at
dialysis bag diffusion method (cellulose membrane, MWCO
540 nm using a microplate reader. The percentage of hemolysis was
12.000–14.000, avg. flat width 10 mm, Orange Scientific, Belgium).
calculated using the following Eq. (3):
Dialysis bags were soaked in distilled water for 30 min under agitation
prior to being filled with 1 mL aqueous NP suspension of each for­ Abs sample Abs negative control
Hemolysis (%) = × 100
mulation, diluted to obtain 49 μg RH, and were then sealed at both Abs positive control Abs negative control
ends. Samples were immersed in glass vials filled with 20 mL pre­ (3)
warmed SNES 35 °C, sealed with parafilm and placed in a shaking water
bath. At predetermined time intervals 500 μL of sample were with­
2.9. Studies on peripheral blood mononuclear cells (PBMCs)
drawn and replaced with an equal volume of fresh and preheated re­
lease medium. Samples were analyzed for RH content in accordance to
2.9.1. PBMCs isolation
the following chromatographic conditions. RH was detected at 250 nm
Peripheral blood was kindly given by IPST,IP (Coimbra, PT) and
using a Discovery HS C18 (15 cm × 4.6 mm, 5 μm) with the mobile
was obtained from normal donors in heparinized syringes followed by
phase consisting of 0.05 M methanol:ammonium acetate buffer (pH 7)
serum depletion. According to the provider’s guidance protocol and as
80:20 (v/v) at a flow rate of 0.9 mL min−1 and injection volume of
published by Jesus et al. (2017). PBMCs were isolated on a density
90 μL. Calibration curve of RH in SNES showed good linearity in the
gradient using Lymphoprep (Axis-Shield, Dundee, Scotland). Isolated
concentration range of 0.01 μg/mL to 2.5 μg/mL (R2 = 0.998).
cells were maintained in RPMI. For cytotoxicity and uptake studies,
PBMCs were plated in well plates at a density of 5 × 106 monocytes/
2.5. Differential scanning calorimetry (DSC) studies mL.

DSC analysis were performed on the raw materials and the lyophi­
2.9.2. Cytotoxicity studies
lized NP formulations using 204 F1 Phoenix DSC apparatus (Netsch
The cytotoxicity of NPs was estimated by MTT cytotoxicity assay. In
GmBH, Germany). Six mg of raw materials and each formulation were
a 96-well plate plated with 100 µL of 5 × 106 monocytes/mL, serial
placed in aluminum pans with perforated lids and their thermal beha­
dilutions of unloaded and RH loaded NPs as well as free RH were added
vior was monitored at a heating rate of 10 °C/min from 40 °C to 260 °C
to cells. After 24 h of incubation, 20 µL of MTT (5 mg/mL in PBS,
under a nitrogen purge of 70 mL/min. Proteus ver. 5.2.1 software
pH = 7.4) was added to each well and plates were incubated for ad­
(Netsch GmBH, Selb, Germany) was used for data analysis.
ditional 4 h. After that time, plates were centrifuged at 800 g for 25 min.
Supernatants were aspirated from all plates and prewarmed DMSO was
2.6. Stability studies added to each well to solubilize formazan crystals. Optical density (OD)
was measured at 540 nm and 630 nm and cell viability was calculated
Accelerated stability studies were conducted by monitoring changes by Eq. (4):
in the particle size and ζ-potential of the NPs over a 3-month period
OD sample (540 nm) OD sample (630 nm)
during storage at 25 °C ± 2 °C and relative humidity (RH) of 60 ± 5 % Cell Viability(%) = × 100
OD control (540 nm) OD control (630 nm)
as recommended by International Conference on Harmonisation (ICH)
Q1A (R2) guidelines, 2003 (Patil and Surana, 2013). Measurements (4)
were performed periodically in a monthly-based period after dilution of The concentration of NPs that inhibits cell viability by 50 % (IC50 -
the aqueous NP suspensions in milli-Q water on a Zetasizer (Nano-ZS, half maximal inhibitory concentration) was estimated by plotting the
Malvern Panalytical, Marlvern Ltd, Malvern, United Kingdom) at 25 °C log concentration of the NP vs inhibition percentage of cell viability and
(Patil and Surana, 2013). extrapolating the value from a non-linear regression using Prism 6.0
(GraphPad Software, San Diego, CA, USA).
2.7. Mucoadhesion studies
2.9.3. Uptake studies by peripheral blood mononuclear cells (PBMCs)
The mucoadhesive properties of the NPs were investigated by 2.9.3.1. Flow cytometry analysis. To investigate NPs cellular uptake,
monitoring alterations on the surface charge properties of the NPs upon two different conditions (BSA-FITC adsorbed NPs and Rhodamine-B
incubation with mucin (Takeuchi et al., 2005). The ζ-potential of the loaded NPs) were evaluated. For the uptake studies, PBMCs were plated
NP suspension and mucin (0.5 mg/mL) in SNES was determined in­ in a 48 well-plate at a concentration of 5 × 106 monocytes/mL.
itially. Mucin solution in SNES was incubated in a 1:1 ratio (w:w) with In the next day, 40 μL of BSA-FITC adsorbed NPs or Rhodamine-B
NP suspensions at room temperature under middle agitation. After 1 h, loaded NPs (prepared as previously described) as well as free protein
samples were centrifuged at 20800 g for 30 min and supernatants were (BSA-FITC) and rhodamine-B solutions (controls) were added to the
discarded in order to remove free soluble protein. Pellets were re­ wells and incubated at 37 °C for 4 h. After incubation, the plate was
suspended in SNES and the ζ-potential of the dispersion was recorded. centrifuged at 800 g for 25 min and after the removal of supernatants,

3
A.-T. Chatzitaki, et al. International Journal of Pharmaceutics 589 (2020) 119776

300 μL cold PBS was added to each well and kept at 4 °C until the compartment of a Franz diffusion cell which was filled with 20 mL SNES
analysis in a BD FACSCalibur Flow Cytometer (BD Biosciences, Bedford, with the mucosal surface facing the donor compartment. Five hundred
MA, USA). The BSA-FITC labeled NP samples were also analyzed after μL of each formulation (corresponding to 52 μg RH) were placed on the
adding 20 μL of 0.4 % trypan blue to quench the extracellular fluores­ donor compartment under stirring at 100 rpm and 35 °C ± 0.2 °C
cence of BSA-FITC (resultant from cellular adsorption) and therefore keeping the donor compartment sealed with Parafilm®. Nasal mucosa
estimate the cellular internalization of the protein. treated only with SNES under the same conditions was used as control.
At regular time intervals 500 μL were withdrawn from the receptor and
2.9.3.2. Confocal microscopy. PBMCs were plated on glass coverslips replaced with an equal volume of fresh prewarmed SNES. Samples were
treated with Poly-D-lysine in 24 well plates at a density of centrifuged at 20800 g for 20 min and drug quantification in the su­
5 × 106 monocytes/mL and incubated at 37 °C in 5 % CO2 overnight. pernatants was performed with HPLC.
In the next day, different formulations (either BSA-FITC adsorbed NPs Steady state flux (Jss) was calculated based on the plot of drug
or Rhodamine-B loaded NPs) were incubated with cells for 4 h. permeation across nasal mucosa per unit area (μg/cm2) against time
Following that period, cells were washed 3 times with phosphate (min) and corresponds to the slope of the linear section of the obtained
buffer saline (PBS) pH 7.4 and fixed with 4 % paraformaldehyde in line. Eq. (6) was used to calculate apparent permeability coefficient
PBS for 20 min at 37 °C. After fixation, cells were washed again 3 times (cm/min) (Paap), while Eq. (7) was used to calculate the steady state
and then, cells were stained with Hoechst 33342 (1 µM) for nucleus diffusion coefficient (D).
stain and Alexa Fluor 594 wheat germ agglutinin (1 μg/mL) for Jss
membrane stain. In case of Rhodamine-B loaded NPs only Hoechst Paap =
initial concentration of drug in the donor compartment
33342 was used to stain the nucleus. After labeling for 10 min, cells
(6)
were washed twice with PBS and coverslips mounted on microscope
slides with DAKO mounting medium and examined under an inverted D= Papp·L/K (7)
laser scanning confocal microscope (Zeiss LSM 710, Carl Zeiss,
where K is the partition coefficient and L is the diffusion path length.
Oberkochen, DE) equipped with imaging software (Zen Black 2012
(Pardeshi and Belgamwar, 2018).
software, Carl Zeiss).

2.12. Histopathological examination


2.10. Studies on RAW 264.7 macrophage cell line
At the end of the ex vivo permeability studies, control tissues and
2.10.1. Cytotoxicity studies
tissues treated with the NP formulations were collected, washed prop­
Raw 264.7 (ATCCR TIB-71TM) were cultured in Dulbecco’s mod­
erly with distilled water and maintained in formalin 10 %. Tissue sec­
ified Eagle medium (DMEM) supplemented with 10 mM HEPES, 3.7 g/L
tions were stained with hematoxylin and eosin and observed under a
Sodium Bicarbonate, 1 % Penicillin/Streptomycin and 10 % of in­
light microscope (Olympus CX31 optical microscope). Images were
activated fetal bovine serum (FBS) and incubated overnight at 37 °C and
acquired using the OLYMPUS analySIS getIT software.
5 % CO2. MTT cytotoxicity assay was also performed in macrophages,
as described previously, for PBMCs with some modifications. Cells were
2.13. Statistical analysis
seeded in a 96-well plate at a density of 2 × 105 cells/well and in­
cubated with MTT solution for 1 h 30 min.
Results were expressed as mean ± standard error or mean ± SEM.
Statistical analysis was performed using one-way ANOVA and
2.10.2. Nanoparticle effect on production of the reactive oxygen species
GraphPad Prism v 5.03 (GraphPad Software Inc., La Jolla, CA, USA).
(ROS Assay)
Data obtained by flow cytometry were analyzed with CellQuest Modfit
The ROS production was evaluated using the dichlorofluorescein
LT analysis software (BD Biosciences, Bedford, MA, USA). P
diacetate probe (DCFH-DA) (Thermo Fisher Scientific Inc., Waltham,
value ≤ 0.05 was considered as statistically significant.
MA, USA) as previously described (da Silva et al., 2019). The RAW
264.7 cells were seeded in a black 96-well plate at density of 5 × 105
3. Results
cells/mL and incubated for 24 h at 37 °C and 5 % CO2. After 24 h, dif­
ferent concentrations (serial dilutions) of unloaded and RH loaded
3.1. PLGA NPs are smaller and have lower polydispersity when compared to
PLGA and PLGA/chit NPs as well as LPS as positive control and cell
PLGA/chit NPs
culture medium (negative control) were incubated with cells. After 24 h
incubation, cell culture medium was replaced by DCFHDA (50 μM) in
It has been previously established that a hydrodynamic diameter
serum free DMEM and the plates incubated for another 2 h. The re­
smaller than 200 nm leads to an easier transportation to the brain by
sulting fluorescence was measured at 485/20 nm and 528/20 nm (ex­
olfactory receptor neuron (ORN) via olfactory region (Kumar et al.,
citation/emission wavelengths).
2008). However, the presence of a cationic ligand, as chitosan in our
The following equation (Eq. (5)) was used to calculate ROS pro­
study, promotes nose-to-brain delivery via absorption-mediated trans­
duction:
cytosis through BBB for NPs with size less than 500 nm (Kumar et al.,
ROS production (mean fluorescence increase) 2013). In this work, PLGA NPs yielded a mean average size of
Fluoroscence (SAMPLE) 98.8 nm ± 10.3 nm with low PDI (0.14) and a negative surface charge
= × 100 in water (−9.28 mV ± 4.45 mV). The negative charge of PLGA nano­
Fluoroscence (NEGATIVE CONTROL) (5)
particles is attributed to the carboxyl groups of PLGA and PVA (Guo and
Gemeinhart, 2008). On the other hand, the blend of chitosan and PLGA
2.11. Ex vivo permeability studies resulted in a significant increase in particle size (468.0 nm ± 40.0 nm)
and also in the PDI (0.29). The presence of chitosan in PLGA NPs is
Ex vivo permeability studies were carried out across sheep nasal responsible for the high positive charge of the NPs, due to the amino
mucosa obtained from the local slaughterhouse according to a pre­ groups of chitosan. The highly positive surface charge (+54.4 mV ±
viously described protocol (Karavasili et al., 2016). Immediately after 2.6 mV), suggested that nanoparticle suspensions were stable and
isolation, tissues were maintained in phosphate buffer (pH 7.4) until protected from aggregate formation (Sharma et al., 2015). The en­
use. The tissues were mounted between the donor and the receptor capsulation of RH did not affect significantly the hydrodynamic

4
A.-T. Chatzitaki, et al. International Journal of Pharmaceutics 589 (2020) 119776

Fig. 1. TEM images of NPs dispersed in pyrogen-free water. A) and B) PLGA NPs; C) and D) PLGA/chit NPs.

diameter, the polydispersion and the surface charge of NPs, which


followed the same tendency as the unloaded NPs (Supplementary ma­
terial, Table S1). Particle size measurements were further corroborated
with TEM imaging of the unloaded NPs revealing their spherical shape
(Fig. 1).

3.2. Chitosan blend enhanced water permeation resulting in higher in vitro


RH release

As previously showed, PLGA and PLGA/chit NPs were successfully


produced by nanoprecipitation. The production yield achieved for the
PLGA and PLGA/chit NPs was 60.8 ± 4.0 % and 33.6 ± 4.9 %, re­
spectively. These values were slightly increased when RH was loaded
into NPs (66.0 ± 9.3 % and 45.7 ± 7.9 %, respectively). Considering
RH loading efficiency (LE), the methodology employed allow to have
5.4 ± 0.7 % LE on PLGA NPs and 5.7 ± 2.5 % on PLGA/chit NPs. Fig. 2. In vitro RH release profiles from RH loaded NPs in SNES. Data are
These low values might be attributed to the hydrophilic nature of the presented as mean ± SD and , n ≥ 3 (more than 3 independent experiments).
RH resulting in low LE during the production process (Govender et al.,
1999).
3.3. Thermal analysis
The in vitro release pattern of RH loaded NPs was evaluated in SNES
(pH 5.5) at 35 ± 0.2 °C for 24 h and results are shown in Fig. 2. RH -
DSC studies were performed for the raw materials and lyophilized
PLGA/chit NPs exhibited a biphasic release profile with a burst RH
NPs and the illustrative spectra are presented in supplementary mate­
release of 34 % in the first 5 min, probably due to the surface adsorbed
rial, Figure S1. PLGA 75:25 polymer exhibited a glass transition at
RH on NPs, followed by a sustained release during 24 h. RH loaded
54.7 °C. PVA had a glass transition temperature at 60 °C and melting
PLGA/chit NPs achieved a release of 89 % of the total drug content,
point at 193 °C. For chitosan an endothermic peak at 100 °C was ob­
whereas only 20 % of RH was released from RH - PLGA NPs. The slow
served due to polymer’s dehydration, as it has been previously de­
release of RH from PLGA NPs might be attributed to the hydrogel ob­
scribed elsewhere (Neto et al., 2005). A sharp endothermic peak at
stacle that PVA residuals form (Sahoo et al., 2002) and to the crystalline
248.1 °C was observed for pure RH. The DSC thermograms of the lyo­
nature of PVA and PLGA polymers. On the other hand, the addition of
philized formulations with and without drug presented an endothermic
the hydrophilic chitosan favors the hydration of the polymeric blend
peak in the temperature range of 50 °C to 100 °C due to the dehydration
matrix and promotes the diffusion of the drug, therefore enhancing RH
of the polymer’s (PVA) hydrophilic moieties (Azevedo et al., 2011; El-
release (Mohammed et al., 2017).
Houssiny et al., 2016) and one endothermic peak at 193 °C corre­
sponding to the melting point of PVA. On the other hand, the absence of

5
A.-T. Chatzitaki, et al. International Journal of Pharmaceutics 589 (2020) 119776

Fig. 3. Stability of unloaded and RH loaded PLGA and PLGA/chit NPs. A) Average size (left y-axis) and PDI (right y-axis) and B) ζ-potential of NP suspensions
(pyrogen-free water) after production and 3 months after storage at environmental conditions (25 ± 2 °C, 60 ± 5 % RH). Data are presented as mean ± standard
deviation (SD), n ≥ 3 (minimum of 3 independent batches).

the drug’s characteristic peak, suggests either the presence of RH in an groups of chitosan, which may favor the interpenetration of the chit­
amorphous state in the NPs or that the drug concentration was lower osan NPs and mucin. Apart from the electrostatic interactions, being the
than the limits of equipment detection (Patil and Surana, 2013). major underlying mechanism of chitosan mucoadhesion, hydrophobic
and hydrogen bonding interactions have been also found to contribute
3.4. Stability studies revealed that RH loaded NPs were stable over a period to the mucoadhesive properties of the cationic polymer (M Ways et al.,
of 3 months 2018). Similar results were previously reported, where chitosan coated
PLGA NPs have higher contact with intestinal mucosa (de Lima et al.,
Stability studies were performed at 25 °C ± 2 °C and 60 % ± 5 % 2018) and chitosan coated liposomes developed ionic interactions with
RH over a period of 3 months. As demonstrated in Fig. 3 and in sup­ the negatively charged mucus layer (Takeuchi et al., 2003) suggesting
plementary Figure S2 (size distribution), no significant changes were the potential mucoadhesive properties of the chitosan coated NPs.
observed in the physicochemical characteristics of the NPs (size, PDI
and ζ-potential) showing good stability over a period of three months. 3.6. Only PLGA/chit NPs at high concentration induced a hemolytic effect
Despite both formulations presented comparable stability, different
mechanisms might be present. For PLGA NPs stability is likely attrib­ Hemolysis is the result of decomposition of erythrocytes membrane
uted to PVA stabilization due to steric repulsion of the adsorbed sta­ and release of hemoglobin, which is oxidized to methemoglobin and at
bilizer at their surface (Gossmann et al., 2015). On their turn, for PLGA/ the end is converted to cyanmethemoglobin (Dobrovolskaia et al.,
chit NPs the repulsive forces originated from the high surface charge 2008). Hemolytic values above 5 % were considered as hemolysis, as
might be the major stabilizer (Sharma et al., 2015). suggested by the American Society for Testing and Materials Interna­
tional (ASTM, 2013) (da Silva et al., 2019).
3.5. Chitosan presence in PLGA/chit NPs promotes mucoadhesion From the formulations tested, only blank PLGA/chit NPs, at the
higher (1700 μg/mL) concentration tested, induced hemolysis
The mucoadhesion ability of PLGA and PLGA/chit NPs was in­ (∼18 %). All other formulations had an hemolytic value below 5 % in
directly evaluated by assessing the ability of the NPs to adsorb mucin, a the concentration rage tested (417–1700 µg/mL). These results are il­
protein typically secreted by mucosal epithelial tissues (Takeuchi et al., lustrated in supplementary Figure S3. Regarding PLGA NPs, we can
2005). The surface charge of NPs and mucin was determined in SNES hypothesize that the negative surface charge contributed to the lack of
prior to incubation of their mixture. Following the incubation and the hemolytic effect. Instead, the high positive charge of PLGA/chit NPs
removal of non-adsorbed mucin, ζ-potential of NPs in SNES were re­ from the presence of amino groups of chitosan could be responsible for
corded. The presence of mucin had little or no effect on the surface the hemolytic action (Dobrovolskaia et al., 2008). Similar results to the
negative charge of PLGA NPs (p > 0.05), as indicated in Table 2, ones observed by us can be found in scientific literature. The absence of
possibly due to their anionic charge and the repulsive forces with mucin hemolytic action of PLGA NPs after different incubation’s time with red
(Takeuchi et al., 2003). On the contrary, the change of the ζ-potential of blood cells in the concentration of 3000 μg/mL (Fornaguera et al.,
PLGA/chit NPs towards negative values indicated the development of 2015) and in another study, NPs modified with chitosan derivatives
electrostatic interactions between the negatively charged sialic acid incubated (range of 250 μg/mL to 2000 μg/mL), caused hemolysis (Zhu
residues of the mucin glycoprotein and the positively charged amino et al., 2007). On the other hand, the encapsulation of RH diminished
the hemolytic action of PLGA/chit NPs. This result is in agreement with
Table 2 Zhu et al. (2007), who referred the diminishing of hemolytic action of
Assessment of the mucoadhesive properties of PLGA and PLGA/chit NPs cationic NPs after insulin encapsulation, due to the development of
through ζ-potential measurements. NPs were suspended in SNES and incubated electrostatic interactions between NPs and insulin instead NPs and er­
for 1 h with mucin solution. ζ-potential measurements were performed before
ythrocytes (Zhu et al., 2007). In the present study, the absence of he­
and after incubation. Data (expressed in mV) are presented as mean ±
molytic effect of the RH loaded PLGA/chit NPs may also be explained
standard deviation (SD), n ≥ 3 (minimum of 3 independent batches).
by the possible presence of the RH on the surface of the particles which
Formulation ζ-potential (mV) hides the effect of chitosan.
NPs suspended in SNES After 1 h incubation with mucin
in SNES 3.7. Only PLGA/chit NPs induce a decrease in the cellular viability of
PBMCs and RAW 264.7
PLGA NPs −3.68 ± 1.90 −4.80 ± 1.26
PLGA/chit NPs +10.45 ± 2.04 −2.03 ± 0.78
Peripheral blood mononuclear cells (PBMCs) contain the precursors

6
A.-T. Chatzitaki, et al. International Journal of Pharmaceutics 589 (2020) 119776

Fig. 4. Cytotoxicity assays (MTT) performed in A) PBMCs and B) RAW 264.7 cell line, after 24 h of incubation with NPs without and with drug. Data are presented as
mean ± SEM, n ≥ 4 (four or more independent experiments, each in triplicate).

of important immune cells present on mucosal surfaces. The macro­ 3.8. The presence of chitosan in the NPs increases the NPs cellular
phages in particular, can be found in the nasal cavity, presenting a interaction, but does not increase the cellular uptake
major role in mucosal immunity (Kiyono and Fukuyama, 2004).
Therefore, PBMCs isolated from human blood and Raw 264.7 murine Protein adsorption studies to NPs surface were initially performed
macrophages were treated with different doses of unloaded and RH with different proteins including BSA. From the range of protein con­
loaded PLGA and PLGA/chit NPs as well as RH solution in order to centrations tested (10 μg/mL to 100 μg/mL), the results revealed that
investigate the influence of these materials on cellular viability of cells. the highest protein adsorption efficacy was observed at the ratio
Results in Fig. 4A illustrate that PLGA NPs and RH loaded PLGA NPs did 100:960 (µg protein:µg NPs) (Supplementary material, Table S2, Figure
not cause a decrease in cellular viability in PBMCs in all tested con­ S4). Therefore, this ratio was used to prepare the protein loaded NPs for
centrations, since cell viability was above 70 % of the control (Paul the cellular (PBMCs) uptake studies. For that, we have used BSA-FITC
et al., 2013) and the same happened with RH solution. On the other adsorbed PLGA and PLGA/chit NPs, as well as rhodamine B loaded
hand, a decrease in cell viability was induced by PLGA/chit NPs (un­ PLGA and PLGA/chit NPs. Results from BSA-FITC adsorbed NPs are
loaded and loaded), probably due to NPs positive surface charge (Deka illustrated in Fig. 5. Both PLGA and PLGA/chit NPs enhanced the up­
et al., 2016) and apoptotic and membrane rupture properties of chit­ take of BSA-FITC. Nonetheless, PLGA/chit NPs induced a higher uptake,
osan (de Lima et al., 2018). This decrease allowed to calculate the half which may be due to the electrostatic interactions between the positive
maximal inhibitory concentration (IC50) induced by unloaded and RH charged NPs surface and the negatively charged cell membrane. The
loaded PLGA/chit NPs, at 1414 μg/mL and 1086 μg/mL, respectively. positive effect of chitosan on cellular uptake has already been described
Similar results were found in Raw 264.7 cells (Fig. 4B). Again, PLGA by Sheng et al. (2015), who used trimethyl chitosan coated PLGA NPs
NPs and RH solution, in concentration range of 1.8 μg/mL to 1875 μg/ and reported the enhancement of cellular uptake in comparison to
mL did not induce cell death. On its turn, unloaded and RH loaded uncoated PLGA NPs (Sheng et al., 2015). To distinguish between the
PLGA/chit NPs induced a decrease in cell viability at concentrations amount of BSA-FITC that was effectively internalized by the cells and
higher than 14.8 μg/mL, with the IC50 calculated at 19.3 μg/mL and the BSA-FITC that was adsorbed at the cell surface, the cells were
15.6 μg/mL, respectively. The difference in the IC50 of PLGA/chit NPs, treated with trypan blue to quench the extracellular fluorescence
observed between PBMCs and Raw 264.7, may be explained by the (Fig. 5A). With this approach, the decrease in the fluorescence intensity
sensitivity of macrophage cell line, which is related to their higher revealed that BSA-FITC and hypothetically the NPs were mainly ad­
degree of cellular differentiation (Pilling et al., 2017; Way et al., 2009). sorbed at cell surface as previously reported elsewhere for chitosan NPs
(Huang et al., 2004). These results were confirmed by a second

7
A.-T. Chatzitaki, et al. International Journal of Pharmaceutics 589 (2020) 119776

Fig. 5. Evaluation of cellular uptake of BSA-FITC adsorbed NPs by PBMCs. A) Total uptake and cellular uptake by lymphocytes and monocytes after incubation with
protein adsorbed NPs1 for 2 h. Bars represent mean ± SEM, n = 3. B) Confocal images of PBMCs after incubation with BSA-FITC adsorbed NPs (green). Cells were
labeled with Hoechst 33342 (blue) to stain the nucleus and Alexa 594 to identify cell membrane. Scale bar: 5 μm.

technique, the confocal laser scanning microscopy (CLSM) (Fig. 5B). study the delivery of RH and also to follow the NPs location after in­
The MTT assay was carried out after 2 h incubation of cells with BSA- cubation with cells. Both NPs were able to be internalized by lympho­
FITC NPs, to elucidate the effect of these NPs under the conditions of cytes and monocytes (Fig. 6A). Despite we did not exclude the extra­
the uptake studies. Results revealed that cell viability during the assay cellular signal of Rhodamine-B as performed with trypan blue for FITC
was superior to 70 % of control for all the formulations tested. signal, confocal images revealed the cell cytoplasm full of Rhodamine-B
Rhodamine-B loaded PLGA and PLGA/chit NPs were prepared to loaded NPs (Fig. 6B). These images support the conclusion that drug

8
A.-T. Chatzitaki, et al. International Journal of Pharmaceutics 589 (2020) 119776

Fig. 6. Assessment of cellular uptake of different Rhodamine-B loaded NPs by PBMCs. A) Cellular uptake by lymphocytes and monocytes after incubation with
Rhodamine B loaded PLGA and PLGA/chit NPs for 2 h. Bars represent mean ± SEM, n = 3 (3 independent blood donors). B) Confocal images of PBMCs after
incubation with Rhodamine-B loaded NPs2 (red). Cells were labeled with Hoechst 33342 (blue) to stain the nucleus. Scale bar: 10 μm.

loaded PLGA and PLGA/chit NPs were able to enter into cells, where counterbalance ROS production. However, upon stimulus, the over­
they have the opportunity to efficiently delivering the loaded cargo. As production of ROS by activated macrophages can promote oxidative
described for BSA-FITC loaded NPs, Rhodamine-B loaded PLGA and stress and consistently cell injury (da Silva et al., 2019).
PLGA/chit NPs did not induce cellular death, as determined with the The RAW 264.7 cell line was incubated for 24 h with RH loaded and
MTT assay at the conditions of experiment. unloaded PLGA and PLGA/chit NPs to assess ROS production. Both
In conclusion, both delivery systems had a similar ability to promote formulations, loaded and unloaded did not induce cells to produce ROS
the intracellular delivery of proteins and hydrophilic drugs, but PLGA/ as presented in Fig. 7A. Importantly, by using the same concentrations
chit NPs showed a higher ability to interact with the cellular mem­ for both formulations, we could observe that PLGA NPs (loaded and
branes, due to electrostatic forces between positive charge of chitosan unloaded) at the higher tested concentrations did not induce cellular
and anionic cell surface characteristic. death, while PLGA/chit NPs (loaded and unloaded), decreased cellular
viability near to 70 % at 100 μg/mL (Fig. 7B). The absence of ROS
3.9. None of the developed formulations induces the production of reactive production in this concentration, might suggest that PLGA/chit NPs
oxygen species induced cellular death is not dependent on ROS signaling and may be
attributed to apoptosis or cell membrane rupture, induced by chitosan
It has been reported that NPs can induce cells to produce ROS as a (de Lima et al., 2018).
result of metabolic activity (Fu et al., 2014; Kim et al., 2015; Magder,
2006). On a regular basis, there is a defence mechanism in order to

9
A.-T. Chatzitaki, et al. International Journal of Pharmaceutics 589 (2020) 119776

Fig. 7. ROS assay. A) ROS production


after 24 h incubation of RAW 264.7
macrophage cell line with different
concentrations of unloaded and RΗ
loaded NPs. LPS (1 µg/mL) was used as
a positive control and unstimulated
cells as a negative control. Results are
presented in fluorescence increase fold
compared to the negative control. B)
Cell viability assay (MTT) after the
performance of ROS production assay.
****p < 0.0001.

3.10. Ex vivo permeability studies confirmed lipid-based NPs and lipid-polymer NPs as efficient and safety
carriers, releasing more than 61 % RH across sheep nasal mucosa
Ex vivo permeation studies with RH loaded NPs were performed (Pardeshi et al., 2013a,b). On their turn, Patil and Surana (2013) de­
across sheep nasal mucosa and results are depicted in Table 3 and veloped PLGA NPs for the nasal administration of RH and showed
Fig. 8. The steady state diffusion coefficient was calculated to be significantly higher ex vivo drug permeation across sheep nasal mucosa
0.16·10−6 cm2 min−1 for the PLGA NPs and 0.52·10−6 cm2 min−1 for (97.9 ± 2.9 %) compared to RH solution (74.43 ± 4.6 %) (Patil and
the PLGA/chit NPs. A slow drug permeation profile across nasal mucosa Surana, 2013). Unfortunately, no direct comparisons can be performed
was observed for the RH loaded PLGA NPs (5.5 µg/cm2 in 4 h), which is with our results since both authors present the percentage of permeated
in accordance with the slow in vitro release of RH from the same NPs, RH (from the total added) instead of μg of drug permeated per mucosal
indicating that drug diffusion from the NPs might be the rate limiting surface area (cm2).
factor for drug permeation across the tissue (Seju et al., 2011). On the It has been previously reported that the mechanism of RH per­
contrary, PLGA/chit NPs exhibited a higher permeation profile (32 µg/ meation following intranasal administration occurs through the para­
cm2 in 4 h) resulting in a 3.22-fold increase of apparent permeability cellular pathway, passing through the hydrophilic passages between
coefficient from 2.16 ± 0.4·10−6 cm min−1 for PLGA NPs to adjacent epithelial cells (Pardeshi and Belgamwar, 2016). Nevertheless,
6.96 ± 2.7·10−6 cm min−1. In comparison to previous work from our the higher RH permeability observed for the PLGA/chit NPs could be
group on RH loaded PLGA microparticles containing TMC (Karavasili attributed not only to the presence of chitosan, acting as both a per­
et al., 2016), the RH – PLGA/chit NPs exhibited a better permeability meation enhancer and an opener of the tight junctions of the nasal
profile. In detail, these NPs induced a 1.68-fold increase in RH per­ epithelium (Khan et al., 2010; Mistry et al., 2009), but also to the higher
meability when compared to the microparticles and a 3.96-fold increase drug release from the chitosan coated NPs. In addition to that, it has
when compared to n RH solution (Karavasili et al., 2016). been previously shown that NPs with size less than 500 nm, as in the
In reports from Pardeshi et al. (2013), solid lipid NPs and lipid present study, interact efficiently with the mucus layer of the nasal
combined with polymer NPs loaded with RH were evaluated regarding cavity, therefore increasing NP retention time and hence drug release
their permeation properties across sheep nasal mucosa. Their results and absorption (Sonvico et al., 2018). Similar observations were made
for chitosan-grafted PLGA NPs with a particle size of 463.9 ± 12 nm
Table 3 formulated for the intranasal delivery of chloropromazine (Chalikwar
Steady state flux (Jss) and apparent permeability values (Papp) of RH-PLGA NPs et al., 2013).
and RH-PLGA/chit NPs across sheep nasal mucosa. Results are the mean The effect of the NP-based formulations on tissue integrity was as­
value ± SD, n ≥ 3. sessed with histopathological analysis after the end of the ex vivo per­
Jss (μg cm−2 min−1) Papp (10−6 cm min−1) meability studies. Untreated epithelium was used as the control. No
significant differences on mucosal morphology were observed after
RH – PLGA NPs 0.056 ± 0.01 2.16 ± 0.4 tissue treatment with RH loaded PLGA and PLGA/chit NPs compared to
RH – PLGA/chit NPs 0.18 ± 0.07 6.96 ± 2.7
the control, whereas only PLGA NPs were found to induce a minor

10
A.-T. Chatzitaki, et al. International Journal of Pharmaceutics 589 (2020) 119776

Fig. 8. Ex vivo permeability studies of RH loaded from PLGA and PLGA/chit NPs across sheep nasal mucosa. A) Permeability studies across nasal mucosa at several
time points (PLGA NPs in red color, PLGA/chit NPs in blue color). Results are presented as mean value ± SEM, n ≥ 3. Histopathological studies of (B) untreated and
(C) treated nasal mucosa with RΗ loaded PLGA NPs and (D) RΗ loaded PLGA/chit NPs after 4 h incubation. Scale bar: 100 μm.

epithelial superficial desquamation. The integrity of the epithelial layer influence the work reported in this paper.
and the presence of goblet cells and mucus layer indicated the absence
of harmful properties of both formulations. Acknowledgements

4. Conclusions This work was financed by the European Regional Development


Fund (ERDF), through the Centro 2020 Regional Operational
In the present work, mucoadhesive polymeric PLGA/chit NPs were Programme and through the COMPETE 2020 – Operational Programme
efficiently developed through a simple nanoprecipitation technique for for Competitiveness and Internationalization and Portuguese national
the nasal administration of RH. The drug loaded PLGA/chit NPs had funds via FCT – Fundação para a Ciência e Tecnologia, under project
acceptable particle size and a strong positive charge, rendering them PROSAFE/0001/2016, POCI-01-0145-FEDER-030331 and the strategic
highly mucoadhesive, compared to PLGA NPs, as indicated by the in project UIDB/04539/2020. The authors thank Dr. Ana Donato for as­
vitro mucin adsorption studies. In vitro drug release and ex vivo per­ sistance in hemolysis studies in the Clinical Analysis Laboratory of the
meability studies through nasal mucosa revealed a significantly faster Faculty of Pharmacy of Coimbra University (Portugal), Dr. Mónica
RH release and a 3.22-fold enhancement in the Papp of RH from the Zuzarte for the TEM microscopy observations performed at
PLGA/chit NPs, while the presence of chitosan also favored PBMC iLAB—Bioimaging Laboratory of the Faculty of Medicine of the
cellular interaction. Although PLGA/chit NPs demonstrated in vitro University of Coimbra and Dr. Luísa Cortes for the Confocal microscopy
lower biocompatibility, toxic effects induced were dose dependent, and analysis performed at the Microscopy Imaging Center of Coimbra, at the
observed only at very high concentrations, not relatable to in vivo do­ Center for Neuroscience and Cell Biology.
sages. Overall, these results show that RH loaded PLGA/chit NPs is a
valuable delivery system that needs to be tested in a suitable animal Appendix A. Supplementary material
model through nasal administration. These results are the basis of hope
needed to find new therapeutic approaches for Parkinson’s disease. Supplementary data to this article can be found online at https://
doi.org/10.1016/j.ijpharm.2020.119776.
CRediT authorship contribution statement
References
Aikaterini-Theodora Chatzitaki: Investigation, Writing - original
Azevedo, J.R., Sizilio, R.H., Brito, M.B., Costa, A.M.B., Serafini, M.R., Araújo, A.A.S.,
draft. Sandra Jesus: Methodology, Validation. Christina Karavasili: Santos, M.R. v, Lira, A.A.M., Nunes, R.S., 2011. Physical and chemical character­
Methodology. Dimitrios Andreadis: Methodology. Dimitrios G. ization insulin-loaded chitosan-TPP nanoparticles. Journal of Thermal Analysis and
Fatouros: Conceptualization, Validation, Resources, Supervision. Olga Calorimetry 106, 685–689. https://doi.org/10.1007/s10973-011-1429-5.
Barcia, E., Boeva, L., García-García, L., Slowing, K., Fernández-Carballido, A., Casanova,
Borges: Conceptualization, Validation, Resources, Supervision. Y., Negro, S., 2017. Nanotechnology-based drug delivery of ropinirole for Parkinson’s
disease. Drug Delivery 24, 1112–1123. https://doi.org/10.1080/10717544.2017.
1359862.
Declaration of Competing Interest Bourganis, V., Kammona, O., Alexopoulos, A., Kiparissides, C., 2018. Recent advances in
carrier mediated nose-to-brain delivery of pharmaceutics. Eur. J. Pharm. Biopharm.
128, 337–362. https://doi.org/10.1016/j.ejpb.2018.05.009.
The authors declare that they have no known competing financial
Chalikwar, S.S., Mene, B.S., Pardeshi, C.v., Belgamwar, V.S., Surana, S.J., 2013. Self-
interests or personal relationships that could have appeared to

11
A.-T. Chatzitaki, et al. International Journal of Pharmaceutics 589 (2020) 119776

assembled, chitosan grafted PLGA nanoparticles for intranasal delivery: design, de­ Mohammed, M.A., Syeda, J.T.M., Wasan, K.M., Wasan, E.K., 2017. An overview of
velopment and ex vivo characterization. Polymer-Plastics Technol. Eng. 52, 368–380. chitosan nanoparticles and its application in non-parenteral drug delivery.
https://doi.org/10.1080/03602559.2012.751999. Pharmaceutics 9, 53. https://doi.org/10.3390/pharmaceutics9040053.
da Silva, J., Jesus, S., Bernardi, N., Colaço, M., Borges, O., 2019. Poly(D, L-lactic acid) Neto, C.G.T., Giacometti, J.A., Job, A.E., Ferreira, F.C., Fonseca, J.L.C., Pereira, M.R.,
nanoparticle size reduction increases its immunotoxicity. Front. Bioeng. Biotechnol. 2005. Thermal analysis of chitosan based networks. Carbohydr. Polym. 62, 97–103.
7, 137. https://doi.org/10.3389/fbioe.2019.00137. https://doi.org/10.1016/j.carbpol.2005.02.022.
Deka, C., Aidew, L., Devi, N., Buragohain, A.K., Kakati, D.K., 2016. Synthesis of curcumin- Pardeshi, C.V., Belgamwar, V.S., 2017. Ropinirole-dextran sulfate nanoplex for nasal
loaded chitosan phosphate nanoparticle and study of its cytotoxicity and anti­ administration against Parkinson’s disease: in silico molecular modeling and in vi­
microbial activity. J. Biomater. Sci. Polym. Ed. 27, 1659–1673. https://doi.org/10. tro–ex vivo evaluation. Artif. Cells Nanomed. Biotechnol. 45, 635–648. https://doi.
1080/09205063.2016.1226051. org/10.3109/21691401.2016.1167703.
Dobrovolskaia, M.A., Aggarwal, P., Hall, J.B., McNeil, S.E., 2008. Preclinical studies to Pardeshi, C.V., Belgamwar, V.S., 2013. Direct nose to brain drug delivery via integrated
understand nanoparticle interaction with the immune system and its potential effects nerve pathways bypassing the blood–brain barrier: an excellent platform for brain
on nanoparticle biodistribution. Mol. Pharm. 5, 487–495. https://doi.org/10.1021/ targeting. Expert Opinion on Drug Delivery 10, 957–972. https://doi.org/10.1517/
mp800032f. 17425247.2013.790887.
El-Houssiny, A.S., Ward, A.A., Mostafa, D.M., Abd-El-Messieh, S.L., Abdel-Nour, K.N., Pardeshi, C.V, Belgamwar, V.S., 2018. N,N,N-trimethyl chitosan modified flaxseed oil
Darwish, M.M., Khalil, W.A., 2016. Drug–polymer interaction between glucosamine based mucoadhesive neuronanoemulsions for direct nose to brain drug delivery.
sulfate and alginate nanoparticles: FTIR, DSC and dielectric spectroscopy studies. International Journal of Biological Macromolecules 120, 2560–2571. https://doi.
Adv. Nat. Sci.: Nanosci. Nanotechnol. 7, 025014. https://doi.org/10.1088/2043- org/https://doi.org/10.1016/j.ijbiomac.2018.09.032.
6262/7/2/025014. Pardeshi, C.V, Belgamwar, V.S., 2016. Controlled synthesis of N,N,N-trimethyl chitosan
Fornaguera, C., Calderó, G., Mitjans, M., Vinardell, M.P., Solans, C., Vauthier, C., 2015. for modulated bioadhesion and nasal membrane permeability. International Journal
Interactions of PLGA nanoparticles with blood components: protein adsorption, of Biological Macromolecules 82, 933–944. https://doi.org/https://doi.org/10.
coagulation, activation of the complement system and hemolysis studies. Nanoscale 1016/j.ijbiomac.2015.11.012.
7, 6045–6058. https://doi.org/10.1039/C5NR00733J. Pardeshi, C.V, Belgamwar, V.S., Tekade, A.R., Surana, S.J., 2013a. Novel surface modified
Fu, P.P., Xia, Q., Hwang, H.-M., Ray, P.C., Yu, H., 2014. Mechanisms of nanotoxicity: polymer–lipid hybrid nanoparticles as intranasal carriers for ropinirole hydro­
Generation of reactive oxygen species. J. Food Drug Anal. 22, 64–75. https://doi.org/ chloride: in vitro, ex vivo and in vivo pharmacodynamic evaluation. Journal of
10.1016/j.jfda.2014.01.005. Materials Science: Materials in Medicine 24, 2101–2115. https://doi.org/10.1007/
Gänger, S., Schindowski, K., 2018. Tailoring formulations for intranasal nose-to-brain s10856-013-4965-7.
delivery: a review on architecture, physico-chemical characteristics and mucociliary Pardeshi, C.V, Rajput, P.V, Belgamwar, V.S., Tekade, A.R., Surana, S.J., 2013b. Novel
clearance of the nasal olfactory mucosa. Pharmaceutics 10, 116. https://doi.org/10. surface modified solid lipid nanoparticles as intranasal carriers for ropinirole hy­
3390/pharmaceutics10030116. drochloride: application of factorial design approach. Drug Delivery 20, 47–56.
Gossmann, R., Langer, K., Mulac, D., 2015. New perspective in the formulation and https://doi.org/10.3109/10717544.2012.752421.
characterization of didodecyldimethylammonium bromide (DMAB) stabilized poly Patil, G.B., Surana, S.J., 2013. Fabrication and statistical optimization of surface en­
(lactic-co-glycolic acid) (PLGA) nanoparticles. PLoS ONE 10https://doi.org/10.1371/ gineered PLGA nanoparticles for naso-brain delivery of ropinirole hydrochloride: in-
journal.pone.0127532. e0127532 e127532. vitro–ex-vivo studies. J. Biomater. Sci. Polym. Ed. 24, 1740–1756. https://doi.org/
Govender, T., Stolnik, S., Garnett, M.C., Illum, L., Davis, S.S., 1999. PLGA nanoparticles 10.1080/09205063.2013.798880.
prepared by nanoprecipitation: drug loading and release studies of a water soluble Paul, A., Das, S., Das, J., Samadder, A., Khuda-Bukhsh, A., 2013. Cytotoxicity and
drug. J. Control. Release 57, 171–185. https://doi.org/10.1016/S0168-3659(98) apoptotic signalling cascade induced by chelidonine-loaded PLGA nanoparticles in
00116-3. HepG2 cells in vitro and bioavailability of nano-chelidonine in mice in vivo. Toxicol.
Guo, C., Gemeinhart, R.A., 2008. Understanding the adsorption mechanism of chitosan Lett. 222. https://doi.org/10.1016/j.toxlet.2013.07.006.
onto poly(lactide-co-glycolide) particles. European journal of pharmaceutics and Pilling, D., Galvis-Carvajal, E., Karhadkar, T.R., Cox, N., Gomer, R.H., 2017. Monocyte
biopharmaceutics : official journal of Arbeitsgemeinschaft fur Pharmazeutische differentiation and macrophage priming are regulated differentially by pentraxins
Verfahrenstechnik e.V 70, 597–604. https://doi.org/10.1016/j.ejpb.2008.06.008. and their ligands. BMC Immunol. 18, 30. https://doi.org/10.1186/s12865-017-
Huang, M., Khor, E., Lim, L.-Y., 2004. Uptake and cytotoxicity of chitosan molecules and 0214-z.
nanoparticles: effects of molecular weight and degree of deacetylation. Pharm. Res. Rao, M., Agrawal, D.K., Shirsath, C., 2017. Thermoreversible mucoadhesive in situ nasal
21, 344–353. https://doi.org/10.1023/B:PHAM.0000016249.52831.a5. gel for treatment of Parkinson’s disease. Drug Dev. Ind. Pharm. 43, 142–150. https://
Jesus, S., Marques, A.P., Duarte, A., Soares, E., Costa, J.P., Colaço, M., Schmutz, M., Som, doi.org/10.1080/03639045.2016.1225754.
C., Borchard, G., Wick, P., Borges, O., 2020. Chitosan nanoparticles: shedding light on Sahoo, S., Panyam, J., Prabha, S., Labhasetwar, V., 2002. Residual polyvinyl alcohol
immunotoxicity and hemocompatibility. Front. Bioeng. Biotechnol. 8, 100. associated with poly (D, L-lactide-co-glycolide) nanoparticles affects their physical
Karavasili, C., Bouropoulos, N., Sygellou, L., Amanatiadou, E.P., Vizirianakis, I.S., properties and cellular uptake. J. Controlled Release : Off. J. Controlled Release Soc.
Fatouros, D.G., 2016. PLGA/DPPC/trimethylchitosan spray-dried microparticles for 82, 105–114. https://doi.org/10.1016/S0168-3659(02)00127-X.
the nasal delivery of ropinirole hydrochloride: in vitro, ex vivo and cytocompatibility Seju, U., Kumar, A., Sawant, K.K., 2011. Development and evaluation of olanzapine-
assessment. Mater. Sci. Eng., C 59, 1053–1062. https://doi.org/10.1016/j.msec. loaded PLGA nanoparticles for nose-to-brain delivery: in vitro and in vivo studies.
2015.11.028. Acta Biomater. 7, 4169–4176. https://doi.org/10.1016/j.actbio.2011.07.025.
Khan, S., Patil, K., Bobade, N., Yeole, P., Gaikwad, R., 2010. Formulation of intranasal Sharma, D., Sharma, R.K., Sharma, N., Gabrani, R., Sharma, S.K., Ali, J., Dang, S., 2015.
mucoadhesive temperature-mediated in situ gel containing ropinirole and evaluation Nose-to-brain delivery of PLGA-diazepam nanoparticles. AAPS PharmSciTech 16,
of brain targeting efficiency in rats. J. Drug Target. 18, 223–234. https://doi.org/10. 1108–1121. https://doi.org/10.1208/s12249-015-0294-0.
3109/10611860903386938. Sheng, J., Han, L., Qin, J., Ru, G., Li, R., Wu, L., Cui, D., Yang, P., He, Y., Wang, J., 2015.
Kim, K.S., Lee, D., Song, C.G., Kang, P.M., 2015. Reactive oxygen species-activated na­ N-trimethyl chitosan chloride-coated PLGA nanoparticles overcoming multiple bar­
nomaterials as theranostic agents. Nanomedicine (London, England) 10, 2709–2723. riers to oral insulin absorption. ACS Appl. Mater. Interfaces 7, 15430–15441. https://
https://doi.org/10.2217/nnm.15.108. doi.org/10.1021/acsami.5b03555.
Kiyono, H., Fukuyama, S., 2004. NALT- versus PEYER’S-patch-mediated mucosal im­ Sonvico, F., Clementino, A., Buttini, F., Colombo, G., Pescina, S., Stanisçuaski Guterres, S.,
munity. Nat. Rev. Immunol. 4, 699–710. https://doi.org/10.1038/nri1439. Raffin Pohlmann, A., Nicoli, S., 2018. Surface-modified nanocarriers for nose-to-brain
Kumar, M., Misra, A., Mishra, A.K., Mishra, P., Pathak, K., 2008. Mucoadhesive nanoe­ delivery: from bioadhesion to targeting. Pharmaceutics 10, 34. https://doi.org/10.
mulsion-based intranasal drug delivery system of olanzapine for brain targeting. J. 3390/pharmaceutics10010034.
Drug Target. 16, 806–814. https://doi.org/10.1080/10611860802476504. Takeuchi, H., Matsui, Y., Yamamoto, H., Kawashima, Y., 2003. Mucoadhesive properties
Kumar, M., Pandey, R.S., Patra, K.C., Jain, S.K., Soni, M.L., Dangi, J.S., Madan, J., 2013. of carbopol or chitosan-coated liposomes and their effectiveness in the oral admin­
Evaluation of neuropeptide loaded trimethyl chitosan nanoparticles for nose to brain istration of calcitonin to rats. J. Control. Release 86, 235–242. https://doi.org/10.
delivery. Int. J. Biol. Macromol. 61, 189–195. https://doi.org/10.1016/j.ijbiomac. 1016/S0168-3659(02)00411-X.
2013.06.041. Takeuchi, H., Thongborisute, J., Matsui, Y., Sugihara, H., Yamamoto, H., Kawashima, Y.,
de Lima, I.A., Khalil, N.M., Tominaga, T.T., Lechanteur, A., Sarmento, B., Mainardes, 2005. Novel mucoadhesion tests for polymers and polymer-coated particles to design
R.M., 2018. Mucoadhesive chitosan-coated PLGA nanoparticles for oral delivery of optimal mucoadhesive drug delivery systems. Adv. Drug Deliv. Rev. 57, 1583–1594.
ferulic acid. Artif. Cells Nanomed. Biotechnol. 46, 993–1002. https://doi.org/10. https://doi.org/10.1016/j.addr.2005.07.008.
1080/21691401.2018.1477788. Way, K.J., Dinh, H., Keene, M.R., White, K.E., Clanchy, F.I.L., Lusby, P., Roiniotis, J.,
M Ways, T.M., Lau, W.M., Khutoryanskiy, V. v, 2018. Chitosan and Its Derivatives for Cook, A.D., Cassady, A.I., Curtis, D.J., Hamilton, J.A., 2009. The generation and
Application in Mucoadhesive Drug Delivery Systems. Polymers 10, 267. https://doi. properties of human macrophage populations from hemopoietic stem cells. J. Leukoc.
org/10.3390/polym10030267. Biol. 85, 766–778. https://doi.org/10.1189/jlb.1108689.
Magder, S., 2006. Reactive oxygen species: toxic molecules or spark of life? Critical care Zhu, S., Qian, F., Zhang, Y., Tang, C., Yin, C., 2007. Synthesis and characterization of PEG
(London, England) 10, 208. https://doi.org/10.1186/cc3992. modified N-trimethylaminoethylmethacrylate chitosan nanoparticles. Eur. Polym. J.
Mistry, A., Stolnik, S., Illum, L., 2009. Nanoparticles for direct nose-to-brain delivery of 43, 2244–2253. https://doi.org/10.1016/j.eurpolymj.2007.03.042.
drugs. Int. J. Pharm. 379, 146–157. https://doi.org/10.1016/j.ijpharm.2009.06.019.

12

You might also like