You are on page 1of 12

Journal of Drug Delivery Science and Technology 63 (2021) 102549

Contents lists available at ScienceDirect

Journal of Drug Delivery Science and Technology


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

Review article

Donepezil—an updated review of challenges in dosage form design


Lalinthip Sutthapitaksakul a, b, Crispin R. Dass c, d, Pornsak Sriamornsak a, b, e, *
a
Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand
b
Pharmaceutical Biopolymer Group (PBiG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand
c
School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin University, Perth, 6845, Australia
d
Curtin Health Innovation Research Institute, Bentley, 6102, Australia
e
Academy of Science, The Royal Society of Thailand, Bangkok, 10300, Thailand

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

Keywords: Donepezil hydrochloride (DPH) was first launched in 1997 as a choline esterase inhibitor (ChEI). The USFDA has
Donepezil approved it for treatment of Alzheimer’s disease since it effectively enhances cholinergic activity by slowing the
Dosage form design clearance of acetylcholine in the brain. Although it is the most prescribed drug among ChEIs, a bitter taste with
Orodispersible
numbness to DPH is a substantial oral administration obstacle, and is a frequently reported adverse event.
Transdermal
Intranasal
Several studies have been conducted to overcome this problem by adding sweeteners and flavors, application of
Parenteral physical barriers, preparation of ion exchange resins, and inclusion complexes. Based on the pharmacological
Nose-to-brain delivery activity, the gastrointestinal effect is a frequently reported adverse event. Therefore, novel dosage forms intended
Taste masking to deliver through oral, topical/transdermal, parenteral, and intranasal routes have been designed and devel­
oped. In this review, findings from recent studies on DPH are summarized, and the challenges and obstacles of
DPH dosage form development are highlighted and discussed.

1. Introduction originated from the abnormal cleavage of amyloid precursor protein


(APP) in the non-amyloidogenic pathway [4,5]. It is believed that the
1.1. Background of Alzheimer’s disease deposition of NFTs and Aβ plaques, glutamate excitotoxicity, and
inflammation are the main causes of neuronal cell death [6].
Alzheimer’s disease (AD) is the most common type of dementia, To date, there are no effective medicines available to terminate or
accounting for approximately 80% of all dementia cases [1]. By 2050, reverse the destruction of neurons. The available medicines mainly
the number of people with dementia is predicted to reach 152 million. alleviate the cognitive dysfunction typical of AD patients. The United
Additionally, dementia treatment cost, which is currently estimated to States Food and Drug Administration (USFDA) currently approved four
be one trillion US dollars, is predicted to rise to two trillion US dollars by medicines to treat AD. Based on their mechanism of action, they can be
2030 [2]. Since dementia increases with age, it can be found in 10% of divided into choline esterase inhibitor (ChEI) and N-methyl-D-aspartate
people over 65 and about 50% of people over 85 years. However, it can (NMDA) receptor antagonist.
be observed in younger people at the age of 20, possibly due to genetic Promising medicines are in the class of ChEIs, including donepezil,
disorder [3]. Owing to the continuous increase in life-expectancy, the galantamine, and rivastigmine that prevent the breakdown of acetyl­
incidence of dementia is anticipated to continuously increase, especially choline (ACh). These medicines were hypothesized to be responsible for
among low-income and middle-income communities. Early-stage de­ the regulation of cognitive function. Another therapeutic agent, mem­
mentia patients are predominantly found in low-level education and antine, an NMDA receptor antagonist, is responsible for receptor
healthcare populations. blocking, resulting in neuron protection from excessive glutamate
Presently, the etiology of AD remains unclear. There are two main excretion. Combining two classes of medicines, including donepezil and
pathological hallmarks of AD, which include neurofibrillary tangles memantine, has also been approved [7]. However, this article will
(NFTs) and amyloid beta (Aβ) plaques. NFTs are generated intra- mainly focus on the development and evaluation of donepezil drug de­
neuronally through the abnormal phosphorylation of tau proteins, livery systems.
which are involved in microtubule assembly, while Aβ plaques

* Corresponding author. Department of Pharmaceutical Technology, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand.
E-mail address: sriamornsak_p@su.ac.th (P. Sriamornsak).

https://doi.org/10.1016/j.jddst.2021.102549
Received 1 March 2021; Received in revised form 23 April 2021; Accepted 23 April 2021
Available online 29 April 2021
1773-2247/© 2021 Elsevier B.V. All rights reserved.
L. Sutthapitaksakul et al. Journal of Drug Delivery Science and Technology 63 (2021) 102549

1.2. Background of donepezil 150 μg/mL [29]. Its molecular weight is 425.96 g/mol with a log P of
4.27 [25]. Although USFDA has approved hydrochloride salt, donepezil
1.2.1. Drug development free base (DPB) has been used as an active drug among several research
In 1983, donepezil was first invented in Tsukuba Research Labora­ groups. It is less water-soluble (33 g/L). The melting point analyzed by
tories, Eisai Co., Ltd., Japan, to overcome the limitations of the first differential calorimetry is 85.6 ◦ C [30]. Thus, DPB might be suitable to
generation ChEIs, such as physostigmine and tacrine (see Fig. 1). develop a lipid-based dosage form. However, its efficacy and toxicity
Although these drugs moderately enhance patients’ cognitive function, should be clearly elucidated.
poor pharmacokinetic profiles of physostigmine, and hepatotoxicity of
tacrine are still significant limitations of the drug. The development is 1.2.5. Adverse events
based on cholinergic hypothesis; it was believed that cholinergic According to its pharmacological activity, the most frequent adverse
innervation deficiency in the basal forebrain is greatly related to the events reported in at least 2% of mild to moderate AD patients at a
unusual memory loss and cognitive function in AD patients [8,9]. As the higher frequency than that of placebo in a controlled clinical trial of
resulting compound had a long half-life with moderate acetylcholine Aricept® are nausea (11%), diarrhea (10%), insomnia (9%), muscle
esterase (AChE) activity inhibition, several indanone derivatives were cramps (6%), fatigue (5%), vomiting (5%), and anorexia (4%). These
synthesized. Among these, E2020 (or donepezil hydrochloride; DPH) events frequently appeared in patients who took a higher dosage.
was discovered, and launched in 1997, in Atlanta, USA. DPH is second However, patients recovered from these events within one to three
generation ChEIs with highly selective inhibition of AChE over butyr­ weeks after continuous usage and can be reduced using six weeks of
ylcholinesterase, thus preventing the breakdown of ACh in synapses of titration dosage. Other adverse events found are insomnia, fatigue,
the central and peripheral nervous system. It enhances the cognitive muscle cramps, and anorexia. Also, DPH may have an effect on the vagus
function of patients with mild to moderate AD symptoms without hep­ nerve because of the cholinergic enhancement effect, causing the
atotoxicity [10]. blockage of sinoatrial and atrioventricular nodes. The incidence of
Torsades de pointes has also been reported. Similarly, the increase in
1.2.2. Dosing strategies cholinergic activity results in gastric acid secretion. Thus, GI hemor­
For mild to moderate AD, DPH is suggested to start at an oral dosage rhage (1%) has been reported. The concurrent use of non-steroidal anti-
of 5 mg daily and slowly titrate to 10 mg daily after four to six weeks of inflammatory drugs (NSAIDs) should be carefully monitored [11,
treatment. A 23-mg extended-release tablet can be given to patients with 31–34].
moderate to severe AD, who have been taking 10-mg DPH for at least To overcome the limitations of DPH including bitter taste and
three months [11]. adverse effect, and patient-related factors, including lack of under­
standing of their disease and medications, several novel dosage forms
1.2.3. Clinical efficacy and development technologies have been developed. In this article, it
DPH has been initially approved for mild to moderate AD in 1997 has been categorized based on DPH-related problems.
and severe AD in 2006. Its benefits have also been shown in other
conditions, including mild cognitive impairment, vascular dementia, 2. Challenges in dosage form design and development
cognitive impairment in multiple sclerosis, Parkinson’s disease, and
dementia with Lewy bodies. Compared to placebo, these studies’ clinical 2.1. Bitter taste of DPH
results showed a significant improvement of behavior, cognitive func­
tion, and daily activities [12–15]. Although recent clinical trials also As the taste sensation is caused by the interaction of soluble sub­
studied the role of DPH in patients with Down’s syndrome [16,17], stances with the taste receptors, the bitter taste with numbness of DPH
posterior cortical atrophy [18], depression, and cognitive impairment might be a major problem affecting patient compliance. The conven­
[19], there were no significant efficacy in these patients. tional tablets do not require taste-masking because the dosage form
Previous studies indicated that DPH is a symptomatic treatment. would not disintegrate or dissolve until passing through the oral cavity.
However, several recent publications have documented the disease- The effective taste-masking technique may be a prerequisite for some
modifying potential of such a drug. The neuroprotective effect has dosage forms such as oral solutions, suspensions, lozenges, chewable
been demonstrated in different AD models, including oxygen-glucose tablets, orally disintegrating tablets (ODTs), and orally disintegrating
deprivation [20], Aβ-induced cell death [21–23] and films (ODFs), where the actives possibly contact the taste receptors [35].
glutamate-induced cell death [24]. Therefore, this effect might help to In the innovator product, Aricept® ODT, carrageenan is incorporated
delay disease progression from cognitive impairment to AD [6]. into the ODTs to suppress the bitterness of DPH in the oral cavity. As
drug molecules intermediated within the sulfate groups of ɩ-carra­
1.2.4. Physicochemical properties geenan, the bitterness was masked [11,36]. Besides this product, Table 1
DPH is freely soluble in water (55 g/L at 25 ◦ C), chloroform, and shows several attempts to develop taste-masking dosage forms to over­
glacial acetic acid. It has a tertiary amine group with 8.9 pKa [25]. By come such a problem.
thermal analysis, the melting point is reported to be 224 ◦ C [26,27]. It is
a white to off-white solid with an intensely bitter taste. The bitterness 2.1.1. Addition of sweetening and flavoring agent
threshold recognized by six volunteers was reported to be 15–20 μg/mL One of the low-cost and straightforward taste-masking techniques is
[28]. However, Zainuddin et al. [29] demonstrated a different result of sensory masking by adding sweeteners and flavors. In a study by Liew

Fig. 1. Chemical structures of (a) physostigmine, (b) tacrine, and (c) DPH.

2
L. Sutthapitaksakul et al. Journal of Drug Delivery Science and Technology 63 (2021) 102549

Table 1
The oral dosage forms developed to mask the bitter taste of DPH.
Dosage Taste-masking Dose Optimized In vitro taste evaluation In vivo taste evaluation Reference
form strategy (mg) formulation
Taste evaluation Taste-masking Taste Bitterness Taste-masking
method criteria evaluation threshold evaluation
condition (μg/mL) procedure

ODTs Preparation of 5 Microspheres The sample was DPH One tablet 15.0–20.0 Bitterness intensity [28]
microspheres of containing DPH- shaken. concentration placed on the scale
DPH-Eudragit® EPO Eudragit® EPO at M: SSF (pH 6.2) after shaking < tongue
by spray drying the ratio of 1:2 V: 10 mL bitterness No.: 6
method Time: 60 s threshold t: after tablet
T: n.s. was
disintegrated
and held in the
mouth for 60s
ODFs Addition of 10 HPMC film based n.s. n.s. One film strip n.s. Palatability scale [37]
sweeteners containing 7% of placed on the (Taste, aftertaste,
aspartame, sucralose tongue. mouth feel, ease of
sucralose, or No.: 16 handling, and
saccharin sodium) t: n.s. acceptance)
ODTs Preparation of drug- 10 DPH-Amberlite® The sample was The Euclidian One tablet n.s. Bitterness intensity [41]
ion exchange resin IRP-64 at the ratio diluted with PW to distances of the placed on the scale
complex of DPH- of 1:2 10% w/v DPH and complexes to tongue
Amberlite® IRP-64 evaluated by native DPH No.: 12
by spray drying electronic tongue should be large. t: immediately
method after tablet was
disintegrated
ODTs Addition of 10 ODTs containing n.s. n.s. One tablet n.s. Palatability scale [42]
sweeteners 50% of Starlac and placed on the (Taste, aftertaste,
(ammonium 10% of tongue. mouth feel, ease of
glycyrrhizinate) ammonium No.: 16 handling, and
glycyrrhizinate t: n.s. acceptance)
ODFs Preparation of DPH/ 5 ODFs containing The sample was The Euclidian n.s n.s. n.s. [43]
hydroxypropyl- 4.7% of DPH/ dissolved to the distances of the
β-cyclodextrin 41.4% of HP-β-CD, concentration of complexes to
complex (DPH/HP- 41.9% of HPMC, 0.05 mg/mL DPH native DPH
β-CD) by liquid- and 16.7% of and evaluated by should be large
liquid extraction glycerol electronic tongue
method
ODFs Preparation of DPH 5 ODFs containing n.s. n.s. Test method n.s. Palatability score [44]
loaded HPMC, HPC, HPMC: HPC at the was not (initial taste,
PVP-K90, or ratio of 5: 1 and specified. mouthfeel,
pullulan films by glycerin of 1.6% No.: 6 disintegration
solvent casting w/w t: n.s. time, flavor,
method aftertaste, and
overall
acceptability)
The score was
calculated to
bitterness index.

Note: M: test medium, V: volume of test medium, T: temperature, No.: number of volunteers, t: time point of evaluation, PW: purified water, n.s.: not specified.

et al. [37], taste-masked hydroxypropyl methylcellulose (HPMC) compatibility of sweeteners and flavors with other ingredients in the
based-ODFs were developed by adding aspartame (180- to 200-fold formulation, stability, toxicity, adverse drug reaction and safety should
sweeter than sucrose), sucralose (300- to 1000-fold sweeter than su­ also be considered [40].
crose), or saccharin sodium (300- to 600-folds sweeter than sucrose)
[37,38]. Since sucralose possesses the highest sweet intensity, the ODF 2.1.2. Application of physical barrier
with 4.67% received the highest acceptance score from volunteers. An alternative taste-masking approach is applying a physical barrier,
However, only a few amounts (less than 1%) are typically recom­ which is a highly effective option in comparison to the addition of
mended, since these sweeteners (aspartame, sucralose, and saccharin sweeteners and flavors. The barrier blocks the taste signal transmission
sodium) have an unpleasant aftertaste. In addition to the synthetic by minimizing the contact of bitter substances and receptors; thus, the
substance, ammonium glycyrrhizinate (AG), was used by the same bitterness could be masked. Taste-masking by incorporating DPH within
research group as sweeteners and herbal flavors for ODTs. The result polymers, such as HPMC, hydroxypropyl cellulose (HPC), polyvinyl
showed that the bitterness of 10-mg DPH was masked by around 6% of pyrrolidone-K90 (PVP-K90), or pullulan film was investigated by Han
AG, whose sweetness potency was 30- to 50-fold greater than that of et al. [44]. The solvent casting technique was used to prepare ODFs in
sucrose [39]. The human palatability test of these two studies also their study. Although the film’s palatability test showed good accept­
demonstrated that the bitterness of DPH was successfully suppressed. ability, it should be noted that the taste-masking effect might be con­
However, the use of a high concentration of sweeteners might be a nected to the synergistic effect of the film-forming agent and sucralose.
limitation of this approach due to the strong bitterness intensity and Based on the fact that the pH of saliva is reported to be 5.5–7.2 [45], a
hydrophilic nature of the DPH. Although special equipment or tech­ polymer with pH-dependent solubility has been studied. Since these
nology is not required for such techniques, a large number of sweeteners polymers are soluble under gastric pH with insoluble or swellable
might result in a rise in cost and processing difficulties. The properties under oral pH, the bitterness of active drugs could be masked

3
L. Sutthapitaksakul et al. Journal of Drug Delivery Science and Technology 63 (2021) 102549

without changing the drug release profile. According to the study of Yan taste evaluation for dosage form development is the design of a test
et al. [28], Eudragit® E, a methacrylate copolymer with pH-dependent method. However, there is no compendial method designed explicitly
solubility, and spray-drying of dried microspheres with a different for taste-masking dosage forms; only bitterness evaluation is suggested
ratio of DPH:polymer have been investigated. The in vitro drug release in in the British Pharmacopeia. Since the human taste perception varies
simulated saliva fluid (SSF) showed that drug release from microspheres depending on age, gender, and eating habits, the bitterness threshold
containing 1:2 ratio of DPH:polymer was 1.55% or 7.75 μg/mL within determination and panelist training are essential steps to ensure the
60 s. Compared with the bitterness threshold of DPH of 15–20 μg/mL, reliability and comparability of results. The bitterness threshold or
this ratio was considered to be a taste-masking formulation. However, it bitterness value is the lowest concentration of the test substance that can
should be noted that 1% of aspartame was also incorporated in ODTs. be perceived as bitter. The panelists training can be conducted by pre­
Therefore, the taste-masking effect might be connected to the synergistic senting pre-defined bitterness score solutions to the volunteers. Thus,
taste-masking effect of Eudragit® E and aspartame. the reliability of sample rating scores of the volunteers is enhanced. The
determination method is reported in details by Suzuki et al. [49] and
2.1.3. Preparation of drug-ion exchange resin complex Albertini et al. [50]. To avoid the limitation of human panelist testing,
Ion exchange resin is a high molecular weight polymer comprising of especially in the preclinical step when the toxicity of new chemical drug
anionic and cationic functional groups. The insoluble drug-resin com­ is not well elucidated, the taste sensing systems (electronic tongues) may
plex or resinate is formed through weak ionic bonding of each oppositely offer a promising approach for taste assessment [51].
charge substance. For taste masking purposes, the drug-resin complex
should not dissociate in the oral cavity. However, at gastric pH, resinate 2.2. Adverse events of DPH
would exchange their drug molecules with counter ions in the GI fluid.
Thus, taste-masking is achieved, and the actives can be absorbed The frequently reported adverse events of DPH are dose-dependent
through the GI tract properly. DPH-ion exchange resin complex was and include nausea, diarrhea, insomnia, muscle cramps, fatigue, vom­
prepared for bitterness taste-masking by ODTs. Amberlite® IRP-64, iting, and anorexia. Among these adverse events, GI adverse event
which is anion exchange resin, was used. Firstly, DPH-Amberlite® (GIAE) including nausea, diarrhea, and vomiting has been extensively
IRP-64 resinate was prepared by spray-drying. The in vitro taste-masking investigated among research groups. As the cholinergic enhancement
efficiency was tested using an electronic taste sensing system or elec­ activity of DPH stimulates the stomach and intestinal motility, gastric
tronic tongue, comparing the taste of the pure drug, ion exchange resin, acid secretion, peptic ulcers, or GI bleeding, or both have also been re­
physical mixture, and optimal resinate. The result showed that the ported in clinical trials and some case reports [52,53]. Thus, these events
Euclidian distance of the physical mixture is close to the pure drug. should be carefully monitored, especially in high-risk patients [10,11,
Alternatively, resinate showed further distance to the pure drug 54]. To alleviate such problems, several novel dosage forms have been
compared with the physical mixture, indicating bitter taste reduction of developed (Table 2). These studies were classified based on the
resinate. The panelist testing of optimal resinate also showed a good administration route of the dosage form intended to be administered
average bitterness index of 1.58, meaning that the bitterness was be­ through the oral, topical/transdermal, parenteral, and intranasal routes.
tween the threshold of bitter to slightly bitter [41].
3. Novel dosage forms and development technologies
2.1.4. Preparation of inclusion complex
Another taste-masking technique is to prepare an inclusion complex. 3.1. Dosage form intended to be administered orally
Among several complexation substances, cyclodextrin is the most
extensively used. The inclusion complex masks the bitter taste of the Clays, layer structured aluminum silicates with good acid absorption
drug by decreasing drug solubility or decreasing drug molecules’ contact capacity, were used to reduce gastric acid by proton absorption and
on taste buds on the tongue. Therefore, the taste signal transmission is control drug release profile. The smectite clays, including laponite XLG,
reduced or terminated. Such an approach was used to prepare DPH/ saponite, and montmorillonite, were studied to intercalate DPH inside
cyclodextrin complexation of ODFs. Hydroxypropyl-β-cyclodextrin (HP- their structure. Due to negative surface charge after hydration, these
β-CD) was applied to mask the bitterness of DPH by liquid-liquid solvent clays possess cation exchange capacity (CEC), which can be arranged in
extraction. After that, ODFs were prepared following the solvent casting descending order as montmorillonite, saponite, and laponite. Eudragit®
method with different film-forming polymers, including HPMC, pre­ E, a cationic polymer, was coated on the hybrid by spray drying to
gelatinized starch, polyethylene oxide, and low-substituted HPC. After replace the cation of DPH molecules through ion exchange reaction.
ODF evaluation, the optimized formulation contained 4.7% of DPH, Drug release of spray-dried hybrid within 180 min showed a fast release
41.4% of HP-β-CD, 41.9% of HPMC, and 16.7% of glycerol. The in vitro of all hybrids [55]. However, the reduction of GIAE was not evaluated in
taste-masking using electronic tongue showed that the Euclidian dis­ this study. Since the smectite clays used were modified to a new hybrid,
tance of the optimized ODFs is larger than others, indicating taste- new clays’ acid absorption capacity should be characterized to confirm
masking has been accomplished [43]. the main objectives of their study.
Apart from the bitter taste problem, age-related or disease-related Since DPH dose increase resulted in an increased GIAE, the incidence
swallowing difficulties may also affect the ability to swallow conven­ of this adverse event increased to approximately 2-fold while using a
tional solid dosage forms since DPH has been used in AD elderly pa­ maximum dose. As it is associated with the fluctuation of DPH plasma
tients. The situation gets worse when several oral drugs have to be concentration levels, the gradual dose titration within four to six weeks
swallow in a day [46]. The studies conducted among this group revealed is recommended to allow patients to adjust to the drug’s pharmacody­
that approximately 84%–93% of patients suffered from a swallowing namic effect. Also, it is hypothesized that GIAE is associated with a rapid
problem [47]. Due to this particular consideration, it is necessary for cholinergic system stimulation caused by rapid drug absorption. Co-
oral formulators to develop “easy-to-swallow” dosage forms, for administration with food can improve the problem by a time delay to
example, ODTs, ODFs (Table 1). For example, DPH-loaded orally dis­ maximum drug concentration [71,72]. To mitigate the GIAE issue,
integrating web with low molecular weight polyvinyl alcohol by elec­ DPB-loaded lyotropic liquid crystalline (LLC) mesophases were prepared
trospinning method was also prepared by Nagy et al. [48]. Since these to assure consistent drug bioavailability. LLC mesophases were prepared
dosage forms rapidly dissolve or disintegrate within seconds after by mixing glyceryl monooleate, DPB-oleic acid solution, and water at
placement on the tongue, a solution or suspension is formed, allowing room temperature. The texture, swelling behavior, and mucoadhesive
easy swallowing without water intake. properties of the formulation were evaluated. Results showed a
Also, the important factor in determining the reliability of the human controlled drug release pattern for over 24 h. The authors mentioned

4
L. Sutthapitaksakul et al. Journal of Drug Delivery Science and Technology 63 (2021) 102549

Table 2
The dosage forms developed to overcome GIAE due to DPH.
Dosage form Administration Dose Strategy to overcome Optimized formulation In vitro evaluation In vivo evaluation Reference
route GIAE of DPH method method

Microparticles Oral n.s. Preparation of DPH- All DPH-clay hybrids Dissolution testing with n.s. [55]
clay (laponite XLG, coated by Eudragit® paddle apparatus under
saponite, and E100 with different drug the paddle speed of 50
montmorillonite) release profile were rpm
hybrid coated with successfully prepared M: SGF (pH 1.2)
Eudragit® E100 by V: 1000 mL
spray drying method Time: 180 min
T: 36.5 ◦ C
Transdermal Transdermal n.s. Preparation of DPB or DPB formulation Skin permeation study Pharmacokinetic study [56]
patches application DPH in PG with or containing 10 mg of DP using Keshary-Chien using Spraque-Dawley
without 1–5% fatty acid with 1% oleic acid diffusion cells rats
which were occluded Test skin: hairless Test sample:
on the skin with mouse skin and human transdermal
parafilm cadaver skin application of DPB or
M: PBS (pH 7.4) DPH in PG (10 mg/mL)
V: 6 mL with or without 1%
T: 37 ◦ C fatty acid
Analyzed sample:
blood
Transdermal Transdermal 4% Preparation of DPH- All DPH-loaded WEDD® n.s. Pharmacokinetic study [25]
patches application loaded WEDD® patches patches with various using hairless skin rats
which propelled the current density gave Test sample: DPH-
actives across the rat different loaded WEDD®
skin by iontophoretic pharmacokinetic profiles patches and control
method without DPH
Analyzed sample:
blood
Transdermal films Transdermal 1% Preparation of DP- The films containing DP- Skin permeation study n.s. [57]
application loaded sodium alginate loaded sodium alginate using Franz diffusion
matrix film with DL- matrix film with 3% DL- cells
limonene as a limonene Test skin: pig skin
penetration enhancer M: PBS (pH 6.5)
using water casting V: 12 mL
method T: 37 ± 1 ◦ C
Nanosuspension Intranasal 5 mg Preparation of DPB- The nanosuspension with DPB of 5 mg and the 1. Estimation of DPB [58]
loaded chitosan the mean size of 150 to nanosuspension content using Sprague-
nanosuspension by 100 nm with PDI of containing 5 mg Dawley rats
ionic-crosslinking 0.341, and EE of equivalent dose of DPB Test sample: intranasal
method 92–96%. were tested in dialysis administration of
bag under the stirring nanosuspension and
speed of 100 rpm. DPB suspension
M: n.s. through nostrils of
V: n.s. rats.
Time: n.s. Analyzed sample:
T: 37 ◦ C brain tissue
2. In vivo safety study
using Sprague-Dawley
rats
Test sample: intranasal
administration of
nanosuspension and
normal saline through
nostrils of rats
Analyzed sample:
blood and vital organs
Microspheres Subcutaneous n.s. Preparation of The microspheres Two milligrams of Pharmacokinetic study [59]
injection sustained release DPB- containing 20 mg of microspheres were using two groups of
loaded PLGA PLGA, 3% of PVA and shaken by horizontal rats (n = 4 per group).
microspheres by oil-in- water phase with 2% of shaker under the speed Test sample: IV
water emulsion solvent PVA for emulsion of 60 rpm. Then, the injection of DPH
evaporation method dilution of 16 mL supernatant was aqueous solution and
Size: mean diameter of analyzed at subcutaneous injection
29.4 ± 17.1 μm and a predetermined time. of DPB-loaded
span value of 0.85 M: PBS (pH 7.4) microspheres
DL: 15.92 ± 0.31% V: 6 mL suspension
EE: 78.79 ± 2.56% Time: up to 10 days Analyzed sample:
T: 37 ± 0.5 ◦ C blood
Lyotropic liquid Oral 16.67 g/g of Preparation of DPB- The formulation The in vitro drug release n.s. [30]
crystalline formulation loaded LLC mesophase containing DPB of 16.67 studies using USP II
(LLC) by direct mixing g/g, glyceryl monooleate apparatus with the
mesophase/ technique of 50 or 60%, oleic acid mixture of 0.1 N
matrix of 25%, and water of 25 hydrochloric acid and
or 15% polysorbate 80
(continued on next page)

5
L. Sutthapitaksakul et al. Journal of Drug Delivery Science and Technology 63 (2021) 102549

Table 2 (continued )
Dosage form Administration Dose Strategy to overcome Optimized formulation In vitro evaluation In vivo evaluation Reference
route GIAE of DPH method method

Tip-loaded Transdermal n.s. Preparation of DPH- Tip-loaded microneedle The amount of DPH Pharmacokinetic study [60]
dissolving application loaded tips of containing DPH-hydroxy delivered into the skin using rats
microneedles dissolving propyl methyl cellulose- analysis using high Test sample: DPH-
microneedles by a ethanol and water performance liquid loaded tips of
micromolding method mixture at the ratio of 80: chromatography microneedle and oral
20 DPH solution
Analyzed sample:
blood
Microneedle films Transdermal n.s. Preparation of DPH- DPH-loaded microneedle In vitro drug delivery In vivo drug delivery [61]
application loaded microneedle films containing study employing Franz study using Sprague-
films by solvent casting Gantrez® AN139 of 15%, diffusion cell Dawley rats
technique and tripropylene glycol Test skin: porcine skin Test sample: DPH-
methyl ether of 7.5% M: PBS (pH 7.4) loaded film with a drug
T: 37 ± 1 ◦ C loading of either 2.5
mg/kg or 5.0 mg/kg
Analyzed sample:
blood
Solid lipid Intranasal DPB to lipid Preparation of DPB- DPB to lipid ratio of 1: 5, In vitro release and In vivo [62]
nanoparticles ratio of 1: 4 loaded solid lipid surfactant concentration release kinetic studies pharmacokinetic study
to 1: 6 nanoparticles by of 2.25% w/v, and in dialysis bag using male albino
solvent emulsification stirring time of 2.75 h M: PBS (pH 7.4) Wistar rats
diffusion technique V: 100 mL Test sample: optimized
Time: up to 24 h DPB solid lipid
T: 37 ± 0.5 ◦ C nanoparticles and DPB
solution
Analyzed sample:
blood, brain, and other
organs
Liposome Intranasal n.s Preparation of DPB- DPB-loaded liposome In vitro drug release in In vivo [63]
loaded liposome by thin containing 1,2-distearyl- dialysis sac pharmacokinetic study
layer hydration method sn-glycero-3-phodpho­ M: simulated nasal using male Wistar rats
choline, cholesterol, and fluid (pH 6.2–6.8) Test sample: DPB-
polyethylene glycol at V: 100 mL loaded intranasal
the ratio of 1: 2: 0.5 Time: up to 360 min liposomes (1 mg/kg)
T: 37 ◦ C Analyzed sample:
blood
Microspheres Subcutaneous n.s. Preparation of The microspheres Four milligrams of Drug release study [64]
injection sustained release DPB- containing 1: 5 of DPB: microspheres were using 42 Kunming
loaded PLGA PLGA (11.83% PLGA), shaken under the speed mice
microspheres by oil-in- 1.005% of PVA, and of 100 rpm. Then, the Test sample:
water emulsion solvent water-organic phase supernatant was subcutaneous injection
evaporation method volume ratio of 13.24 analyzed at of DPB-loaded
Size: mean diameter of predetermined time. microspheres
71.75 ± 0.35 μm and a M: PBS (pH 7.4) suspension
size distribution of 0.825 V: 8 mL Analyzed sample:
± 0.007 Time: up to 42 days residual microspheres
DL: 14.48 ± 0.41% T: 37 ◦ C after peeling skin off
Nanostructured Transdermal 0.25 or Preparation of DPB- The NLCs containing In vitro skin permeation n.s. [65]
lipid carrier- application 0.50% of loaded NLCs by hot DPB, stearic acid of study employing Franz
based gels formulation microemulsion 1.25%, oleic acid of diffusion cell
technique 0.75%, lecithin of 1.00%, Test skin: porcine ear
and sodium skin
taurodeoxycholate of M: PBS (pH 5.0)
0.25% T: 37 ◦ C
Nanoemulsions Intranasal 6.25 mg/mL Preparation of DPB- DPB-loaded In vitro release study Histological analysis [66]
loaded nanoemulsions nanoemulsions with using Franz diffusion using pigs
and DPB-loaded Pluronic® F-1227 cell Test sample: intranasal
nanoemulsions with Test tissue: dialysis isopropyl alcohol,
Pluronic F-1227 membrane DPB-loaded
M: methanol: nanoemulsions and
Transcutol-P DPB-loaded
T: 37 ± 0.5 ◦ C nanoemulsions with
Ex vivo permeation Pluronic® F-1227
study Analyzed sample:
Test tissue: porcine nasal mucosa
nasal mucosa
M: Transcutol-P: water
Ionic liquid (ILs) Transdermal n.s. Preparation of ILs form Donepezil- Skin PAMPA and BBB n.s. [67]
form of DPH application of DPH with different docosahexaenoic acid PAMPA
types of counterion by and donepezil-α linolenic Test skin: artificial skin
metathesis reaction acid M: standard Britton-
formulation Robinson buffer (pH
7.4)
T: 32 ◦ C
Molecular docking
(continued on next page)

6
L. Sutthapitaksakul et al. Journal of Drug Delivery Science and Technology 63 (2021) 102549

Table 2 (continued )
Dosage form Administration Dose Strategy to overcome Optimized formulation In vitro evaluation In vivo evaluation Reference
route GIAE of DPH method method

study
In vitro skin permeation
study
Microemulsions Intranasal n.s. Preparation of DPH- DPH-butter oil and DPH- Ex vivo nasal diffusion In vivo [68]
butter oil and DPH- omega-3 fatty acid-rich study pharmacokinetic study
omega-3 fatty acid-rich fish oil microemulsions Test tissue: goat nasal using Sprague-Dawley
fish oil microemulsions tissue rats
by physical mixing M: PBS (pH 6.4) Test sample: DPH
method In vitro cell solution, DPH-
permeability study microemulsion, DPH-
Test cell: mouse butter oil and DPH-
cerebral microvascular omega-3 fatty acid-
endothelial cell line rich fish oil
(bEnd3) microemulsion
Analyzed sample:
blood, and brain
Solid lipid Intranasal n.s. Preparation of DPB- DPB-loaded solid lipid In vitro release study n.s. [69]
nanoparticles loaded solid lipid nanoparticles labeled using the dialysis
nanoparticles labeled with rhodamine B and membrane method
with rhodamine B and targeted with M: PBS (pH 7.4)
targeted with apolipoprotein E containing 0.1% Tween
apolipoprotein E 80
V: 50 mL
Time: up to 24 h
T: 37 ◦ C
Cellular uptake study
Test cell: rat brain
endothelial cell line,
human cerebral
microvascular
endothelial cell line,
and human
neuroblastoma cell line
BBB co-culture model
and permeability study
Test cell: primary rat
brain endothelial cells,
pericytes, and
astrocytes
Nanofibers and Oral n.s. Preparation of chitosan Nanofibers containing Dissolution testing with In vivo bioavailability [70]
thin film blended with chitosan/hyperbranched basket apparatus study using male
hyperbranched chitosan blend M: PBS (pH 6.8) Wistar rats
polyester and V: 600 mL Test sample: orally
hyperbranched Time: 60 min administration of thin
chitosan T: 37 ± 0.5 ◦ C film and nanofiber
sheet
Analyzed sample:
blood

Note: M: test medium, V: volume of test medium, T: temperature, DL: drug loading, EE: encapsulation efficiency, n.s.: not specified.

that these formulations could provide a controlled DPB release for oral absorption and permeation. The molecular weight of DPH is 425.96
administration, which will be beneficial to improve GIAE [30]. g/mol with log P of 4.27 [25]. The number of hydrogen bond acceptors
and donors are four and zero, respectively. Although none of the factors
break the rule, the molecular weight and log P of DPH are approximate
3.2. Dosage form intended to administer through topical/transdermal to these factors’ margin (molecular weight <500 g/mol and log P < 5).
route Therefore, an effective delivery device is necessary to achieve a desired
therapeutic effect.
The topical/transdermal route offers good advantages over others
since it is a non-invasive administration route. Also, it also provides 3.2.1. Transdermal patches/films
several benefits, including first-pass metabolism circumvention, easy-to- Recently, donepezil transdermal patches were developed by Icure
use, and more convenience for patients to self-administer. Pharmaceutical Inc. (South Korea) and tested in phase III clinical trial
It also provided constant plasma level of drug and minimized the with 399 participants. This study compared the efficacy and safety of 25-
GIAE of DPH [73]. In addition, the transdermal route has got a lot of cm2 and 50-cm2 transdermal patches to 5-mg and 10-mg Aricept®
attention from the pharmaceutical industry. Teikoku Pharma USA, Inc. tablets in mild to moderate AD patients [75].
developed Aricept® transdermal patches for once-weekly administra­ DPB or DPH-loaded transdermal patches were prepared based on
tion under a licensing agreement with Eisai Co., Ltd., and then submitted clinical trial results of rivastigmine transdermal patches, which sug­
to US FDA in 2010. The application was, however, withdrawn from the gested that the patches’ efficacy is similar to that of the capsules [76].
US FDA in 2012 [74]. Although this route might be an excellent choice On the other hand, GIAE was found to be lower. To improve drug skin
for DPH administration, it is known that only limited amounts of drugs permeability, the transdermal patches were developed using different
are administrable via this route. Following the rule of five, this corre­ carbon length fatty acids as a skin penetration enhancer. The
lates with drug physicochemical properties to determine high

7
L. Sutthapitaksakul et al. Journal of Drug Delivery Science and Technology 63 (2021) 102549

formulation was prepared by dissolving various DPB and DPH dosages in vinyl ether and maleic anhydride (PMVE/MAH), copolymers of methyl
propylene glycol (PG) with or without fatty acid. The in vitro permeation vinyl ether and maleic acid (PMVE/MA), or PVP and plasticizers were
study was carried out on diffusion cells using hairless mouse skin, blended to prepare a film using a solvent casting method. The in vitro
showing that the permeation rate of DPB and DPH was most enhanced drug release study was conducted using Franz diffusion cells and
by stearic acid (C18) and palmitic acid (C16), respectively. The in vitro demonstrated that the permeation of DPH was enhanced. Drug delivery
permeability result of DPB showed that the permeation rate decreased of these two formulations was further analyzed in vivo with
with an increasing number of double bonds in unsaturated fatty acids. Sprague-Dawley rats. After blood sample analysis, the results showed
Oleic acid with one double bond (C18:1) was the most appropriate that DPH levels detected in a film containing 5.0 mg/kg dose were
enhancer for DPB as the permeation rate increased by approximately higher than that of 2.5 mg/kg. It continuously increased over 24 h. The
19-fold. In contrast to DPH, the results showed that the permeation rate authors suggested that these dosage forms may be used to deliver hy­
was increased up to 35-fold while unsaturated fatty acid with only one drophilic drugs since such hydrogel-forming microneedle could create
double bond (palmitoleic acid, C16:1) was incorporated. The in vivo an aqueous environment facilitating drug release [61].
pharmacokinetic study in rats showed that pharmacokinetic parameters
of the DPB with 1% oleic acid and DPH with fatty acids were higher than 3.2.2. Iontophoretic delivery
those without fatty acids. The steady-state concentration of all formu­ Iontophoretic transdermal delivery uses a low electrical current to
lations was maintained for approximately 48 h. Therefore, the authors drive a charged drug molecule into the skin. This electrical current
suggested that this transdermal drug delivery system with skin en­ would interfere with skin structures, facilitating drug delivery based on
hancers might be an appropriate product for clinical use [56]. The skin two main delivery mechanisms, including electromigration and elec­
permeation enhancer was also employed to prepare DPH-loaded sodium troosmosis. Electromigration is the term defining the repulsion of the
alginate matrix type film by the water casting method. The effect of anode to positive charge ions and cathode to negative charge ions. On
DL-limonene concentration as a skin penetration enhancer was investi­ the other hand, electroosmosis is the movement of water caused by
gated. The in vitro skin permeation study was carried out with Franz convection [79,80]. According to the study of Salujaand et al. [25], DPH
diffusion cells using pig skin. The results showed that the amount of delivery by iontophoretic method was investigated. The gel formulation
permeated DPH was enhanced by 2.82-fold while DL-limonene concen­ containing DPH was injected into absorbent pads and attached to
tration increased from 1% to 3% [57]. However, there was no significant wearable electronic drug delivery (WEDD) patches, an iontophoretic
difference in enhancing effect than of DL-limonene (5%). delivery platform built with active electrodes and connected to a power
In another study, the skin permeability of DPH was improved by supply. After that, the active drug was propelled across abdominal re­
transforming ionic liquid (ILs). ILs improve permeability property by gions of rat skin by voltage-direct current. Blood was collected over 72 h
disrupting cell membrane integrity and fluidization, thus facilitating and analyzed. The transdermal patches’ pharmacokinetic profile
drug delivery across or into skin [77,78]. ILs were prepared by reacting showed that the plasma levels increased with increasing current density
DPH with anionic counter ions, including docusate sodium, oleate so­ [25]. However, the ability of the patches to overcome the cholinergic
dium, linoleate sodium, ibuprofen sodium, α-linolenic sodium, and do­ side effect issues, which the authors raised in the introduction, were not
cosahexaenoic sodium. The skin parallel artificial membrane evaluated.
permeability assays (skin PAMPA) results during the first 6 h showed
that permeation percentage of donepezil-docosahexaenoic acid and 3.2.3. Gels
donepezil-α linolenic acid ILs was higher than that of donepezil. How­ DPB-loaded nanostructured lipid carrier (NLC)-based gel was pre­
ever, these permeation percentage differences were lower in 12 h of pared to enhance drug skin permeability and overcome the adverse ef­
testing due to steady-state flux. The blood-brain barrier (BBB) parallel fect of donepezil. Hot microemulsion technique was used to prepare NLC
artificial membrane permeability assay (BBB PAMPA) was also by employing stearic acid as a solid lipid, oleic acid as a liquid lipid,
employed to investigate the effect of IL formation on BBB permeation. lecithin as a surfactant, and sodium taurodeoxycholate as a co-
The results showed that IL formulation had the ability to cross the BBB. surfactant. The authors also mentioned that lecithin was used as a
After that, drug-in-adhesive transdermal patches of ILs were prepared. cutaneous absorption enhancer as it can interact with skin lipid. The size
The result showed that donepezil-docosahexaenoic acid and donepezil-α of NLCs was around 200 nm. In vitro drug release test employing Franz
linolenic acid has higher permeability capacity than native donepezil. diffusion cell and cellulose acetate dialysis membrane (12–14 kDa cut
Therefore, the authors suggested that ILs may be an alternative trans­ off) of DPB-loaded NLC gels showed an initial burst release, followed by
dermal way for effective delivery of DPH [67]. sustained release of 50% over 48 h.
To enhance skin permeability by circumventing the stratum cor­ In contrast to free drug gels, a complete drug release of around 100%
neum, which is a permeation barrier, DPH was loaded in the tips of was obtained within 24 h. In vitro, skin permeation study using porcine
dissolving microneedles prepared by micromolding to overcome its low ear skin with Franz diffusion cell showed a lower permeated percentage
skin permeability and improve drug loading. DPH was added to than drug release test. Cumulative DPB permeated over 40% was ob­
hydroxypropyl methyl cellulose solution at different concentrations. The tained from 0.50% of the DPB-loaded NLCs while that of 0.25% of the
tip was prepared by blending DPH-hydroxypropyl methylcellulose- DPB-loaded NLCs was around 15% within 72 h of the test. The authors
ethanol and water, while the needle base was prepared by carboxy concluded that the skin permeation enhancing effect might contribute to
methylcellulose-water. The insertion test was conducted using porcine the increased contact with the skin, occlusive effect, and sustained
skin and three arrays of 100 microneedles with different amounts of release property of the formulation [65]. However, it should be noted
DPH. The pharmacokinetic results showed that a high drug loading that the skin permeation result of free drug gels was not shown in the
could be encapsulated in the tips. DPH delivered amount was over 95% study.
within 5 min of insertion while the tips were fully dissolved within 15
min. Also, the Cmax of tip-loaded microneedle was four times higher than 3.3. Dosage form intended for administration via injection
that of the oral administration. The authors suggested that their tip-
loaded microneedle may be a transdermal alternative to deliver DPH Some researchers prefer the injection method to deliver DPH
with good patient acceptance [60]. Another example also produced DPB directly. Based on the study of Guo et al. [59] and Fang et al. [64],
and DPH-loaded hydrogel-forming microneedle films to enhance drug injectable DPH-loaded poly(d,l-lactic-co-glycolic acid) (PLGA) micro­
delivery across the skin. Gantrez® S-97, polyethylene glycol, and spheres have been prepared using the emulsion solvent evaporation
Na2CO3 were blended to fabricate microneedle arrays by the molding technique. PLGA, a biocompatible and biodegradable polymer approved
method. DPB or DPH, Gantrez® AN 139 or S-97, copolymers of methyl by the FDA to be used as a drug delivery system, was used as a sustained

8
L. Sutthapitaksakul et al. Journal of Drug Delivery Science and Technology 63 (2021) 102549

drug release to overcome GIAE and compliance problems. DPB-loaded chitosan nanosuspension was prepared for intranasal
Guo et al. [59] prepared oil-in-water emulsions using dichloro­ administration by Bhavna et al. [58], to mitigate GI side effects and
methane as a solvent or oil phase. To enhance drug loading of the mi­ prolong drug action. The nanosuspension was prepared by the
crospheres, DPH was modified to DPB, which has low water solubility ionic-crosslinking method of DPB and chitosan; tripolyphosphate was
before encapsulation. After that, it was mixed with PLGA to produce the used as a crosslinker. The in vitro drug release test showed an
oil phase. This phase was then emulsified into a water phase containing improvement of cumulative drug release in comparison with conven­
different concentrations of polyvinyl alcohol. After that, the oil-in-water tional suspension. Similarly, in vivo drug content determination of DPB
emulsion was centrifuged and dried to obtain microspheres. The in vitro nanosuspension showed a higher drug concentration both in the plasma
drug release profile in PBS showed a sustained cumulative drug release and brain as compared to the intranasal solution. Moreover, the safety
of 88.44% over 10 days. The pharmacokinetics study in rats showed that test showed no toxicity in all rats, as there was no mortality, body
the DPB concentration peaked in plasma within a day after a single dose weight, and hematological changes. Therefore, the authors concluded
of subcutaneous administration, and drug levels were detected up to 15 that the intranasal DPB-loaded nanosuspension might be used as a novel
days. alternative route to deliver DPB to the brain [58].
Moreover, the authors also reported a good in vitro-in vivo release The DPB-loaded liposomal formulation was developed by Al Asmari
profile correlation of the microspheres. Therefore, they concluded that et al. [63], aiming to improve the bioavailability of intranasal formu­
the microspheres developed can be a sustained release dosage form for lation. The liposomes were prepared by thin-film hydration method
AD [59]. Since the modification of the salt form of DPH might affect such using cholesterol, polyethylene glycol and 1,2-distearyl-sn-glycero-3-­
a drug’s safety and efficacy, it should be noted that these issues are phosphocholine. In comparison to oral and intranasal administration
considered re-evaluated. In another study, sustained-release micro­ of DPB, the intranasal DPB-loaded liposomes resulted in a major
spheres were also prepared by a similar technique. DPB was also used as improvement in pharmacokinetic parameters. Under a microscope,
an active. However, the design of an experimental approach was used to histopathological examination of formed liposomes revealed no major
find the optimal formulation. Drug release kinetics of the microspheres changes in tissue after intranasal administration, suggesting the safety of
was also investigated. A pharmacokinetic study demonstrated that the the formulation [63].
microspheres successfully sustained drug release of about 90% up to 42 To improve the permeation across BBB, Khunt et al. [68] used a
days for in vitro testing, whereas only 21 days for in vivo testing in permeation enhancer, i.e., butter oil and omega-3 fatty acid-rich fish oil,
Kunming mice. The result showed that erosion was the main mechanism to prepare DPH microemulsions for intranasal administration. The ex
affecting the drug release, with the rate of erosion in vivo being faster vivo nasal diffusion study using Franz diffusion cells with goat nasal
than that in vitro. The authors hypothesized that this effect contributed tissue of DPH-butter oil and DPH-omega-3 fatty acid-rich fish oil
to some physiological reactions that might enhance water absorption or microemulsions showed an improved drug diffusion over DPH solution
accelerate polymer degradation [64]. Although GIAE was mentioned as and DPH microemulsion. The in vitro cell permeability study using cell
a primary objective of these studies, an effort to overcome the problem line of these two formulations showed a decrease of transepithelial
was not directly evaluated. Moreover, despite patient compliance being electrical resistance, indicating the permeation change. The drug extent
mentioned as a main aim, the authors administered the dosage form after intranasal administration of DPH-butter oil and DPH-omega-3 fatty
through subcutaneous injection, an invasive method. So, the compliance acid-rich fish oil microemulsion was higher than the control samples in
of developed dosage might be questionable. an in vivo pharmacokinetic study in rat brain, suggesting a promising use
of these oils as a nose-brain permeation enhancer [68].
3.4. Dosage form intended to be administered through intranasal route Apart from microemulsions, DPB nanoemulsions and DPB-Pluronic®
F-127 nanoemulsions were developed by Espinoza et al. [66]. Pluronic®
According to previous studies, it is preferred to deliver DPH orally F-127 nanoemulsions had significantly higher drug amount and pre­
and transdermally, which are peripheral administration routes. Drug diction parameters than DPB nanoemulsions, according to findings of an
molecules are supposed to be delivered to the brain through the blood ex vivo permeation study. It was suggested that using a nanoemulsion
circulation. However, this route’s efficacy is limited because of the BBB, with a permeation enhancer to deliver DPB through the BBB might be a
which prevents hazardous substances from entering the central nervous promising alternative.
system. Briefly, a low brain permeability and consequent low drug There has been an increased interest in the production of solid lipid
concentration in the brain are the main limitations of this route. nanoparticles for nose-to-brain drug delivery in recent years. Solvent
Compared to intranasal, drug molecules are mainly delivered through emulsification diffusion was used to make solid lipid nanoparticles of
the neuronal route, which is a direct nasal–brain route. The nasal cavity glyceryl monostearate blended with polysorbate 80 and poloxamer 188.
anatomy can be divided into three cavities: the vestibular region, res­ The effect of processing parameters and optimization was studied by
piratory region, and olfactory region. Among these regions, the respi­ Box-Behnken design. As compared to intravenous DPB solution and
ratory region covers the largest area of the nasal cavity. After drug intranasal DPB solution in pharmacokinetic and biodistribution studies
molecules pass into the nasal cavity, it will get through the mucociliary in rats, the optimized nanoparticles showed an increase in AUC of
clearance of the vestibular region, followed by enzymatic degradation approximately 2 folds [62]. Topal et al. [83] have also developed
and short retention time. These barriers might be an important challenge DPB-loaded solid lipid nanoparticles by homogenization-sonication
for the formulator to develop drug delivery systems to overcome such method. The particles were labeled with rhodamine B and targeted
systems. After passing through the vestibular region, drug molecules with apolipoprotein E, a ligand of the lipoprotein receptor in the BBB, to
enter the posterior region to contact the respiratory and olfactory re­ ensure that DPB was delivered effectively. Apolipoprotein E increased
gions. The respiratory region is highly supplied by blood vessels and DPB cellular uptake in a cell line, according to the findings. Further­
trigeminal sensory neurons, facilitating high-extent drug transport to the more, in the co-culture BBB model, permeation was increased [69]. It
pons and cerebrum. However, only a low drug extent is delivered to the was discovered that combining solid lipid nanoparticles with a targeting
olfactory and frontal brain. In addition to the major pathway, fewer ligand may be a promising technique for overcoming the BBB’s
amounts of drugs are delivered through the BBB. Compared with others, limitations.
the nose–brain delivery route offers several advantages, including pa­ Some studies prefer to use DPB as an active instead of DPH, espe­
tient convenience, BBB circumvention, first-pass metabolism avoidance, cially for lipid-based drug delivery systems, such as LLC, patches,
and quicker attainment of therapeutic drug level [81–83]. The microneedles, NLC, injectable microspheres, and intranasal nano­
nanotechnology-based drug delivery system may be a promising suspension. Since DPB is a lipophilic form, their compatibility with lipid
approach to overcome the limitations of BBB. excipients is more than that of DPH. However, it should be noted that the

9
L. Sutthapitaksakul et al. Journal of Drug Delivery Science and Technology 63 (2021) 102549

therapeutic efficacy and toxicity of DPB should be clearly elucidated. [10] H. Sugimoto, H. Ogura, Y. Arai, Y. Iimura, Y. Yamanishi, Research and
development of donepezil hydrochloride, a new type of acetylcholinesterase
inhibitor, Jpn. J. Pharmacol. 89 (2002) 7–20, https://doi.org/10.1254/jjp.89.7.
4. Conclusion and future prospects [11] Eisai Co Ltd, Aricept (donepezil hydrochloride) label. http://www.accessdata.fda.
gov/drugsatfda_docs/label/2012/020690s035, 021720s008, 022568s005lbl.pdf,
Recently, there is no effective drug for AD; most available drugs are 2012. (Accessed 10 December 2020).
[12] A.H. Barfejani, M. Jafarvand, S.M. Seyedsaadat, R.T. Rasekhi, Donepezil in the
symptomatic. Current research is focused on the clinical role of DPH as a treatment of ischemic stroke: review and future perspective, Life Sci. 263 (2020)
disease-modifying agent in both preclinical and clinical studies. Due to 118575, https://doi.org/10.1016/j.lfs.2020.118575.
AD’s extremely complicated and ambiguous pathogenesis, there are still [13] J. Cummings, T. Lai, S. Hemrungrojn, E. Mohandas, S.Y. Kim, G. Nair, Role of
donepezil in the management of neuropsychiatric symptoms in Alzheimer’ s
some controversies about a suitable model selection. Among several disease and dementia with Lewy bodies, CNS Neurosci, Ther 22 (2016) 159–166,
studies, the bitter taste is a significant problem for conventional oral https://doi.org/10.1111/cns.12484.
dosage forms, which is an original administration route of DPH. Several [14] G. Román, S.J. Rogers, Donepezil: a clinical review of current and emerging
indications, Expert Opin, Pharmacother 5 (2004) 161–180, https://doi.org/
approaches have been employed for taste-masking, including the addi­ 10.1517/14656566.5.1.161.
tion of sweeteners and flavors, application of physical barriers, prepa­ [15] B. Seltzer, Donepezil: an update, expert opin, Pharmacother 8 (2007) 1011–1023,
ration of ion exchange resins, and inclusion complexes. Future research https://doi.org/10.1517/14656566.8.7.1011.
[16] N. Livingstone, J. Hanratty, R. Mcshane, G. Macdonald, Pharmacological
should emphasize a combinatorial approach, which may prove to be an interventions for cognitive decline in people with Down syndrome (Review),
efficient taste-masking strategy. Due to the fluctuation of plasma DPH Cochrane Database Syst. Rev. 10 (2015), https://doi.org/10.1002/14651858.
concentration levels, GIAE was frequently reported as an adverse event. CD011546.pub2. CD011546.
[17] M. Mohan, P.K. Carpenter, C. Bennett, Donepezil for dementia in people with
To alleviate the adverse effects, studies should focus on the development
Down syndrome, Cochrane Database Syst. Rev. 1 (2009), https://doi.org/10.1002/
of controlled drug release formulations for maintaining a constant level 14651858.CD007178.pub2. CD007178.
of drug concentration in the plasma. The dosage forms intended to be [18] B.H. Ridha, S. Crutch, D. Cutler, C. Frost, W. Knight, S. Barker, N. Epie, E.
administered through oral, topical/transdermal, injection, and intra­ K. Warrington, R. Kukkastenvehmas, J. Douglas, M.N. Rossor, A double-blind
placebo-controlled cross-over clinical trial of donepezil in Posterior cortical
nasal routes have been developed to overcome such adverse events and atrophy due to underlying Alzheimer’s disease: DONIPAD study,, Alzheimer’s Res.
control the drug release. Transdermal drug delivery systems might be an Ther. 10 (2018), https://doi.org/10.1186/s13195-018-0363-1.
excellent alternative choice. Recent studies show that intranasal drug [19] D.P. Devanand, G.H. Pelton, K. D’Antonio, A. Ciarleglio, J. Scodes, H. Andrews,
J. Lunsford, J.L. Beyer, J.R. Petrella, J. Sneed, M. Ciovacco, P.M. Doraiswamy,
delivery systems’ potential as it directly delivered DPH to the brain. A Donepezil treatment in patients with depression and cognitive impairment on
key consideration for successful delivery includes efficient delivery to stable antidepressant treatment: a randomized controlled trial, Am, J. Geriatr.
the BBB and therapeutic concentration achievement. Since a compre­ Psychiatr. 26 (2018) 1050–1060, https://doi.org/10.1016/j.jagp.2018.05.008.
[20] J. Zhou, Y. Fu, X.C. Tang, Huperzine A and donepezil protect rat
hensive knowledge of AD target factors is essential for designing drug pheochromocytoma cells against oxygen–glucose deprivation, Neurosci. Lett. 306
delivery systems for clinical use, most studies at this stage are preclin­ (2001) 53–56, https://doi.org/10.1016/S0304-3940(01)01855-9.
ical. The mechanism of drug transfer from the nose to the brain must be [21] E.I. Solntseva, N.A. Kapai, O.V. Popova, P.D. Rogozin, V.G. Skrebitsky, The
involvement of sigma1 receptors in donepezil-induced rescue of hippocampal LTP
clarified to facilitate drug delivery system development to target a impaired by beta-amyloid peptide, Brain Res, Bull. (Arch. Am. Art) 106 (2014)
suitable region of the nasal cavity. Further functionalization with li­ 56–61, https://doi.org/10.1016/j.brainresbull.2014.06.002.
gands targeting suitable regions on the BBB might improve DPH brain [22] M.Y. Noh, S.H. Koh, S.M. Kim, T. Maurice, S.K. Ku, S.H. Kim, Neuroprotective
effects of donepezil against Aβ42-induced neuronal toxicity are mediated through
entry.
not only enhancing PP2A activity but also regulating GSK-3β and nAChRs activity,
J. Neurochem. 127 (2013) 562–574, https://doi.org/10.1111/jnc.12319.
Declaration of competing interest [23] Y. Takada-Takatori, S. Nakagawa, R. Kimata, Y. Nao, Y. Mizukawa, T. Urushidani,
Y. Izumi, A. Akaike, K. Tsuchida, T. Kume, Donepezil modulates amyloid precursor
protein endocytosis and reduction by up-regulation of SNX33 expression in
None. primary cortical neurons, Sci. Rep. 9 (2019), https://doi.org/10.1038/s41598-
019-47462-4.
Acknowledgements [24] H. Shen, T. Kihara, H. Hongo, X. Wu, W.R. Kem, S. Shimohama, A. Akaike,
T. Niidome, H. Sugimoto, Neuroprotection by donepezil against glutamate
excitotoxicity involves stimulation of alpha7 nicotinic receptors and
Financial funding from the Ministry of Higher Education, Science, internalization of NMDA receptors, Br. J. Pharmacol. 161 (2010) 127–139, https://
Research and Innovation (Thailand) through Reinventing University doi.org/10.1111/j.1476-5381.2010.00894.x.
[25] S. Saluja, P.C. Kasha, J. Paturi, C. Anderson, R. Morris, A.K. Banga, A novel
System Program (fiscal year 2021) is gratefully acknowledged. electronic skin patch for delivery and pharmacokinetic evaluation of donepezil
following transdermal iontophoresis, Int, J. Pharm 453 (2013) 395–399, https://
References doi.org/10.1016/j.ijpharm.2013.05.029.
[26] Y.A. Asiri, G.A.E. Mostafa, Donepezil, in: profiles of drug substances. Excipients
and Related Methodology, first ed., Elsevier Inc., New York, 2010, pp. 117–150,
[1] Alzheimer’s association, 2020 Alzheimer’s disease facts and figures, Alzheimer’s
https://doi.org/10.1016/S1871-vol. 5125(10, 35003–35005.
Dementia 16 (2020) 391–460, https://doi.org/10.1002/alz.12068.
[27] A. Moffat, M. Osseltion, P. Widdop (Eds.), Clarke’s Analysis and Poisons, fourth
[2] C. Patterson, World Alzheimer Report 2018: the State of the Art of Dementia
ed., Pharmaceutical Press, London, 2011.
Research - New Frontiers, 2018.
[28] Y.D. Yan, J.S. Woo, J.H. Kang, C.S. Yong, H.-G. Choi, Preparation and evaluation of
[3] M. Zvěřová, Clinical aspects of Alzheimer’s disease, Clin. Biochem. 72 (2019) 3–6,
taste-masked donepezil hydrochloride orally disintegrating tablets, Biol. Pharm.
https://doi.org/10.1016/j.clinbiochem.2019.04.015.
Bull. 33 (2010) 1364–1370, https://doi.org/10.1248/bpb.33.1364.
[4] M.S. Uddin, M.T. Kabir, A. Al Mamun, G.E. Barreto, M. Rashid, A. Perveen, G.
[29] R. Zainuddin, A. Kulkarni, H. Chavan, G. Patil, Z. Zaheer, Taste masking of
M. Ashraf, Pharmacological approaches to mitigate neuroinflammation in
donepezil hydrochloride using different ion exchange resins - a comparative study,
Alzheimer’s disease, Int. Immunopharm. 84 (2020), 106479, https://doi.org/
Indones. J. Pharm. 24 (2013) 107–115.
10.1016/j.intimp.2020.106479.
[30] A.L.M. Ruela, F.C. Carvalho, G.R. Pereira, Exploring the phase behavior of
[5] Q. Li, S. He, Y. Chen, F. Feng, W. Qu, H. Sun, Donepezil-based multi-functional
monoolein/oleic acid/water systems for enhanced donepezil administration for
cholinesterase inhibitors for treatment of Alzheimer’s disease, Eur. J. Med. Chem.
Alzheimer disease treatment, J. Pharm. Sci. 105 (2016) 71–77, https://doi.org/
158 (2018) 463–477, https://doi.org/10.1016/j.ejmech.2018.09.031.
10.1016/j.xphs.2015.10.016.
[6] S.H. Kim, N. Kandiah, J.-L. Hsu, C. Suthisisang, C. Udommongkol, A. Dash, Beyond
[31] IBM Watson Health, Donepezil hydrochloride. http://doi.org/10.32388/zao7wr,
symptomatic effects: potential of donepezil as a neuroprotective agent and disease
2020. (Accessed 11 December 2020).
modifier in Alzheimer’s disease, Br. J. Pharmacol. 174 (2017) 4224–4232, https://
[32] R. Cacabelos, Donepezil in Alzheimer’s disease: from conventional trials to
doi.org/10.1111/bph.14030.
pharmacogenetics, Neuropsychiatric Dis. Treat. 3 (2007) 303–333.
[7] Alzheimer’s Association, FDA-approved treatments for Alzheimer’s. https://www.
[33] A.S. Gurbuz, S. Ozturk, E. Acar, S.Ç. Efe, T. Akgun, A. Kilicgedik, A. Guler,
alz.org/media/documents/fda-approved-treatments-alzheimers-ts.pdf, 2019.
C. Kirma, Acquired long QT syndrome and Torsades de Pointes related to donepezil
(Accessed 10 December 2020).
use in a patient with Alzheimer disease, Egypt. Hear. J. 68 (2016) 197–199,
[8] R.T. Bartus, R.L. Dean, B. Beer, A.S. Lippa, The cholinergic hypothesis of geriatric
https://doi.org/10.1016/j.ehj.2015.07.004.
memory dysfunction,, Science 217 (1982) 408–417.
[34] California workgroup on guidelines for Alzheimer’s disease management,
[9] J.P. Whitehouse, L.D. Price, G.R. Struble, W.A. Clark, T.J. Coyle, R.M. DeLong,
Guideline for Alzheimer’s disease management. https://www.cdph.ca.gov/prog
Alzheimer’s disease and senile dementia: loss of neurons in the basal forebrain,
Science 215 (1982) 1237–1239.

10
L. Sutthapitaksakul et al. Journal of Drug Delivery Science and Technology 63 (2021) 102549

rams/alzheimers/Documents/professional_GuidelineFullReport.pdf, 2008. evaluation, Int. J. Biol. Macromol. 67 (2014) 418–425, https://doi.org/10.1016/j.


(Accessed 12 December 2020). ijbiomac.2014.03.022.
[35] M. Slavkova, J. Breitkreutz, Orodispersible drug formulations for children and [59] W. Guo, P. Quan, L. Fang, D. Cun, M. Yang, Sustained release donepezil loaded
elderly, Eur. J. Pharmaceut. Sci. 75 (2015) 2–9, https://doi.org/10.1016/j. PLGA microspheres for injection: preparation, in vitro and in vivo study, Asian J.
ejps.2015.02.015. Pharm. Sci. 10 (2015) 405–414, https://doi.org/10.1016/j.ajps.2015.06.001.
[36] T. Harada, T. Uchida, M. Yoshida, Y. Kobayashi, R. Narazaki, T. Ohwaki, A new [60] J.Y. Kim, M.R. Han, Y.H. Kim, S.W. Shin, S.Y. Nam, J.H. Park, Tip-loaded
method for evaluating the bitterness of medicines in development using a taste dissolving microneedles for transdermal delivery of donepezil hydrochloride for
sensor and a disintegration testing apparatus, Chem. Pharm. Bull. 58 (2010) treatment of Alzheimer’s disease, Eur, J. Pharmacokinet. Biopharm. 105 (2016)
1009–1014, https://doi.org/10.1248/cpb.58.1009 (Tokyo). 148–155, https://doi.org/10.1016/j.ejpb.2016.06.006.
[37] K. Bin Liew, Y.T. Tan, K.K. Peh, Characterization of oral disintegrating film [61] M.C. Kearney, E. Caffarel-Salvador, S.J. Fallows, H.O. McCarthy, R.F. Donnelly,
containing donepezil for Alzheimer disease, AAPS PharmSciTech 13 (2012) Microneedle-mediated delivery of donepezil: potential for improved treatment
134–142, https://doi.org/10.1208/s12249-011-9729-4. options in Alzheimer’s disease, Eur, J. Pharmacokinet. Biopharm. 103 (2016)
[38] R.C. Rowe, P.J. Sheskey, M.E. Quinn (Eds.), Handbook of Pharmaceutical 43–50, https://doi.org/10.1016/j.ejpb.2016.03.026.
Excipients, sixth ed., Pharmaceutical Press, London, 2009. [62] M. Yasir, U.V. Singh-Sara, I. Chauhan, P.K. Guar, A.P. Singh, D. Puri,
[39] M. Zeece, Flavors, in: Introduction to the Chemistry of Food, Academic Press, New Ameeduzzafar, Solid lipid nanoparticles for nose to brain delivery of donepezil
York, 2020, pp. 213–250, https://doi.org/10.1016/b978-0-12-809434-1.00006-2. formulation optimization by Box Behnken design in vitro and in vivo evaluation,
[40] J. Walsh, A. Cram, K. Woertz, J. Breitkreutz, G. Winzenburg, R. Turner, C. Tuleu, Artif. Cells. Nanomed. Biotechnol. 46 (2018) 1838–1851, https://doi.org/
Playing hide and seek with poorly tasting paediatric medicines: do not forget the 10.1080/21691401.2017.1394872.
excipients, Adv. Drug Deliv. Rev. 73 (2014) 14–33, https://doi.org/10.1016/j. [63] F. Asmari, Z. ullah, M. tariq, Preparation, characterization, and in vivo evaluation
addr.2014.02.012. of intranasally administered liposomal formulation of donepezil, Drug Des. Devel,
[41] J. Il Kim, S.M. Cho, J.H. Cui, Q.R. Cao, E. Oh, B.J. Lee, In vitro and in vivo Ther (2016) 205–215. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC471
correlation of disintegration and bitter taste masking using orally disintegrating 6722/pdf/dddt-10-205.pdf.
tablet containing ion exchange resin-drug complex, Int. J. Pharm 455 (2013) [64] Y. Fang, N. Zhang, Q. Li, J. Chen, S. Xiong, W. Pan, Characterizing the release
31–39, https://doi.org/10.1016/j.ijpharm.2013.07.072. mechanism of donepezil-loaded PLGA microspheres in vitro and in vivo, J. Drug
[42] K.B. Liew, Y.T. Tan, K.K. Peh, Taste-masked and affordable donepezil Deliv. Sci. Technol. 51 (2019) 430–437, https://doi.org/10.1016/j.
hydrochloride orally disintegrating tablet as promising solution for non- jddst.2019.03.029.
compliance in Alzheimer ’ s disease patients, Drug Dev. Ind. Pharm. 9045 (2015) [65] I.T. Mendes, A.L.M. Ruela, F.C. Carvalho, J.T.J. Freitas, R. Bonfilio, G.R. Pereira,
583–593, https://doi.org/10.3109/03639045.2014.884130. Development and characterization of nanostructured lipid carrier-based gels for
[43] T. Liu, X. Wan, Z. Luo, C. Liu, P. Quan, D. Cun, L. Fang, A donepezil/cyclodextrin the transdermal delivery of donepezil, Colloids Surf, B Biointerfaces 177 (2019)
complexation orodispersible film: effect of cyclodextrin on taste-masking based on 274–281, https://doi.org/10.1016/j.colsurfb.2019.02.007.
dynamic process and in vivo drug absorption, Asian J, Pharmaceut. Sci. 14 (2019) [66] L.C. Espinoza, M. Silva-Abreu, C. Beatriz, M.J. Rodriguez-Lagunas, L. Halbaut, M.-
183–192, https://doi.org/10.1016/j.ajps.2018.05.001. A. Canas, A.C. Calpena, Formulation strategies to improve nose-to-brain delivery of
[44] X. Han, J. Yan, L. Ren, M. Xue, Z. Yuan, T. Wang, Z. Yan, L. Yin, L. Yang, C. Qin, donepezil, Pharmaceutics 11 (2019) 1–16, https://doi.org/10.3390/
Preparation and evaluation of orally disintegrating film containing donepezil for pharmaceutics11020064.
Alzheimer disease, J. Drug Deliv. Sci. Technol, 54 101321 (2019), https://doi.org/ [67] H. Wu, F. Fang, L. Zheng, W. Ji, M. Qi, M. Hong, G. Ren, Ionic liquid form of
10.1016/j.jddst.2019.101321. donepezil: preparation, characterization and formulation development, J. Mol. Liq,
[45] F.G. Bellagambi, T. Lomonaco, P. Salvo, F. Vivaldi, M. Hangouët, S. Ghimenti, 300 112308 (2020), https://doi.org/10.1016/j.molliq.2019.112308.
D. Biagini, F. Di Francesco, R. Fuoco, A. Errachid, Saliva sampling: methods and [68] D. Khunt, M. Shrivas, P. Suryanarayana, P. Gondaliya, M. Manju, Role of omega-3
devices, Trends Anal. Chem. 124 (2020), https://doi.org/10.1016/j. fatty acids and butter oil in targeting delevery of donepezil hydrochloride
trac.2019.115781. microemulsion to brain via the intranasal route: a comparative study, AAPS
[46] S. Stegemann, F. Ecker, M. Maio, P. Kraahs, R. Wohlfart, J. Breitkreutz, A. Zimmer, PharmSciTech 21 (2020) 1–11, https://doi.org/10.1208/s12249-019-1585-7.
D. Bar-Shalom, P. Hettrich, B. Broegmann, Geriatric drug therapy: neglecting the [69] G.R. Topal, M. Meszaros, G. Porkolab, A. Szecsko, T.F. Polgar, L. Siklos, M.A. Deli,
inevitable majority, Ageing Res. Rev. 9 (2010) 384–398, https://doi.org/10.1016/ S. Veszelka, A. Bozkir, ApoE-targeting increases the transfer of solid lipid
j.arr.2010.04.005. nanoparticles with donepezil cargo across a culture model of the blood-brain
[47] R.H. Affoo, N. Foley, J. Rosenbek, J. Kevin Shoemaker, R.E. Martin, Swallowing barrier, Pharmaceutics 13 (2020) 1–19, https://doi.org/10.3390/
dysfunction and autonomic nervous system dysfunction in Alzheimer’s disease: a pharmaceutics13010038.
scoping review of the evidence, J. Am. Geriatr. Soc. 61 (2013) 2203–2213, https:// [70] K. AnjiReddy, S. Karpagam, In vitro and in vivo evaluation of oral disintegrating
doi.org/10.1111/jgs.12553. nanofiber and thin-film contains hyperbranched chitosan/donepezil for active drug
[48] Z.K. Nagy, K. Nyúl, I. Wagner, K. Molnár, M. Gy, Z.K. Nagy, K. Nyúl, I. Wagner, delivery, J. Polym. Environ. 29 (2021) 922–936, https://doi.org/10.1007/s10924-
K. Molnár, G. Marosi, Electrospun water soluble polymer mat for ultrafast release 020-01937-y.
of donepezil HCl, Express Polym. Lett. 4 (2010) 763–772, https://doi.org/ [71] M.W. Jann, K.L. Shirley, G.W. Small, Clinical pharmacokinetics and
10.3144/expresspolymlett.2010.92. pharmacodynamics of cholinesterase inhibitors, Clin. Pharmacokinet. 41 (2002)
[49] H. Suzuki, H. Onishi, Y. Takahashi, M. Iwata, Development of oral acetaminophen 719–739.
chewable tablets with inhibited bitter taste, Int. J. Pharm 251 (2003) 123–132, [72] B. Pietro Imbimbo, Pharmacodynamic-tolerability relationships of cholinesterase
https://doi.org/10.1016/S0378-5173(02)00595-1. inhibitors for Alzheimer ’ s disease, CNS Drugs 15 (2001) 375–390.
[50] B. Albertini, C. Cavallari, N. Passerini, D. Voinovich, M.L. González-Rodrı ́guez, [73] P. Sozio, L.S. Cerasa, L. Marinelli, A. Di Stefano, Transdermal donepezil on the
L. Magarotto, L. Rodriguez, Characterization and taste-masking evaluation of treatment of Alzheimer’s disease, Neuropsychiatric Dis. Treat. 8 (2012) 361–368.
acetaminophen granules: comparison between different preparation methods in a [74] Eisai Co, Eisai Amends License Agreement with Teikoku Pharma USA for Aricept®
high-shear mixer, Eur. J. Pharmaceut. Sci. 21 (2004) 295–303, https://doi.org/ Transdermal Patch System, Ltd., 2012. https://www.eisai.com/news/news20
10.1016/j.ejps.2003.10.017. 1217.html.
[51] K. Woertz, C. Tissen, P. Kleinebudde, J. Breitkreutz, Taste sensing systems for [75] Icure Pharmaceutical Inc, A multinational, multi-center, randomized, double-blind,
pharmaceutical applications, Int. J. Pharm 417 (2010) 256–271, https://doi.org/ active comparator, phase III clinical trial to evaluate the efficacy and safety of
10.1016/j.ijpharm.2010.11.028. donepezil transdermal patch in patients with Alzheimer’s disease. https://clinicaltr
[52] S.L. Rogers, R.S. Doody, R.C. Mohs, L.T. Friedhoff, Donepezil improves cognition ials.gov/ct2/show/NCT03197740, 2017. (Accessed 17 April 2021).
and global function in Alzheimer disease: a 15-week, double blind, placebo- [76] G. Grossberg, C. Sadowsky, H. Förstl, L. Frölich, J. Nagel, S. Tekin, S. Zechner,
controlled study, Clin. Chem. 158 (1998) 1021–1031, https://doi.org/10.1001/ J. Ros, J.M. Orgogozo, Safety and tolerability of the rivastigmine patch: results of a
archinte.158.9.1021. 28-week open-label extension, Alzheimer Dis. Assoc. Disord. 23 (2009) 158–164,
[53] M. Boada-Rovira, H. Brodaty, P. Cras, S. Baloyannis, M. Emre, R. Zhang, R. Bahra, https://doi.org/10.1097/WAD.0b013e31818b1c2c.
Efficacy and safety of donepezil in patients with Alzheimer’s disease: results of a [77] Z. Sidat, T. Marimuthu, P. Kumar, L.C. du Toit, P.P.D. Kondiah, Y.E. Choonara,
global, multinational, clinical experience study, Drugs Aging 21 (2004) 43–53, V. Pillay, Ionic liquids as potential and synergistic permeation enhancers for
https://doi.org/10.2165/00002512-200421010-00004. transdermal drug delivery, Pharmaceutics 11 (2019), https://doi.org/10.3390/
[54] B. Seltzer, Donepezil: a review, expert opin. Drug metab, Toxicol 1 (2005) pharmaceutics11020096.
527–536, https://doi.org/10.1517/17425255.1.3.527. [78] S.N. Pedro, C.S.R. Freire, A.J.D. Silvestre, M.G. Freire, The role of ionic liquids in
[55] J.K. Park, Y. Bin Choy, J. Oh, J.Y. Kim, S. Hwang, J. Choy, Controlled release of the pharmaceutical field: an overview of relevant applications, Int, J. Mol. Sci. 21
donepezil intercalated in smectite clays, Int. J. Pharm 359 (2008) 198–204, (2020) 1–50, https://doi.org/10.3390/ijms21218298.
https://doi.org/10.1016/j.ijpharm.2008.04.012. [79] K. Ita, Transdermal iontophoretic drug delivery: advances and challenges, J. Drug
[56] J. Choi, M.K. Choi, S. Chong, S.J. Chung, C.K. Shim, D.D. Kim, Effect of fatty acids Target. 24 (2016) 386–391, https://doi.org/10.3109/1061186X.2015.1090442.
on the transdermal delivery of donepezil: in vitro and in vivo evaluation, Int. J. [80] P. Bakshi, D. Vora, K. Hemmady, A.K. Banga, Iontophoretic skin delivery systems:
Pharm 422 (2012) 83–90, https://doi.org/10.1016/j.ijpharm.2011.10.031. success and failures, Int. J. Pharm. 586 (2020), 119584, https://doi.org/10.1016/j.
[57] M. Galipoğlu, M.S. Erdal, S. Güngör, Biopolymer-based transdermal films of ijpharm.2020.119584.
donepezil as an alternative delivery approach in Alzheimer’s disease treatment,
AAPS PharmSciTech 16 (2014) 284–292, https://doi.org/10.1208/s12249-014-
0224-6.
[58] S. Bhavna, M. Md, R. Ali, A. Ali, S. Bhatnagar, J. Baboota, Ali, Donepezil
nanosuspension intended for nose to brain targeting: in vitro and in vivo safety

11
L. Sutthapitaksakul et al. Journal of Drug Delivery Science and Technology 63 (2021) 102549

[81] J. Rohrer, N. Lupo, A. Bernkop-Schnürch, Advanced formulations for intranasal [83] M. Agrawal, S. Saraf, S. Saraf, S.G. Antimisiaris, M.B. Chougule, S.A. Shoyele,
delivery of biologics, Int. J. Pharm 553 (2018) 8–20, https://doi.org/10.1016/j. A. Alexander, Nose-to-brain drug delivery: an update on clinical challenges and
ijpharm.2018.10.029. progress towards approval of anti-Alzheimer drugs, J. Contr. Release 281 (2018)
[82] T.P. Crowe, M.H.W. Greenlee, A.G. Kanthasamy, W.H. Hsu, Mechanism of 139–177, https://doi.org/10.1016/j.jconrel.2018.05.011.
intranasal drug delivery directly to the brain, Life Sci. 195 (2018) 44–52, https://
doi.org/10.1016/j.lfs.2017.12.025.

12

You might also like