ArticlePDF AvailableLiterature Review

Alzheimer’s Disease Targeted Nano-Based Drug Delivery Systems

Authors:

Abstract and Figures

Alzheimer’s disease (AD) is the most common neurodegenerative disease, and is part of a massive and growing health care burden that is destroying the cognitive function of more than 50 million individuals worldwide. Today, therapeutic options are limited to approaches with mild symptomatic benefits. The failure in developing effective drugs is attributed to, but not limited to the highly heterogeneous nature of AD with multiple underlying hypotheses and multifactorial pathology. In addition, targeted drug delivery to the central nervous system (CNS), for the diagnosis and therapy of neurological diseases like AD, is restricted by the challenges posed by blood-brain interfaces surrounding the CNS, limiting the bioavailability of therapeutics. Research done over the last decade has focused on developing new strategies to overcome these limitations and successfully deliver drugs to the CNS. Nanoparticles, that are capable of encapsulating drugs with sustained drug release profiles and adjustable physiochemical properties, can cross the protective barriers surrounding the CNS. Thus, nanotechnology offers new hope for AD treatment as a strong alternative to conventional drug delivery mechanisms. In this review, potential application of nanoparticle based approaches in Alzheimer’s disease and their implications in therapy is discussed.
Content may be subject to copyright.
Send Orders for Reprints to reprints@benthamscience.net
628 Current Drug Targets, 2020, 21, 628-646
REVIEW ARTICLE
1389-4501/20 $65.00+.00 ©2020 Bentham Science Publishers
Alzheimer’s Disease Targeted Nano-Based Drug Delivery Systems
Gülcem Altinoglu1,2 and Terin Adali1,2,*
1Department of Biomedical Engineering, Faculty of Engineering, Near East University, P.O. Box: 99138, North Cyprus
via Mersin 10 Turkey; 2Tissue Engineering and Biomaterials Research Centre, Centre of Excellence, Near East Univer-
sity, P.O. Box: 99138, North Cyprus via Mersin 10 Turkey
!
A R T I C L E H I S T O R Y
Received: September 19, 2019
Revised: November 07, 2019
Accepted: November 11, 2019
DOI:
10.2174/1389450120666191118123151
Abstract: Alzheimer’s disease (AD) is the most common neurodegenerative disease, and is part of a
massive and growing health care burden that is destroying the cognitive function of more than 50 mil-
lion individuals worldwide. Today, therapeutic options are limited to approaches with mild sympto-
matic benefits. The failure in developing effective drugs is attributed to, but not limited to the highly
heterogeneous nature of AD with multiple underlying hypotheses and multifactorial pathology. In ad-
dition, targeted drug delivery to the central nervous system (CNS), for the diagnosis and therapy of
neurological diseases like AD, is restricted by the challenges posed by blood-brain interfaces sur-
rounding the CNS, limiting the bioavailability of therapeutics. Research done over the last decade has
focused on developing new strategies to overcome these limitations and successfully deliver drugs to
the CNS. Nanoparticles, that are capable of encapsulating drugs with sustained drug release profiles
and adjustable physiochemical properties, can cross the protective barriers surrounding the CNS.
Thus, nanotechnology offers new hope for AD treatment as a strong alternative to conventional drug
delivery mechanisms. In this review, the potential application of nanoparticle based approaches in
Alzheimer’s disease and their implications in therapy is discussed.
Keywords: Alzheimer’s disease, nanoparticles, nanotechnology, drug delivery, targeted delivery, physiochemical.
1. INTRODUCTION
Alzheimer’s Disease (AD) is the most common type of
dementia, accounting for more than 80% of reported demen-
tias around the globe [1]. The increasing prevalence of AD
stands as one of the most severe burden on socio-economy
[2]. There are currently around 50 million people suffering
from the disease, that is expected to rise over 100 million by
the year 2050 [3]. It is characterised by irreversible neurode-
generation due to a progressive loss of brain cells and their
connections [4], leading to impairments in memory, mental
functioning, thinking and behaviour. According to the Diag-
nostic and Statistical Manual of Mental Disorders V (DSM-
V), AD can be diagnosed on having a new onset memory
impairment with a gradual progression of cognitive decline
in at least one or more of other cognitive domains including
complex attention, executive function, language, perceptual-
motor, or social cognition [5]. AD combines both genetic
and environmental factors in the formation of its pathology
[6]. Advanced age is considered as the main environmental
trigger along with other metabolic and vascular conditions
such as obesity, diabetes, trauma, stroke, high cholesterol,
*Address correspondence to this author at the Department of Biomedical
Engineering, Faculty of Engineering, Near East University, P.O. Box:
99138, North Cyprus via Mersin 10 Turkey; and Tissue Engineering and
Biomaterials Research Centre, Centre of Excellence, Near East University,
P.O. Box: 99138, North Cyprus via Mersin 10 Turkey; Tel: 03926802002;
E-mail: terin.adali@neu.edu.tr
hypertension and cardiovascular disorders [7]. Genetic re-
search suggests the involvement of 5 genes that may be
linked to the pathology of AD; including mutations of amy-
loid precursor protein (APP), presenilin 1 (PS1) and preseni-
lin 2 (PS2) genes and having the apolipoprotein E (ApoE) e4
allele and/or rare variants in the triggering receptor ex-
pressed on myeloid cells 2 (TREM2) gene that have been
revealed to increase individuals’ risk of developing AD [8-
10].
The available pharmacological treatments against AD are
yet unable to prevent or slow down the progression of the
neurodegenerative process. They can only offer symptomatic
solutions, targeting the cognitive manifestations of the disor-
der with severe side effects i.e. acetylcholinesterase inhibi-
tors (rivastigmine, galantamine, donepezil and tacrine) and
N-methyl D-aspartate receptor antagonist (memantine) [11].
The lack of effective therapeutic agents highlights the impor-
tance of gaining a deeper understanding of the underlying
mechanisms of AD. There have been several hypotheses, yet
not a single theory is able to explain all aspects of this highly
heterogeneous disorder [12]. The most established underly-
ing hypotheses that have been put forward include the amy-
loid beta, tau, cholinergic and oxidative stress hypotheses
(Fig. 1), along with other secondary pathological mecha-
nisms such as the glutamate excitotoxicity and inflammation
hypotheses. Numerous candidate drugs with diverse pharma-
cological mechanisms have been tested [13]. However, de-
spite impressive progress in clinical trials, most of the thera-
Alzheimer’s Disease Targeted Nano-Based Drug Delivery Systems Current Drug Targets, 2020, Vol. 21, No. 7 629
peutic approaches have failed to modify the course of the
disease irrespective of the form of therapy and/or pathologi-
cal hallmark targeted [14].
Drug delivery to the central nervous system (CNS) is
restricted due to the challenges posed by the blood-brain
barrier (BBB). Nanoparticle-mediated drug delivery repre-
sents a viable approach to facilitate the delivery of therapeu-
tic agents to the CNS bypassing the BBB. In the context of
targeting AD, different nanocarriers are being explored. The
aim of this review is to discuss the potential application of
nanoparticle based approaches in Alzheimer’s disease and
their implications in therapy.
2. ALZHEIMER’S DISEASE HYPOTHESES
2.1. Amyloid Cascade Hypothesis
One of the most established pathological hallmarks of the
disease that has been accepted as the main explanation for
AD for over 25 years is the “amyloid hypothesis”. According
to this hypothesis, there is an extracellular amyloid-beta (Aβ)
peptide accumulation, leading to the formation of Aβ
plaques, that represent an early and central event in AD pa-
thology [15]. It accepts that Aβ plaques are the causative
factor, and that other pathophysiological features of the dis-
order follow its deposition in the brain [16].
Normally, Aβ is derived from the proteolytic cleavage
of a larger transmembrane protein, amyloid precursor pro-
tein (APP), and subsequently gets degraded in the brain.
APP is sequentially cleaved by the action of β- and γ-
secretases. β- and γ-secretase yield several isoforms of the
Aβ peptide [17] but those ending at position 40 (Aβ40) and
position 42 (Aβ42) are the most pronounced Aβ species in
AD patients’ brains. Pathological Aβ accumulation is likely
the result of an altered balance between overproduction and
degradation. Spontaneous aggregation of Aβ forms small
clusters called oligomers, which develops into insoluble
fibrils with a beta-sheet structure, and eventually into senile
plaques [18] (Fig. 2). These processes trigger neurotoxicity,
neural dysfunction and ultimately neural death, leading to
neurodegeneration.
Although the majority of research in literature still sup-
ports the significance of Aβ as the main initiating trigger of
the pathogenic cascade of AD, growing evidence also sug-
gests that Aβ pathology alone is inadequate in the late stages
of the disease [19]. Advances in neuroimaging have allowed
the observation of Aβ accumulation in vivo. It has been
consistently shown that there is a lack of correlation between
Aβ aggregates and AD-related cognitive impairment [20],
with many healthy individuals having amyloid deposits, and
AD patients with very few aggregates [17, 21]. Additionally,
the failure in developing effective Aβ targeted drugs with no
significant improvements to disease symptoms and/or pro-
gression (via the blockage of Aβ aggregation, reducing its
soluble peptides or destroying already existing Aβ plaques)
formed another growing line of evidence questioning the
impact of Aβ in AD pathology. Following numerous failures
of developing Aβ targeted drugs, attention has shifted to neu-
rofibrillary tangles (NFTs), the second most established
pathological hallmark of AD, that are composed of tau pro-
tein.
2.2. Tau Hypothesis
Tau is a soluble, microtubule associated scaffolding pro-
tein modulating the cytoskeletal dynamics of neural cells
[22]. Tau hypothesis accepts tau to be the main causative
factor in AD development [18]. Under normal conditions,
tau undergoes many modifications one of which is phos-
phorylation by various kinases. In the case of AD, there is a
state of hyper-phosphorylation [23], which raises the ques-
tion as to what mechanisms trigger the abnormal phosphory-
lation state. Although the transmission mechanism leading to
tauopathy is not fully comprehended, literature suggests the
Fig. (1). Simple overview of the most established underlying hypotheses of Alzheimer’s disease. (A higher resolution / colour version of this
figure is available in the electronic cop y of the article).
630 Current Drug Targets, 2020, Vol. 21, No. 7 Altinoglu and Adali
involvement of damage signals such as Aβ oligomers (as
stated by the amyloid cascade hypothesis), iron overload,
oxygen free radicals, cholesterol levels in the neural raft and
the involvement of innate immune system through microglial
cell activation [24]. Once hyper-phosphorylated, tau starts to
dissociate from microtubules and pair with other threads of
tau, leading to its aggregation into oligomers [25]. Eventu-
ally, they form intracellular NFTs, destroying the structure of
the neural cytoskeleton and ultimately resulting in neu-
ronal/synaptic dysfunction and cell death (Fig. 3). Although
most of the efforts have failed in clinical trials, tau-targeting
treatment strategies focus on blocking tau aggregation,
modulating tau modifications and stabilizing microtubules
[11].
2.3. Oxidative Stress Hypothesis
Increasing evidence supports the hypothesis that oxida-
tive stress, triggered by different mechanisms, may play a
causative role in initiating neurodegeneration [26, 27]. Oxi-
dative stress indicates an imbalance between anti-oxidants
and oxidants, in favor of oxidants, and have been associated
with triggering or enhancing Aβ accumulation and NFT
deposition in AD patients. Mitochondrial dysfunction fol-
lowed by an increase in toxic free-radicals is reported to be
the biological basis of the anti-oxidant and oxidant imbal-
ance, leading to oxidative damage, which in turn can initiate
the apoptotic pathway [28]. It has also been evidenced that
there is a strong link between oxidation reactions and in-
creased local concentration of transition metals such as iron
(Fe2+), copper (Cu2+), aluminium (Al3+), and zinc (Zn2+)
that may act as the source of free radicals prior to triggering
oxidative damage and neurotoxicity [29].
2.4. Cholinergic Hypothesis
The cholinergic hypothesis of AD is based on the finding
that a dysfunction of cholinergic neurons which use acetyl-
choline (involved important physiological functions e.g.
learning and memory) as a neurotransmitter is commonly
seen in people with advanced age and AD patients, and that
it correlates with cognitive decline and memory impairments
observed in these individuals [30]. Acetylcholine (ACh) is
synthesized in the cytosol of presynaptic cholinergic neurons
from choline and Acetyl Coenzyme A (Ac CoA) by choline
acetyltransferase (ChAT) enzyme and binds to postsynaptic
cholinergic receptors to cause a response. Brains of patients
with AD revealed a reduced choline uptake and ChAT activ-
ity in ACh synthesizing neurons, in addition to impairments
in post synaptic Ach receptors (Fig. 4). Therefore, the cho-
linergic system remains a highly viable target for sympto-
matic AD treatment [31]. Cholinergic drugs that are cur-
rently on the market (acetylcholinesterase inhibitors
(AChEIs)), work by inhibiting the enzyme, acetylcho-
linesterase (AChE), that breaks down ACh in the synaptic
cleft. This improves cholinergic neurotransmission and thus
the cognitive symptoms of the disorder. This hypothesis is
supported by various findings in the literature, which illus-
trates that AD-like symptoms (e.g. memory and cognitive
decline) can be induced in young subjects by blocking cho-
linergic transmission via cholinergic antagonists and re-
versed by agonists [32, 33]. However, it is important to note
that not all early stage AD patients reveal a cholinergic dys-
function and are responsive to cholinergic drugs [12], chal-
lenging the validity of this hypothesis.
3. STRUCTURAL AND FUNCTIONAL BARRIERS
The disappointing results of AD therapy are the result of
its multifactorial pathology and the failure to detect it in its
early stages before any clinical manifestations [34]. Drug
delivery to the CNS is also limited by the challenges posed
by blood-brain interfaces, including the BBB and the blood
cerebrospinal fluid barrier (BCSFB) [35]. BBB is the major
interface between the brain and circulating blood. It is a
semi-permeable, highly selective barrier. It acts as a special-
ized gatekeeper to maintain the chemical composition of the
neuronal environment at normal physiological levels by
regulating the exchange of ions/molecules [36]. BBB con-
sists of a network of endothelial cells connected by tight
junctions, enzymatic barriers and transport proteins, and
Fig. (2). Formation of amyloid beta plaques in Alzheimer’s disease pathogenesis. Abbreviations: APP, Amyloid precursor protein; AB,
Amyloid beta. (A higher resolution / colour version of this figure is available in the electronic copy of the article).
Alzheimer’s Disease Targeted Nano-Based Drug Delivery Systems Current Drug Targets, 2020, Vol. 21, No. 7 631
Fig. (3). Formation on neurofibrillary tangles in Alzheimer’s disease pathogenesis. Abbreviations: AB, Amyloid beta; nAchR, Nicotinic ace-
tylcholine receptors; NMDAR, NMDA receptors on neuronal membrane. (A higher resolution / colour version of this figure is available in the
electronic cop y of the a rticle).
Fig. (4). Schematic representation of cholinergic neurotransmission in normal, mild and late stages of Alzheimer’s disease. Abbreviations:
ACh, Acetylcholine; AChE, Acetylcholinesterase; ChAT, Choline acetyltransferase; Ch transporter, Choline transporter; NGF, Nerve growth
factor; M1AChR/M2AChR; Muscarinic acetylcholine receptors Type 1/2; nAChR, Nicotinic acetylcholine receptors. (A higher resolution /
colour version of this figure is available in the electronic copy of the article).
632 Current Drug Targets, 2020, Vol. 21, No. 7 Altinoglu and Adali
functionally restricts the entrance of solutes or drugs that
may be of good neuro-therapeutic value [37]. Thus, the main
obstacle for CNS drug delivery stems from the presence of
the BBB. BBB prevents the entry of “foreign” substances
into the CNS, and stops the transfer of almost all high mo-
lecular weight drugs and more than 98% of low molecular
weight drugs [38]. Lipophilicity and surface activity of
molecules are other physiochemical parameters that deter-
mine the extent to which compounds can penetrate through
this physical barrier [39]. Therapeutics or compounds having
a low molecular weight (400 Da), that are lipophilic, and not
ionized at physiological pH can transport through the BBB
by diffusion [40]. Essential compounds e.g. amino acids,
hexoses and neuropeptides, however, require specific carri-
ers to cross the BBB [40], while proteins and peptides need
saturable transport systems [41]. Consequently, the first
thing to consider during drug development is their ability to
override the BBB.
The BCSFB is another physical obstacle separating the
blood from the cerebrospinal fluid (CSF) in the subarachnoid
space, located at the choroid plexus of epithelial cells in-
volved in movement restriction [42]. Even supposing some
drugs pass these biological barriers, CNS employs other de-
fence mechanisms to reduce the potential for systemic expo-
sure [43-45]. Additionally, therapeutic agents display poor
solubility, bioavailability, and targeting potency in reaching
their pathogenic targets adding to the limitations posed by
blood-brain interfaces [34]. Therefore, it is of great impor-
tance to understand the structural and functional barriers
responsible for the failure in the treatment of CNS diseases
while designing new potential drugs and delivery mecha-
nisms [46].
Over the last decade, continuous research efforts have
been made to overcome these limitations and increase the
bioavailability of therapeutic compounds. Among these, drug
delivery systems based on nanotechnology, which combines
the fields of engineering and technology, have gained a lot of
scientific interest and offer new hope for AD treatment with
encouraging success [14] (Table 2).
4. NANOPARTICLES: DRUG DELIVERY SYSTEMS
Recent advances in nanotechnology have allowed to offer
a novel opportunity for AD treatment. Due to having a con-
trol over their shape, size, hydrophobicity, coating, chemis-
try and surface charge, nanoparticle-based approaches have
been utilized as promising tools as effective drug carriers
since their first development as carrier materials by Birren-
bach and Speiser in the 1970s [47]. By definition, nanoparti-
cles (NPs) are microscopic particles with at least one dimen-
sion below 100 nm and are able to interact with biological
systems. They are composed of a surface layer, that can be
functionalized with different molecules, and an internal core
[48]. NPs are capable of overcoming the limitations posed by
biological barriers and conventional treatment methods, al-
lowing for the encapsulation of therapeutics and their trans-
port into the CNS bypassing the BBB [49]. They can be de-
signed and modified with different surface characteristics to
optimize their pharmacokinetic and pharmacodynamic pro-
files [37]. Nano-mediated drug delivery systems offer good
stability, biocompatibility, controlled/sustained drug release
and biodegradability, as well as low toxicity and immuno-
genicity response. NPs can also provide a reduction of the
quantity and frequency of dosing, and any probably side ef-
fects (Table 1) [50].
4.1. Important NP Characteristics for Brain Drug Delivery
Biocompatible, biodegradable, non-reactive and non-
toxic nanomaterials are desired when developing nano-
particulate drug delivery systems [46]. The size, the struc-
tural conformation, the molecular weight and charge, the
encapsulating materials used, the lipophilicity, affinity for
cellular proteins, the zeta potential, the concentration gradi-
ents of encapsulated agents, the affinity of the agent and the
dosage form for the receptors are all among the important
characteristics that need to be considered while designing
NPs. Having a control over these features allows for the abil-
ity to optimize their BBB transport efficiency, drug release,
stability, and the ability to escape from their rapid circulation
clearance mechanism, the reticuloendothelial system [51-53].
Several parameters influence the passage and efficacy of
NPs across the BBB. Numerous findings have indicated an
inverse correlation between NP size and the ability to over-
come the restrictive mechanism of the BBB [54-56]. Specifi-
cally, NPs that are 50 nm 100 nm in diameter have been
preferred for a successful transport of NPs in AD animal
models [2]. Smaller particles have also been associated with
Table 1. Potential advantages and disadvantages of nanoparticle based approaches for CNS drug delivery.!
Disadvantages
1. BBB transport is not indicative of the biological activity
2. May influence the physiology of any cell in the body
3. Limited information on metabolic fate and pathway
4. Possible aggregation
5. May cause toxicity
6. Possibility to further disease progression
Alzheimer’s Disease Targeted Nano-Based Drug Delivery Systems Current Drug Targets, 2020, Vol. 21, No. 7 633
Table 2. Nano-enabled d rug delivery systems for AD therapy.!
Nano-delivery
Systems
Therapeutic Agent
Category
Proposed Target Hypothesis
Carrier Material
References
Polymeric NPs
Rivastigmine
AChEI
Cholinergic system
PnBCA
[117, 118,
120, 151]
-
-
-
-
PLGA
-
-
-
-
-
Chitosan
-
-
Galantamine-
hydrobromide
AChEI
Cholinergic system
Chitosan
[119, 152]
-
-
-
-
PLGA
-
-
Donepezil
AChEI
Cholinergic system
Chitosan
[121, 153, 154]
-
-
-
-
PLGA
-
-
-
-
-
(PEG)PLGA
-
-
Galantamine
AChEI
Cholinergic system
Chitosan
[127]
-
Tacrine
AChEI
Cholinergic system
Chitosan
[124, 125]
-
-
-
-
PnBCA
-
-
Acetylcholine
Proteins and peptides
Cholinergic system
Human serum albumin
[128]
-
Aβ sub-fragments
Anti-amyloid
Amyloid cascade
Chitosan
[91, 155, 156]
-
-
-
-
Gold
-
-
Coenzyme Q10
Anti-amyloid and
Amyloid cascade
PLGA
[157]
-
-
antioxidant
and oxidative damage
-
-
-
PPAR- γ agonist
Anti-amyloid
Amyloid cascade
PEG-PLGA
[158]
-
Methylene blue
Tau aggregation in-
hibitor
Tau cascade
PEG-PLGA
[108]
-
MKT-077
Tau aggregation in-
hibitor
Tau cascade
PEG-PLGA
[111]
-
Fibroblast growth
factor
Proteins and peptides
Neuroprotective and choliner-
gic system
PEG-PLGA
[159]
-
Nerve growth factor
Proteins and peptides
Cholinergic system
PnBCA
[160]
-
Vasoactive intestinal
Proteins and peptides
Neuroprotective peptide with
PEG-PLA
[99, 100, 98]
-
-
peptide
with anti-amyloid and
-
-
-
-
-
antioxidant effects
-
-
-
iAβ5- β-sheet breaker
Proteins and peptides
Amyloid cascade
PLGA
[94]
-
Peptide
-
-
-
-
-
TGN and QSH
Proteins and peptides
Targeted delivery to amyloid
PEG-PLA
[29]
-
-
-
cascade
-
-
-
NAPVSIPQ (NAP)
Proteins and peptides
Neuroprotective peptide
PEG-PLA
[161]
-
Estradiol
Hormones
Amyloid cascade and
Chitosan, PLGA
[103-105]
-
-
-
cholinergic system
-
-
-
Melatonin
Hormones
Oxidative stress
Eudragit S10
[162]
(Table 2) contd….
634 Current Drug Targets, 2020, Vol. 21, No. 7 Altinoglu and Adali
Nano-delivery
Systems
Therapeutic Agent
Category
Proposed Target Hypothesis
Carrier Material
References
-
Mifepristone
Hormones
Amyloid cascade
PLGA
[163]
-
Curcumin
Polyphenols
Oxidative stress
PnBCA
[135, 136,
164, 165]
-
-
(Antioxidant)
-
PLGA
-
-
Resveratrol
Polyhphenols
Oxidative stress
PEG-PCL
[141]
-
-
(Antioxidant)
-
-
-
-
ECGC
Polyphenols
Oxidative stress
Chitosan
[166]
-
-
(Antioxidant)
-
-
-
-
Piperine
Polyphenols
Oxidative stress
Chitosan
[167]
-
-
(Antioxidant)
-
-
-
-
Nano-N2PY
Chelating agent (Iron)
Metal induced
Polystyrene
[168]
-
-
-
oxidative stress
-
-
-
D-penicillamine
Chelating agent (Cop-
per)
Metal induced
MPB-PE
[169]
-
-
-
oxidative stress
-
-
Solid Li pid NPs
Galantamine
AChEI
Cholinergic system
Glycerylbehnate
[170]
-
hydrobromide
-
-
(compritol)
-
-
Rivastigmine
AChEI
Cholinergic system
Compritol 888 ATO
[171]
-
Curcumin
Polyphenols (Antioxi-
dant)
Cholinergic system
-
[134]
-
Resveratrol
Polyphenols
Oxidative stress
-
[140, 49]
-
-
(Antioxidant)
-
-
-
-
Ferulic acid
Polyphenols (Antioxi-
dant)
Oxidative stress
Compritol 888 ATO
[142, 172]
-
epigallocatechin-3-
gallate
Polyphenols (Antioxi-
dant)
Oxidative stress
monophasic liquid
[173]
-
(ECGC)
-
and amyloid cascade
preparations
-
-
D-penicillamine
Chelating agent (Cop-
per)
Metal induced oxidative stress
hexadecanol and
[169]
-
-
-
-
1,2-dioleoyl-sn-glycero-
-
-
-
-
-
3-phospho- ethanolamine-
-
-
-
-
-
N-[3-(2-pyridyldithio)-
-
-
-
-
-
propionate]
-
-
Nerve growth factor
Proteins and peptides
Cholinergic system
(Heparin-conjugated stearic
[174]
-
-
-
-
acid; stearylamine-
-
-
-
-
-
cationic lipid; esterquat
-
-
Nicotinamide
Neuroprotective agent
Tau cascade
Polysorbate 80,
[114]
-
-
-
-
phosphatidylserine or
-
-
-
-
-
phosphatidic acid coating
-
(Table 2) contd….
Alzheimer’s Disease Targeted Nano-Based Drug Delivery Systems Current Drug Targets, 2020, Vol. 21, No. 7 635
Nano-delivery
Systems
Therapeutic Agent
Category
Proposed Target Hypothesis
Carrier Material
References
Nanostructured
Curcumin-donepezil
Antioxidant-AChEI
Oxidative stress and
-
[133]
Lipid carriers
-
-
cholinergic system
-
-
Liposomes
Galantamine
AChEI
Cholinergic system
soya phosphatidylcholine,
[175]
-
-
-
-
cholesterol, and propylene
-
-
-
-
-
glycol
-
-
Rivastigmine
AChEI
Cholinergic system
EPC, cholesterol,
[176-178]
-
-
-
-
DSPE-PEG-CPP
-
-
-
-
-
phosphatidylcho-
line/cholesterol
-
-
-
-
-
soya lecithin/cholesterol
-
-
Donepezil
AChEI
Cholinergic system
1,2-distearyl-sn-glycero-3-
[179]
-
-
-
-
phosphocholine (DSPC),
-
-
-
-
-
cholesterol (CHE), polyethylene
-
-
-
-
glycol (PEG)
-
-
Brain derived neu-
rotrophic factor
Proteins and peptides
Neuroprotective peptide
-
[180]
-
Curcumin
Polyphenols
Oxidative stress and amyloid
-
[181]
-
-
(Antioxidant)
cascade
-
-
Nano-emulsions
Huperzine A
Natural AChEI
Cholinergic system
-
[182]
-
Ginkgo biloba
Antioxidant
Oxidative stress
-
[183]
-
Tabernaemontana
Natural AChEI
Oxidative stress
-
[184]
-
divaricate
-
-
-
-
-
Beta-asarone
Neuroprotective agent
Oxidative stress
-
[185]
Nanocomposites
Methylene blue
Tau aggregation in-
hibitor
Tau cascade
CeNC/IONC/MSN-T807
[106]
a more uniform body distribution [57], and prolonged dura-
tion of action [58]. Drug loading and release are also influ-
enced by NP size [59]. Larger NPs with relatively large cores
allow for drug encapsulation to a greater extent per particle.
Additionally, larger NPs provide slower drug release rates
whereas smaller NPs offer faster drug release profiles. This
is due to the fact that most of the drugs encapsulated in
smaller particles would be at or near the NP surface due to
the large surface area to volume ratio of smaller NPs [60]. It
is important to note that drug loading and release efficacy is
also dependent on other important characteristics of the drug
of interest and the biomaterial of choice.
Regarding the surface characteristics, NPs can have dif-
ferent shapes varying from spherical, rod-shaped, cubic and
cylindrical models, and charges (positive, zwitterionic, and
negative). The shape of NPs influences not only BBB up-
take, but also their distribution and clearance [57]. Zeta po-
tential is another key element affecting the tendency of NPs
to penetrate through the BBB [39]. It is a measure of the
overall surface charge on the NP surface [61], and is an indi-
cator of particle stability [62]. It has been shown that most
NPs for CNS delivery have moderate or highly negative zeta
potentials, between -1 to -15 mV or -15 to -45 mV respec-
tively [63-65]. Highly positive zeta potentials are linked with
inducing BBB toxicity, yet some NPs up to 15 mV or above
have been successful in permeating through the BBB and are
used as effective delivery mechanisms [66, 67]. Being neu-
tral and zwitterionic in nature they also contribute to longer
circulation times and clearance as opposed to negative and
positive NPs [68]. The correct mechanism(s) of transport of
NPs across the BBB is not clearly established. Yet, studies
suggest the involvement of various processes including dif-
fusion, endocytosis and/or transcytosis [69], depending on
the physiochemical characteristics of NPs.
NPs can be classified into various categories. In this re-
view, we have focused on polymeric NPs, lipid-based NPs
(solid lipid NPs and nanostructured lipid carriers), liposomes
and nano-emulsions.
636 Current Drug Targets, 2020, Vol. 21, No. 7 Altinoglu and Adali
4.2. Polymeric Nanoparticles
Polymeric NPs are solid colloidal particles that can
transport therapeutic agents by encapsulating, entrapping or
bounding covalently to them [70, 52, 53]. These polymers
are the most extensively studied nano-carriers due to a sta-
ble, biocompatible and biodegradable nature with low toxic-
ity and immunogenicity [71]. Polymeric NPs may be pre-
pared from both natural polymers (e.g. polysaccharides (chi-
tosan and alginate), amino acids (poly(lysine), poly(aspartic
acid) (PASA)), or proteins (gelatin and albumin) [72], and
synthetic polymers (e.g. poly(ethylenimine) (PEI),
poly(alkylcyanoacrylates), poly(amidoamine) dendrimers
(PAMAM), poly(ε-caprolactone) (PCL), poly (lactic-co-
glycolic acid) (PLGA), polyesters (poly(lactic acid) (PLA),
or from inorganic materials, such as gold, silicon dioxide
(silica) and alumina) [2]. NPs obtained from natural poly-
mers offer the benefit of providing biological signals for
specific receptors and transporters, while expressing some
intrinsic limitations due to challenges with which they can be
modified, as opposed to synthetic NPs that can be designed
to obtain a wide range of necessary characteristics [70]. Po-
lymeric NPs are obtained by employing various preparation
techniques such as ionic gelation, polymer polymerization,
nano-precipitation, spray drying and emulsion solvent
evaporation [73], where the technique employed is deter-
mined by the characteristics of encapsulated material and the
particular polymer used [74].
4.3. Lipid-based Nanoparticles
Lipid-based nano-carriers are another type of nano-
particulate systems that offer good drug loading capacities,
protection against drug degradation, sustained and controlled
drug release and low toxicity issues. Lipid NPs are colloidal
particles that can cross the BBB via endocytosis due to their
lipophilic nature, which would also have a beneficial effect
on the ease of loading lipophilic drugs and surface function-
alization [75]. Additionally, lipid carriers have compara-
tively enhanced drug loading efficacies compared to polym-
eric NPs, allowing them to have greater control over drug
release [76]. Yet, they also express some limitations includ-
ing poor in vivo stability and poor loading of hydrophilic
agents [35].
Lipid NPs consist of solid lipid nanoparticles (SLNs) and
nanostructured lipid carriers (NLCs), both of which have been
used as drug delivery vehicles for AD treatment. SLNs are
typically composed of a solid lipid core matrix, in which the
therapeutic drugs can be dispersed or dissolved [77]. Their
nano-size (40 nm 200 nm) offer them the ability to elude the
liver and the reticuloendothelial system, and thus penetrate
through the endothelial cells of the BBB [78]. SLNs may be
prepared from lipids, emulsifying agents, and water/solvent
that are biocompatible for application in humans. Different
preparation techniques can be utilized in their manufacture
comprising ultra-sonication/high-shear technique, high pres-
sure homogenization, solvent emulsification diffusion,
evaporation, double emulsion and spray drying techniques
[79]. NLCs, on the other hand, are drug delivery vehicles with
both solid and liquid lipid cores, that were developed to avoid
some of the limitations of SLNs such as limited drug-loading
capacity and expulsion during storage [80].
4.4. Liposomes
Liposomes are vesicles comprising one or multiple phos-
pholipid bilayers that self-assemble around an aqueous core
[81], and are therefore categorized as uni-lamellar or multi-
lamellar [82], with sizes ranging from 50 nm to 100 µm.
Liposomes can be utilized as biocompatible and non-toxic
carriers of not only lipophilic or hydrophilic drugs [83], but
also of hydrophobic and amphiphilic agents that can be en-
trapped within the aqueous core of liposomes, due to their
phospholipid nature. Once administered, liposomes are ca-
pable of BBB transport via lipid mediated diffusion or endo-
cytosis. As with other nano-particulate systems, different
techniques can be employed to prepare liposomes including
hydration of a thin lipid film followed by agitation, sonica-
tion, high-pressure homogenization, reverse-phase evapora-
tion, and extrusion [84].
4.5. Nano-Emulsions
Nano-emulsions are nanometric-scale oil-in-water (O/W)
or water-in-oil (W/O) emulsions, typically having a droplet
size ranges of 20 nm-200nm [85], making them interesting
candidates to enhance drug delivery. They are transparent
heterogeneous mixtures comprised of oil droplets, surfac-
tant(s) as stabilizers and water or other aqueous medium
[50]. They are kinetically stable systems while being metas-
table thermodynamically [35]. They may be prepared by
different processes including high-energy input or low-
energy methods (such as high-pressure homogenizers and
spontaneous emulsification respectively), ultrasound genera-
tors and phase inversion temperature [85]. The choice of oil
employed in the preparation process has been reported to
have an effect on their uptake into the CNS. Despite posing
limitations regarding their instability during storage leading
to phase separation and burst-release effect [50, 86], several
studies have employed nano-emulsion systems to improve
drug delivery to the brain [87].
5. NANO-ENABLED ANTI-AD STRATEGIES
5.1. Nano-enabled Anti-amyloid Strategies
As discussed previously, Aβ aggregation is a commonly
reported pathological hallmark of AD. The search for new
drug candidates for AD has indicated that neuroprotective
peptides hold promise therapy-wise [2]. They can act in a
variety of ways by breaking down Aβ plaque formation, de-
grading Aβ toxic peptide and modulating Aβ cleaving en-
zymes [88]. Additionally, more futuristic neuro-regenerative
approaches aspire to rebuild the damaged tissue and reverse
the disease pathology [89, 90]. In this context, several nano-
carrier systems have been studied.
Sub-fragments of Aβ are proposed to protect brain cells
from AD. Yet, their bioavailability for in vivo applications is
restricted owing to their low permeability through the BBB.
To this end, Songjiang and Lixing synthesized chitosan NPs
loaded with Aβ sub-fragments via mechanical stirring emul-
sification methods [91]. Chitosan is a biocompatible and
biodegradable natural polymer, that has been used exten-
sively as a carrier for controlled drug delivery applications
[92]. Their results confirmed that this nano-mediated deliv-
ery has successfully carried out Aβ to the brain with a sig-
Alzheimer’s Disease Targeted Nano-Based Drug Delivery Systems Current Drug Targets, 2020, Vol. 21, No. 7 637
nificant immunogenic response, underlining the importance
of research on peptide vaccines for AD [91]. iAβ5 peptide is
also a shorter anti-β-sheet peptide. Despite its significant
activity to inhibit Aβ fibrillogenesis, iAβ5 peptide is unstable
and easily degradable by proteases interrupting its use in in
vivo applications [93]. To this end, iAβ5 loaded NPs can be a
promising strategy. A research group loaded iAβ5 peptides
in PLGA NPs functionalized with two different types of an-
tibodies, an anti-transferrin receptor monoclonal antibody
OX26 (surface receptor on the brain capillary endothelium)
and anti-Aβ DE2B4 (targeting peptide for Aβ) [94].
Poly(lactic-co-glycolic acid) (PLGA) NPs are another type
of extensively studied polymeric carriers for drug delivery in
relation to their ability to offer surface functionalization,
enhanced drug loading capacity and biocompatibility with
low toxicity issues [95]. Results showed that when function-
alized, the CNS delivery of iAβ5 loaded PLGA NPs was
significantly enhanced compared to NPs with no functionali-
zation, with the purpose of increasing their bioavailability
and weakening the brain Aβ aggregates. Vasoactive intesti-
nal neuropeptide (VIP) is another neuroprotective major neu-
ropeptide found extensively in the central and peripheral
nervous system [96]. Besides possessing anti-inflammatory
effects, in vivo and in vitro studies of VIP demonstrated an
ability to inhibit excitotoxicity mediated neural death [97],
and reduce Aβ toxicity [98]. However, developing VIP-
based drug design against AD was not a success owing to its
instability and limited bioavailability. Encapsulation of VIP
by polymeric NPs has been proposed as an alternative solu-
tion by various research groups. Gao and his colleagues syn-
thesized I125 vasoactive intestinal peptide (I125VIP) loaded
poly (ethylene glycol)-poly (lactic acid) (PLA) NPs modified
with wheat germ agglutinin (WGA). Bio-distribution and
uptake of I125 VIP exhibited enhanced concentrations of the
peptide in the mice brain after intranasal administration
when loaded in WGA-conjugated and non-conjugated NPs
compared to the free drug, with an increase of 5.67.7-folds
and 3.54.7-folds, respectively [99]. Another study from
Kanwar and associates have incorporated I125 VIP into the
PEG-PLA NPs surface modified with lectins and wheat germ
agglutinin (WGA). Lectins are proteins or glycoproteins that
bind to Aβ protein. They reported a 2-fold increased uptake
of lectin modified NPs in comparison to uncoated I125 VIP
loaded NPs [100]. Zhang et al. also developed a dual-
functional anti-amyloid NP system targeting the Aβ plaques
in the brains of transgenic AD mice based on a PEGylated
PLA NPs surface functionalized with two targeting peptides;
TGN and QSH. TGN targets ligands at the BBB for BBB
penetration, while QSH has a good affinity for Aβ1-42 for
specific targeting of the plaques [29]. This study demon-
strated an enhanced and precise delivery in AD model mice,
proving effective as a valuable targeting system and improv-
ing research on disease-modifying strategies. It can be con-
cluded that surface modified polymeric NPs can serve as
promising delivery systems for carrying anti-amyloid agents
like peptides or proteins to the brain.
Besides peptides and proteins, hormones can also be
loaded in NPs that can be of good use therapeutically. There
is significant preclinical evidence that gonadal steroids (e.g.
estrogens and androgens) are key players of brain function-
ing, influencing a wide range of brain activities [101]. Addi-
tionally, estrogen has been suggested to reduce the cerebral
amyloid load and stimulate the growth and survival of cho-
linergic neurons in the brain [102]. Taking this into consid-
eration, Wang et al synthesized estradiol loaded chitosan
NPs by ionic gelation technique that was administered to
Wistar rats either intranasally or intravenously. Following
intranasal administration, plasma concentrations (32.7 ± 10.1
ng/ml) were significantly lower compared to its intravenous
administration (151.4 ± 28.2 ng/ml), whereas CSF concen-
trations after intranasal administration (76.4 ± 14.0 ng/ml)
were found to be considerably higher than those after intra-
venous administration (29.5 ± 7.4 ng/ml). Their results indi-
cate that nasal delivery could be a more optimal delivery
option targeting of drugs to the CNS directly, making it more
likely for estradiol to reach the brain [103]. Estradiol loaded
NPs have also been employed by Mittal and associates who
encapsulated estradiol in PLGA NPs for oral administration
to study the impact of polymer molecular weight and co-
polymer composition on release behavior in vivo and in vi-
tro. Fine tuning these two variables yielded a 10-fold in-
crease in the bioavailability of the drug as compared with the
free drug [104]. The same group also loaded estradiol in
PLGA NPs coated with tween-80 (T-80) via single emulsion
technique to study the effect of surface coating on the NPs.
24 h after oral administration, T-80 coated PLGA NPs re-
sulted in a significantly higher brain estradiol levels (1.969 ±
0.197 ng/g tissue) when compared to non-coated PLGA NPs
(1.105 ± 0.136 ng/g tissue). Comparing the efficacy of both
NPs also revealed a successful inhibition of Aβ42 im-
munoreactivity expression in the hippocampus area for
coated NPs as opposed to non-coated NPs, indicating the
importance of NP surface coating for brain drug delivery
[105].
5.2. Nano-enabled Anti-tau Strategies
Tau aggregation is closely linked with clinical manifesta-
tion of AD symptoms [106]. Yet, to the best of our knowl-
edge, nano-encapsulation of tau-inhibitors is still scarce.
Several approaches have employed nanoparticles and loaded
them with anti-tau agents to study their therapeutic effect.
Among them, methylene blue (MB), also known as meth-
ylthioninium chloride, is considered as a redox regulator and
a tau aggregation inhibitor. MB has been shown to delay the
progression of AD along with other tauopathies. Nonethe-
less, it suffers from limited brain availability due to its
highly hydrophilic nature [107]. Taking this into considera-
tion, glutathione coated hydrophobic poly-(lactide-co-
glycolide) (PLGA-b-PEG) NPs have been utilized to in-
crease its brain availability. These nano-carriers were found
suitable for CNS penetration. Transwell in vitro BBB model
studies revealed a greater BBB penetration of drug loaded
NPs compared to the drug solution alone over 48 hours. Syn-
thesized NPs showed a steady and sustained drug release
profile (up to 144 hrs) with no initial burst release effect.
Therapeutically, MB-NP administration was tested on two
different AD cell culture models expressing the tau protein.
According to their results, MB-NPs led to a significant re-
duction in both endogenous and over-expressed tau protein
levels, and thus could offer an alternative solution to AD
638 Current Drug Targets, 2020, Vol. 21, No. 7 Altinoglu and Adali
therapy [108]. Chen et al and colleagues have also encapsu-
lated MB in CeNC/IONC/MSN-T807 nanocomposite capa-
ble of targeting tau pathology owing to its high affinity for
the hyper-phosphorylated state of tau. In vitro and in vivo
tests showed a reduction not only in tau hyper-
phosphorylation but also in mitochondrial oxidative stress
and neuronal loss. Additionally, behavioral measures re-
vealed a considerable memory reversal of AD rats upon NP
administration, indicating its potential as a tau-focused
treatment modality [106]. Considering the involvement of
protein misfolding in protein aggregation, targeting molecu-
lar chaperones might represent a promising therapeutic ap-
proach. Among these, heat shock protein 70 (Hsp 70), which
has a crucial role in preventing protein misfolding, has re-
ceived great attention as a potential target in AD pathogene-
sis [109]. MKT-077 is one such drug that has shown promis-
ing results in AD pathology in vitro and ex vivo through a
tau-mediated pathway by targeting Hsp70. It is a highly wa-
ter soluble rhodacyanine dye previously known as FJ-776
and implicated in possessing anti-cancerous effects [110].
Yet, its potential use as an anti-AD agent is restricted due to
its limited passage across the BBB and renal toxicity. To
overcome these issues, MKT-077 loaded PEG-PLGA NPs
coated in 2% glutathione were synthesized. These NPs have
yielded a successful permeation across a Transwell in vitro
BBB model, with a higher permeability than the MKT-077
solution alone over 48 hours. Also, MKT-077 NPs demon-
strated a sustained drug release and tau reduction in in vitro
experimental models [111], holding a promise for AD and
other tau-related disorders. Similarly, Nicotinamide, the am-
ide form of vitamin B3, appears to have neuroprotective ef-
fects and has been associated with neuronal development and
survival [112]. Preclinical studies displayed an enhancement
of cognition following nicotinamide treatment. Thus, the
effectiveness of nicotinamide therapy on AD progression has
been evaluated and established as successful [113]. How-
ever, despite its ability to readily cross the BBB, nicotina-
mide suffers from a sink condition and is only available at
relatively low concentrations, making it necessary to take
multiple doses per day [114]. As a solution, a nano-based
delivery system has been employed. Nicotinamide loaded
SLNs were prepared by Vakilinezhad and associates, and
functionalized with one of the three different coatings;
polysorbate 80, phosphatidylserine or phosphatidic acid.
Their results confirmed the benefits of functionalization in
improving brain bio-distribution. In vitro cytotoxicity tests
also indicated the safety of these particles excluding the
polysorbate 80 coated NPs. Reduction in tau hyper-
phosphorylation and conservation of neuronal cells in AD
rats were demonstrated for phosphatidylserine coated
Nicotinamide-NPs in parallel with the results of spatial and
memory test [114]. Based on these reasons, these NPs can be
used as delivery vehicles to overcome the limitation of con-
ventional nicotinamide administration and target AD more
effectively.
5.3. Nano-enabled Cholinergic Strategies
As discussed earlier, the dysfunction of the cholinergic
system is considered to have a significant influence on the
learning and memory impairments of AD patients [115]. The
enhancement of cholinergic transmission via acetylcho-
linesterase (AChE) inhibition is the most established thera-
peutic approach for symptomatic AD treatment [107]. Rivas-
tigmine, an FDA-approved non-competitive and reversible
inhibitor of AChE enzyme, is one of the few drugs that are
commonly used among AD patients for long-term sympto-
matic treatment. Yet, its clinical efficacy is still limited
mostly due to poor brain uptake that leads to adverse cho-
linergic effects on peripheral organs, making it a target can-
didate for nano-encapsulation [116]. For this reason, many
studies have encapsulated rivastigmine in NPs aiming to
enhance its brain delivery and overcome the above men-
tioned limitations. Wilson et al prepared polysorbate 80-
coated Poly(n-butylcyano-acrylate) (PnBCA) NPs by emul-
sion polymerization method to improve the uptake and
bioavailability of rivastigmine via intravenous injection into
rats. A 3.82-fold increase in the uptake of the drug was ob-
served when compared with its administration as a free drug
[117]. A similar study by Joshi et al who encapsulated rivas-
tigmine not only with PnBCA NPs but also with
poly(lactide-co-glycolide) (PLGA) NPs reported improved
uptake of rivastigmine within the brain compartment. They
further observed a positive therapeutic outcome with a faster
memory regain on scopolamine-induced amnesic mice [118].
Furthermore, many studies have employed chitosan NPs to
load rivastigmine for AD therapy due to its ease of manufac-
ture and reduced cost compared to other biodegradable
polymers [119]. Fazil et al synthesized chitosan loaded ri-
vastigmine NPs using ionic gelation technique for intranasal
administration to increase drug transport efficiency across
the BBB. The findings revealed a higher transport efficiency
(355 ± 13.52%) with direct transport (71.80 ± 6.71%) in
comparison to other formulations employed [120]. Various
other experiments also showed that chitosan NPs can be em-
ployed to increase the brain concentrations of drugs via the
intranasal route [103, 121, 122]. These findings indicate the
suitability of employing AChEI-loaded NPs due to better
brain targeting efficiency. Further studies are required to
support the efficiency of such formulations in vivo.
Not only rivastigmine but many other AChE inhibitors
have also been encapsulated in NPs for the same purposes.
Tacrine, another AChE inhibitor, was the first drug to be
approved for AD treatment in 1993 [123]. However, due to
poor tolerability, tacrine use was discontinued. Nonetheless,
nano-mediated brain targeting of tacrine has been investi-
gated. Tacrine loaded PnBCA NPs were synthesized by
emulsion polymerization process by varying drug polymer
ratios. In vivo results showed that brain concentrations of
intravenously injected 1% polysorbate-80 coated tacrine NPs
were improved up to 4.07 fold compared to the free drug
[124]. Additionally, polysorbate 80 coated NPs demonstrated
a higher brain concentration of the drug in comparison to
non-coated NPs. In a similar study, Wilson et al have devel-
oped tacrine loaded chitosan NPs and tested its bio-
distribution on rats after intravenous injection. Their findings
revealed that NPs coated with 1% polysorbate 80 enhanced
brain drug concentrations, with minimal reticuloendothelial
system uptake and long half-lives [125], highlighting the
importance of this coating in brain drug delivery. According
to Sun and colleagues, brain targeting is achieved via an in-
teraction between polysorbate 80 coating and brain micro-
vessel endothelial cells making polysorbate-80 necessary for
Alzheimer’s Disease Targeted Nano-Based Drug Delivery Systems Current Drug Targets, 2020, Vol. 21, No. 7 639
NP brain delivery [126]. To overcome the limitations of
BBB, AChEI drug Galantamine, has also been encapsulated
in NPs to enhance its bioavailability in the brain. Galan-
tamine was successfully delivered to the brain following its
intranasal administration in hydro-bromide incorporated chi-
tosan NPs with higher transport efficiency in comparison to
its oral administration, with no toxicity issues [119]. More
recently, galantamine loaded thiolated chitosan NPs were
successfully delivered to the brain and demonstrated a sig-
nificant recovery in amnesia induced mice [127]. Apart from
AChEIs, the effects of direct acetylcholine brain delivery
have been studied. In a recent study, Fan L et al and associ-
ates were the first to demonstrate that acetylcholine-loaded
human serum albumin nanoparticles might lead to a better
therapeutic outcome with enhanced spatial learning and
memory, while decreasing oxidative stress in mice [128].
The discussed formulations prove that nano-based delivery
of AChE inhibitors and ACh itself have the potential to be
developed as a treatment option due to better brain targeting
efficiency.
5.4. Nano-enabled Antioxidant Strategies
Another method for AD therapy is focused on the brain
delivery of antioxidants due to their ability to neutralize oxi-
dative stress mediated free radicals and association with the
prevention of neurodegenerative diseases [37]. However,
most antioxidants have limited bioavailability due to being
rapidly metabolized and eliminated from the body. One po-
tential approach is the use of antioxidant-loaded NPs for
brain-specific delivery. Curcumin, a yellow curry spice ob-
tained from the rhizome of Curcuma species, has strong neu-
roprotective, anti-oxidant and anti-inflammatory properties
besides other important medicinal values [36]. In vitro and in
vivo studies have also confirmed that curcumin has positive
effects on AD pathology. It decreases amyloid beta forma-
tion from APP protein, interacts with metal ions and inhibit
metal induced Aβ formation, inhibits the aggregation of Aβ,
and destabilize preformed Aβ fibrils [129-132]. Based on
these reasons, Sood et al have encapsulated curcu-
min/donepezil with solid lipid NPs (SLNs). Higher brain
concentrations of the drug after intranasal administration
were also supported by behavioral experiments on rats which
showed enhanced learning and memory behaviors [133].
Kakkar and associates also loaded curcumin into SLNs and
evaluated their brain targeting via the oral route in rats. Cur-
cumin loaded SLNs revealed a 2-fold increase of curcumin
concentration within the brain and improved AChE activity
when compared with the free drug [134]. Not only lipid-
based NPs but also polymeric NPs such as PnBCA NPs have
been employed to encapsulate curcumin. The encapsulation
process significantly improved the curcumin circulation time
and concentration in the brain in comparison to curcumin
alone, although its biological activity had not been evaluated
[135, 136]. Resveratrol, a natural polyphenol found in the
seeds and skins of grapes and red wine [137], is another
agent that received increased attention due to its antioxidant,
anti-inflammatory [138], and neuroprotective properties
against Aβ induced toxicity [139]. However, as with curcu-
min, resveratrol is rapidly metabolized and fails to reach the
target organs. Therefore, it is essential to stabilize resveratrol
to preserve its biological activity and to increase its bioavail-
ability in the CNS. To this end, Frozza et al developed res-
veratrol loaded SLNs to increase its brain concentration as a
result of the nano-encapsulation process. They demonstrated
a 3 to 6-fold increase in resveratrol concentration in the
brain, liver and kidneys along with improvements in memory
impairments in mouse models [140]. Lu and co-workers also
prepared a nano-particulate delivery system of resveratrol in
AD. They loaded resveratrol in PEGylated poly-caprolactone
(PCL) NPs and showed that after 48 hours, the encapsulation
process could protect PC12 cells against Aβ and were non-
toxic, as compared to the ineffective free resveratrol [141].
Another important potential antioxidant in AD therapy is
ferulic acid. It is a natural and abundant phenolic found in
plant cell walls. Bondi et al. synthesized ferulic acid-SLNs
and demonstrated that ferulic acid loaded SLNs were able to
cause a greater reduction in radical oxygen species in com-
parison to cells treated with free ferulic acid [142]. It is im-
portant to note that although the protective effects of these
antioxidant compounds are well-established, no human clini-
cal trials have been completed regarding their role in AD
therapy.
6. LIMITATIONS OF USING NANO-DELIVERY SYS-
TEMS IN AD
Despite the fact that NPs hold a significant promise as
innovative drug delivery systems, they face some issues re-
garding their use. Initially, it is important to note that in vitro
and in vivo experimental models currently used for NP stud-
ies are significant oversimplifications of real physiological
systems under study. Therefore, these models are limited to
the study of such complex NP interactions [143]. One should
be cautious of the effects of NPs on disease pathology, as a
successful delivery of drug loaded NPs across the BBB is not
predictive of their biological activity [39]. AD is a highly
heterogeneous and multi-variate disorder with multiple hy-
potheses, and testing the impact of treatment might be a
challenge across a wide range of pathways. Potentially, NPs
have the capacity to interact and influence the physiology of
any given cell in the body [144, 145]. Therefore, it is impor-
tant to develop systems that only remotely release the drug
on entering the CNS [146]. Additionally, systemic admini-
stration of NPs generates non-specific interactions between
the NP surface and biomolecules in the bloodstream leading
to their adsorption onto the NP surface, forming a layer
called the protein corona [147]. This layer may change the
morphology and surface characteristics of NPs, which has an
influence on their cellular uptake and interaction with AD
pathology [148], further complicating the use of NPs. To
overcome this limitation, further modifications at the NP
surface can be performed (e.g. PEGylating the surface). The
potential toxicity of NPs is another issue that needs to be
evaluated carefully. Clinical data on NP toxicity are still
scant, and the existing ones are primarily on non-primate
animals. The majority of these studies have focused on short-
term NP exposure, with very few reports on long-term ef-
fects and toxicity [34], that should be addressed in future
studies. Furthermore, certain NPs are not easily removed
from the body. This increases their likelihood of accumula-
tion that could interfere with blood flow or further aggravate
disease progression [149]. To this end, benefit-to-risk ratio
of using NPs as drug delivery systems for neurodegenerative
640 Current Drug Targets, 2020, Vol. 21, No. 7 Altinoglu and Adali
diseases like AD should be thoroughly assessed from a
therapeutic point of view, addressing all of these issues in
future studies.
7. AUTHORS INSIGHT ON THE TOPIC
The application of nanotechnology is increasing rapidly
by addressing the shortcomings of conventional therapy of a
wide range of chronic diseases. Alzheimer’s disease is one
such disorder that can benefit from nanoscale drug delivery
systems. Despite screening numerous drug candidates
against various biological mechanisms of AD, only a few are
currently on the market today. These treatment strategies are
also restricted due to their poor solubility, low bioavailability
and low permeability across the BBB. Therefore, one can
come to the conclusion that it may not entirely be the drugs
that are failing to target AD, but also the way they are deliv-
ered. Current advances in nanotechnology offer the potential
to overcome these limitations. Apart from drugs, other novel
therapeutic agents (e.g. peptides, hormones, antioxidants)
have also revealed significant promise in targeting AD and
thus should not be overlooked. However, extensive clinical
trials are still required to examine the effect of NPs in hu-
mans.
CONCLUSION
AD is the most common neurodegenerative disorder,
affecting the lives of approximately 50 million individuals
worldwide with a substantial impact on socio-economy. This
number is expected to double every 20 years [150], empha-
sizing the urgent need to develop new and effective treat-
ment strategies against AD. Yet, the challenges posed by the
BBB and CNS defence mechanisms significantly limit the
bioavailability of potentially effective therapeutic agents.
Nanotechnology-based approaches may provide a solution to
these limitations and thus hold a great potential as drug de-
livery mechanisms. In the context of AD, nanoparticles can
cross the BBB and establish a new frontier for drugs and
other neuroprotective molecules. Careful design of these
particles offers numerous advantages by facilitating the CNS
delivery of therapeutics with enhanced bioavailability, stabil-
ity and half-life, with controlled and sustained release pro-
files along with decreasing the frequency of dosing and ad-
verse effects [46]. Although very promising, there are still
various challenges concerning the use of NPs as drug deliv-
ery mechanisms. A crucial gap is still waiting to be filled for
improved comprehension of nano-mediated transport of
drugs to the CNS [147] and thus continuous research is es-
sential to successfully employ NPs in AD therapy.
LIST OF ABBREVIATIONS
AcCoA = Acetyl Coenzyme A
AChE = Acetylcholinesterase
AChEIs = Acetylcholinesterase Inhibitors
AD = Alzheimer’s Disease
ApoE = Apolipoprotein E
APP = Amyloid Precursor Protein
Aβ = Amyloid-beta
BBB = Blood Brain Barrier
ChAT = Choline Acetyltransferase
Ch Transporter = Choline Transporter
BCSFB = Blood Cerebrospinal Fluid Barrier
CNS = Central Nervous System
CSF = Cerebrospinal Fluid
Hsp70 = Heat Shock Protein 70
M1AChR/M2AChR = Muscarinic Acetylcholine Recep-
tors Type 1/2
MB = Methylene Blue
NFTs = Neurofibrillary Tangles
NLCs = Nanostructured Lipid Carriers
NMDAR = NMDA Receptors
nAChR = Nicotinic Acetylcholine Receptors
NPs = Nanoparticles
O/W = Oil-in-Water
PAMAM = Poly(amidoamine) Dendrimers
PASA = Poly(aspartic acid)
PCL = Poly(ε-caprolactone)
PEG = Polyethylene Glycol
PEI = Poly(ethylenimine)
PLA = Poly(lactic acid)
PLGA = Poly (lactic-co-glycolic acid)
PnBCA = Poly(n-butylcyano-acrylate)
PS1 = Presenilin 1
PS2 = Presenilin 2
SLNs = Solid Lipid Nanoparticles
T-80 = Tween-80
TREM2 = Triggering Receptor Expressed on
Myeloid Cells 2
VIP = Vasoactive Intestinal Neuropeptide
W/O = Water-in-Oil
WGA = Wheat Germ Agglutinin
CONSENT FOR PUBLICATION
Not applicable.
FUNDING
None.
CONFLICT OF INTEREST
The authors declare no conflict of interest, financial or
otherwise.
ACKNOWLEDGEMENTS
Declared none.
REFERENCES
[1] Kumar A, Singh A, Ekavali. A review on Alzheimer’s disease
pathophysiology and its management: an update. Pharmacol Rep
2015; 67(2): 195-203.
[http://dx.doi.org/10.1016/j.pharep.2014.09.004] [PMID:
25712639]
[2] Saraiva C, Praça C, Ferreira R, Santos T, Ferreira L, Bernardino L.
Nanoparticle-mediated brain drug delivery: Overcoming blood-
brain barrier to treat neurodegenerative diseases. J Control Release
2016; 235: 34-47.
[http://dx.doi.org/10.1016/j.jconrel.2016.05.044] [PMID:
27208862]
[3] Brookmeyer R, Johnson E, Ziegler-Graham K, Arrighi HM. Fore-
casting the global burden of Alzheimer’s disease. Alzheimers De-
ment 2007; 3(3): 186-91.
[http://dx.doi.org/10.1016/j.jalz.2007.04.381] [PMID: 19595937]
[4] Singh A. Alzheimer’s disease Inhibitors: Current status and future
Alzheimer’s Disease Targeted Nano-Based Drug Delivery Systems Current Drug Targets, 2020, Vol. 21, No. 7 641
prospects. IJPLS 2014; 5(8): 3734-40.
[5] American Psychiatric Association. Diagnostic and Statistical Man-
ual of Mental Disorders: Diagnostic and Statistical Manual of Men-
tal Disorders. 5th ed. Arlington, VA: American Psychiatric Asso-
ciation 2013.
[6] Morris R, Mucke L. Alzheimer’s disease: A needle from the hay-
stack. Nature 2006; 440(7082): 284-5.
[http://dx.doi.org/10.1038/440284a] [PMID: 16541054]
[7] Hugo J, Ganguli M. Dementia and cognitive impairment: epidemi-
ology, diagnosis, and treatment. Clin Geriatr Med 2014; 30(3):
421-42.
[http://dx.doi.org/10.1016/j.cger.2014.04.001] [PMID: 25037289]
[8] Kim J, Basak JM, Holtzman DM. The role of apolipoprotein E in
Alzheimer’s disease. Neuron 2009; 63(3): 287-303.
[http://dx.doi.org/10.1016/j.neuron.2009.06.026] [PMID:
19679070]
[9] Jendresen C, Årskog V, Daws MR, Nilsson LN. The Alzheimer’s
disease risk factors apolipoprotein E and TREM2 are linked in a
receptor signaling pathway. J Neuroinflammation 2017; 14(1): 59.
[http://dx.doi.org/10.1186/s12974-017-0835-4] [PMID: 28320424]
[10] Obulesu M, Somashekhar R, Venu R. Genetics of Alzheimer’s
disease: an insight into presenilins and apolipoprotein E instigated
neurodegeneration. Int J Neurosci 2011; 121(5): 229-36.
[http://dx.doi.org/10.3109/00207454.2010.551432] [PMID:
21545304]
[11] Du X, Wang X, Geng M. Alzheimer’s disease hypothesis and re-
lated therapies. Transl Neurodegener 2018; 7: 2.
[http://dx.doi.org/10.1186/s40035-018-0107-y] [PMID: 29423193]
[12] Craig LA, Hong NS, McDonald RJ. Revisiting the cholinergic
hypothesis in the development of Alzheimer’s disease. Neurosci
Biobehav Rev 2011; 35(6): 1397-409.
[http://dx.doi.org/10.1016/j.neubiorev.2011.03.001] [PMID:
21392524]
[13] Becker RE, Greig NH, Giacobini E. Why do so many drugs for
Alzheimer’s disease fail in development? Time for new methods
and new practices? J Alzheimers Dis 2008; 15(2): 303-25.
[http://dx.doi.org/10.3233/JAD-2008-15213] [PMID: 18953116]
[14] Ansari SA, Satar R, Perveen A, Ashraf GM. Current opinion in
Alzheimer’s disease therapy by nanotechnology-based approaches.
Curr Opin Psychiatry 2017; 30(2): 128-35.
[http://dx.doi.org/10.1097/YCO.0000000000000310] [PMID:
28009724]
[15] Aprahamian I, Stella F, Forlenza OV. New treatment strategies for
Alzheimer’s disease: is there a hope? Indian J Med Res 2013;
138(4): 449-60.
[PMID: 24434253]
[16] Hardy JA, Higgins GA. Alzheimer’s disease: the amyloid cascade
hypothesis. Science 1992; 256(5054): 184-5.
[http://dx.doi.org/10.1126/science.1566067] [PMID: 1566067]
[17] Kametani F, Hasegawa M. Reconsideration of Amyloid Hypothesis
and Tau Hypothesis in Alzheimer’s Disease. Front Neurosci 2018;
12: 25.
[http://dx.doi.org/10.3389/fnins.2018.00025] [PMID: 29440986]
[18] Bharadwaj PR, Dubey AK, Masters CL, Martins RN, Macreadie
IG. Abeta aggregation and possible implications in Alzheimer’s
disease pathogenesis. J Cell Mol Med 2009; 13(3): 412-21.
[http://dx.doi.org/10.1111/j.1582-4934.2009.00609.x] [PMID:
19374683]
[19] Musiek ES, Holtzman DM. Three dimensions of the amyloid hy-
pothesis: time, space and ‘wingmen’. Nat Neurosci 2015; 18(6):
800-6.
[http://dx.doi.org/10.1038/nn.4018] [PMID: 26007213]
[20] Ricciarelli R, Fedele E. The Amyloid Cascade Hypothesis in Alz-
heimer’s Disease: It’s Time to Change Our Mind. Curr Neuro-
pharmacol 2017; 15(6): 926-35.
[http://dx.doi.org/10.2174/1570159X15666170116143743] [PMID:
28093977]
[21] Chételat G, La Joie R, Villain N, et al. Amyloid imaging in cogni-
tively normal individuals, at-risk populations and preclinical Alz-
heimer’s disease. Neuroimage Clin 2013; 2: 356-65.
[http://dx.doi.org/10.1016/j.nicl.2013.02.006] [PMID: 24179789]
[22] Pooler AM, Polydoro M, Wegmann S, Nicholls SB, Spires-Jones
TL, Hyman BT. Propagation of tau pathology in Alzheimer’s dis-
ease: identification of novel therapeutic targets. Alzheimers Res
Ther 2013; 5(5): 49.
[http://dx.doi.org/10.1186/alzrt214] [PMID: 24152385]
[23] Gong CX, Iqbal K. Hyperphosphorylation of microtubule-
associated protein tau: a promising therapeutic target for Alzheimer
disease. Curr Med Chem 2008; 15(23): 2321-8.
[http://dx.doi.org/10.2174/092986708785909111] [PMID:
18855662]
[24] Maccioni RB, Farías G, Morales I, Navarrete L. The revitalized tau
hypothesis on Alzheimer’s disease. Arch Med Res 2010; 41(3):
226-31.
[http://dx.doi.org/10.1016/j.arcmed.2010.03.007] [PMID:
20682182]
[25] Meraz-Ríos MA, Lira-De León KI, Campos-Peña V, De Anda-
Hernández MA, Mena-López R. Tau oligomers and aggregation in
Alzheimer’s disease. J Neurochem 2010; 112(6): 1353-67.
[http://dx.doi.org/10.1111/j.1471-4159.2009.06511.x] [PMID:
19943854]
[26] Albarracin SL, Stab B, Casas Z, et al. Effects of natural antioxi-
dants in neurodegenerative disease. Nutr Neurosci 2012; 15(1): 1-9.
[http://dx.doi.org/10.1179/1476830511Y.0000000028] [PMID:
22305647]
[27] Khan TA, Hassan I, Ahmad A, et al. Recent Updates on the Dy-
namic Association Between Oxidative Stress and Neurodegenera-
tive Disorders. CNS Neurol Disord Drug Targets 2016; 15(3): 310-
20.
[http://dx.doi.org/10.2174/1871527315666160202124518] [PMID:
26831262]
[28] Huang WJ, Zhang X, Chen WW. Role of oxidative stress in Alz-
heimer’s disease. Biomed Rep 2016; 4(5): 519-22.
[http://dx.doi.org/10.3892/br.2016.630] [PMID: 27123241]
[29] Zhang C, Wan X, Zheng X, et al. Dual-functional nanoparticles
targeting amyloid plaques in the brains of Alzheimer’s disease
mice. Biomaterials 2014; 35(1): 456-65.
[http://dx.doi.org/10.1016/j.biomaterials.2013.09.063] [PMID:
24099709]
[30] Terry AV Jr, Buccafusco JJ. The cholinergic hypothesis of age and
Alzheimer’s disease-related cognitive deficits: recent challenges
and their implications for novel drug development. J Pharmacol
Exp Ther 2003; 306(3): 821-7.
[http://dx.doi.org/10.1124/jpet.102.041616] [PMID: 12805474]
[31] Colović MB, Krstić DZ, Lazarević-Pašti TD, Bondžić AM, Vasić
VM. Acetylcholinesterase inhibitors: pharmacology and toxicol-
ogy. Curr Neuropharmacol 2013; 11(3): 315-35.
[http://dx.doi.org/10.2174/1570159X11311030006] [PMID:
24179466]
[32] Bartus RT. Evidence for a direct cholinergic involvement in the
scopolamine-induced amnesia in monkeys: effects of concurrent
administration of physostigmine and methylphenidate with sco-
polamine. Pharmacol Biochem Behav 1978; 9(6): 833-6.
[http://dx.doi.org/10.1016/0091-3057(78)90364-7] [PMID:
106402]
[33] Drachman DA. Memory and cognitive function in man: does the
cholinergic system have a specific role? Neurology 1977; 27(8):
783-90.
[http://dx.doi.org/10.1212/WNL.27.8.783] [PMID: 560649]
[34] Hajipour MJ, Santoso MR, Rezaee F, Aghaverdi H, Mahmoudi M,
Perry G. Advances in Alzheimer’s Diagnosis and Therapy: The
Implications of Nanotechnology. Trends Biotechnol 2017; 35(10):
937-53.
[http://dx.doi.org/10.1016/j.tibtech.2017.06.002] [PMID:
28666544]
[35] Md S, Bhattmisra SK, Zeeshan F, et al. Nano-carrier enabled drug
delivery systems for nose to brain targeting for the treatment of
neurodegenerative disorders. J Drug Deliv Sci Technol 2018; 43:
295-310.
[http://dx.doi.org/10.1016/j.jddst.2017.09.022]
[36] Brambilla D, Le Droumaguet B, Nicolas J, et al. Nanotechnologies
for Alzheimer’s disease: diagnosis, therapy, and safety issues.
Nanomedicine (Lond) 2011; 7(5): 521-40.
[http://dx.doi.org/10.1016/j.nano.2011.03.008] [PMID: 21477665]
[37] Leszek J, Md Ashraf G, Tse WH, et al. Nanotechnology for Alz-
heimer Disease. Curr Alzheimer Res 2017; 14(11): 1182-9.
[http://dx.doi.org/10.2174/1567205014666170203125008] [PMID:
28164767]
[38] Pardridge WM. Blood-brain barrier delivery. Drug Discov Today
2007; 12(1-2): 54-61.
[http://dx.doi.org/10.1016/j.drudis.2006.10.013] [PMID:
17198973]
642 Current Drug Targets, 2020, Vol. 21, No. 7 Altinoglu and Adali
[39] Sahni JK, Doggui S, Ali J, Baboota S, Dao L, Ramassamy C. Neu-
rotherapeutic applications of nanoparticles in Alzheimer’s disease.
J Control Release 2011; 152(2): 208-31.
[http://dx.doi.org/10.1016/j.jconrel.2010.11.033] [PMID:
21134407]
[40] Md S, Mustafa G, Baboota S, Ali J. Nanoneurotherapeutics ap-
proach intended for direct nose to brain delivery. Drug Dev Ind
Pharm 2015; 41(12): 1922-34.
[http://dx.doi.org/10.3109/03639045.2015.1052081] [PMID:
26057769]
[41] Banks WA. Peptides and the blood-brain barrier. Peptides 2015;
72: 16-9.
[http://dx.doi.org/10.1016/j.peptides.2015.03.010] [PMID:
25805003]
[42] Engelhardt B, Sorokin L. The blood-brain and the blood-
cerebrospinal fluid barriers: function and dysfunction. Semin Im-
munopathol 2009; 31(4): 497-511.
[http://dx.doi.org/10.1007/s00281-009-0177-0] [PMID: 19779720]
[43] Gao H. Progress and perspectives on targeting nanoparticles for
brain drug delivery. Acta Pharm Sin B 2016; 6(4): 268-86.
[http://dx.doi.org/10.1016/j.apsb.2016.05.013] [PMID: 27471668]
[44] Pardridge WM. Blood-brain barrier biology and methodology. J
Neurovirol 1999; 5(6): 556-69.
[http://dx.doi.org/10.3109/13550289909021285] [PMID:
10602397]
[45] Graff CL, Pollack GM. Drug transport at the blood-brain barrier
and the choroid plexus. Curr Drug Metab 2004; 5(1): 95-108.
[http://dx.doi.org/10.2174/1389200043489126] [PMID: 14965253]
[46] Fazil M, Shadab , Baboota S, Sahni JK, Ali J. Nanotherapeutics for
Alzheimer’s disease (AD): Past, present and future. J Drug Target
2012; 20(2): 97-113.
[http://dx.doi.org/10.3109/1061186X.2011.607499] [PMID:
22023651]
[47] Birrenbach G, Speiser PP. Polymerized micelles and their use as
adjuvants in immunology. J Pharm Sci 1976; 65(12): 1763-6.
[http://dx.doi.org/10.1002/jps.2600651217] [PMID: 1036442]
[48] Khan I, Saeed K, Khan I. Nanoparticles: Properties, applications
and toxicities. Arab J Chem 2017.
[49] Loureiro JA, Andrade S, Duarte A, et al. Resveratrol and Grape
Extract-loaded Solid Lipid Nanoparticles for the Treatment of Alz-
heimer’s Disease. Molecules 2017; 22(2): 277.
[http://dx.doi.org/10.3390/molecules22020277] [PMID: 28208831]
[50] Wen MM, El-Salamouni NS, El-Refaie WM, et al. Nanotechnol-
ogy-based drug delivery systems for Alzheimer’s disease manage-
ment: Technical, industrial, and clinical challenges. J Control Re-
lease 2017; 245: 95-107.
[http://dx.doi.org/10.1016/j.jconrel.2016.11.025] [PMID:
27889394]
[51] Goldsmith M, Abramovitz L, Peer D. Precision nanomedicine in
neurodegenerative diseases. ACS Nano 2014; 8(3): 1958-65.
[http://dx.doi.org/10.1021/nn501292z] [PMID: 24660817]
[52] Barbu E, Molnàr E, Tsibouklis J, Górecki DC. The potential for
nanoparticle-based drug delivery to the brain: overcoming the
blood-brain barrier. Expert Opin Drug Deliv 2009; 6(6): 553-65.
[http://dx.doi.org/10.1517/17425240902939143] [PMID:
19435406]
[53] Lockman PR, Mumper RJ, Khan MA, Allen DD. Nanoparticle
technology for drug delivery across the blood-brain barrier. Drug
Dev Ind Pharm 2002; 28(1): 1-13.
[http://dx.doi.org/10.1081/DDC-120001481] [PMID: 11858519]
[54] Etame AB, Smith CA, Chan WC, Rutka JT. Design and potential
application of PEGylated gold nanoparticles with size-dependent
permeation through brain microvasculature. Nanomedicine (Lond)
2011; 7(6): 992-1000.
[http://dx.doi.org/10.1016/j.nano.2011.04.004] [PMID: 21616168]
[55] Hanada S, Fujioka K, Inoue Y, Kanaya F, Manome Y, Yamamoto
K. Cell-based in vitro blood-brain barrier model can rapidly evalu-
ate nanoparticles’ brain permeability in association with particle
size and surface modification. Int J Mol Sci 2014; 15(2): 1812-25.
[http://dx.doi.org/10.3390/ijms15021812] [PMID: 24469316]
[56] Sonavane G, Tomoda K, Makino K. Biodistribution of colloidal
gold nanoparticles after intravenous administration: effect of parti-
cle size. Colloids Surf B Biointerfaces 2008; 66(2): 274-80.
[http://dx.doi.org/10.1016/j.colsurfb.2008.07.004] [PMID:
18722754]
[57] Decuzzi P, Godin B, Tanaka T, et al. Size and shape effects in the
biodistribution of intravascularly injected particles. J Control Re-
lease 2010; 141(3): 320-7.
[http://dx.doi.org/10.1016/j.jconrel.2009.10.014] [PMID:
19874859]
[58] Banerjee T, Mitra S, Kumar Singh A, Kumar Sharma R, Maitra A.
Preparation, characterization and biodistribution of ultrafine chito-
san nanoparticles. Int J Pharm 2002; 243(1-2): 93-105.
[http://dx.doi.org/10.1016/S0378-5173(02)00267-3] [PMID:
12176298]
[59] Singh R, Lillard JW Jr. Nanoparticle-based targeted drug delivery.
Exp Mol Pathol 2009; 86(3): 215-23.
[http://dx.doi.org/10.1016/j.yexmp.2008.12.004] [PMID:
19186176]
[60] Redhead HM, Davis SS, Illum L. Drug delivery in poly(lactide-co-
glycolide) nanoparticles surface modified with poloxamer 407 and
poloxamine 908: in vitro characterisation and in vivo evaluation. J
Control Release 2001; 70(3): 353-63.
[http://dx.doi.org/10.1016/S0168-3659(00)00367-9] [PMID:
11182205]
[61] Pan H, Marsh JN, Christenson ET, et al. Postformulation peptide
drug loading of nanostructures. Methods Enzymol 2012; 508: 17-
39.
[http://dx.doi.org/10.1016/B978-0-12-391860-4.00002-1] [PMID:
22449919]
[62] Hans ML, Lowman AM. Biodegradable nanoparticles for drug
delivery and targeting. Curr Opin Solid St M 2002; 6(4): 319-27.
[http://dx.doi.org/10.1016/S1359-0286(02)00117-1]
[63] Bramini M, Ye D, Hallerbach A, et al. Imaging approach to
mechanistic study of nanoparticle interactions with the blood-brain
barrier. ACS Nano 2014; 8(5): 4304-12.
[http://dx.doi.org/10.1021/nn5018523] [PMID: 24773217]
[64] Choi CHJ, Alabi CA, Webster P, Davis ME. Mechanism of active
targeting in solid tumors with transferrin-containing gold nanopar-
ticles. Proc Natl Acad Sci USA 2010; 107(3): 1235-40.
[http://dx.doi.org/10.1073/pnas.0914140107] [PMID: 20080552]
[65] Wiley DT, Webster P, Gale A, Davis ME. Transcytosis and brain
uptake of transferrin-containing nanoparticles by tuning avidity to
transferrin receptor. Proc Natl Acad Sci USA 2013; 110(21): 8662-
7.
[http://dx.doi.org/10.1073/pnas.1307152110] [PMID: 23650374]
[66] Jallouli Y, Paillard A, Chang J, Sevin E, Betbeder D. Influence of
surface charge and inner composition of porous nanoparticles to
cross blood-brain barrier in vitro . Int J Pharm 2007; 344(1-2): 103-
9.
[http://dx.doi.org/10.1016/j.ijpharm.2007.06.023] [PMID:
17651930]
[67] Gao X, Qian J, Zheng S, et al. Overcoming the blood-brain barrier
for delivering drugs into the brain by using adenosine receptor
nanoagonist. ACS Nano 2014; 8(4): 3678-89.
[http://dx.doi.org/10.1021/nn5003375] [PMID: 24673594]
[68] Arvizo RR, Miranda OR, Moyano DF, et al. Modulating pharma-
cokinetics, tumor uptake and biodistribution by engineered
nanoparticles. PLoS One 2011; 6(9)e24374
[http://dx.doi.org/10.1371/journal.pone.0024374] [PMID:
21931696]
[69] Doggui S, Dao L, Ramassamy C. Potential of drug-loaded nanopar-
ticles for Alzheimer’s disease: diagnosis, prevention and treatment.
Ther Deliv 2012; 3(9): 1025-7.
[http://dx.doi.org/10.4155/tde.12.84] [PMID: 23035588]
[70] Cupaioli FA, Zucca FA, Boraschi D, Zecca L. Engineered nanopar-
ticles. How brain friendly is this new guest? Prog Neurobiol 2014;
119-120: 20-38.
[http://dx.doi.org/10.1016/j.pneurobio.2014.05.002] [PMID:
24820405]
[71] Naahidi S, Jafari M, Edalat F, Raymond K, Khademhosseini A,
Chen P. Biocompatibility of engineered nanoparticles for drug
delivery. J Control Release 2013; 166(2): 182-94.
[http://dx.doi.org/10.1016/j.jconrel.2012.12.013] [PMID:
23262199]
[72] Karatas H, Aktas Y, Gursoy-Ozdemir Y, et al. A nanomedicine
transports a peptide caspase-3 inhibitor across the blood-brain bar-
rier and provides neuroprotection. J Neurosci 2009; 29(44): 13761-
9.
[http://dx.doi.org/10.1523/JNEUROSCI.4246-09.2009] [PMID:
19889988]
[73] Cheng CJ, Tietjen GT, Saucier-Sawyer JK, Saltzman WM. A holis-
Alzheimer’s Disease Targeted Nano-Based Drug Delivery Systems Current Drug Targets, 2020, Vol. 21, No. 7 643
tic approach to targeting disease with polymeric nanoparticles. Nat
Rev Drug Discov 2015; 14(4): 239-47.
[http://dx.doi.org/10.1038/nrd4503] [PMID: 25598505]
[74] Silva AC, Santos D, Ferreira D, Souto EB, Nalwa HS. In: Encyclo-
pedia of Nanoscience and Nanotechnology; H.S Nalwa, Ed.
American: Scientific. 2011; pp. 1-14.
[75] Gastaldi L, Battaglia L, Peira E, et al. Solid lipid nanoparticles as
vehicles of drugs to the brain: current state of the art. Eur J Pharm
Biopharm 2014; 87(3): 433-44.
[http://dx.doi.org/10.1016/j.ejpb.2014.05.004] [PMID: 24833004]
[76] Mishra B, Patel BB, Tiwari S. Colloidal nanocarriers: a review on
formulation technology, types and applications toward targeted
drug delivery. Nanomedicine (Lond) 2010; 6(1): 9-24.
[http://dx.doi.org/10.1016/j.nano.2009.04.008] [PMID: 19447208]
[77] Kaur IP, Bhandari R, Bhandari S, Kakkar V. Potential of solid lipid
nanoparticles in brain targeting. J Control Release 2008; 127(2):
97-109.
[http://dx.doi.org/10.1016/j.jconrel.2007.12.018] [PMID:
18313785]
[78] Neves AR, Queiroz JF, Weksler B, Romero IA, Couraud PO, Reis
S. Solid lipid nanoparticles as a vehicle for brain-targeted drug de-
livery: two new strategies of functionalization with apolipoprotein
E. Nanotechnology 2015; 26(49)495103
[http://dx.doi.org/10.1088/0957-4484/26/49/495103] [PMID:
26574295]
[79] Robinson M, Lee BY, Leonenko Z. Drugs and drug delivery sys-
tems targeting amyloid- β in Alzheimer’s disease. AIMS Mol Sci
2015; 2(3): 332-58.
[http://dx.doi.org/10.3934/molsci.2015.3.332]
[80] Fang CL, Al-Suwayeh SA, Fang JY. Nanostructured lipid carriers
(NLCs) for drug delivery and targeting. Recent Pat Nanotechnol
2013; 7(1): 41-55.
[http://dx.doi.org/10.2174/187221013804484827] [PMID:
22946628]
[81] Hadavi D, Poot AA. Biomaterials for the treatment of Alzheimer’s
disease. Front Bioeng Biotechnol 2016; 4: 49.
[http://dx.doi.org/10.3389/fbioe.2016.00049] [PMID: 27379232]
[82] Kaur IP, Garg A, Singla AK, Aggarwal D. Vesicular systems in
ocular drug delivery: an overview. Int J Pharm 2004; 269(1): 1-14.
[http://dx.doi.org/10.1016/j.ijpharm.2003.09.016] [PMID:
14698571]
[83] Gulati M, Grover M, Singh S, Singh M. Lipophilic drug derivatives
in liposomes. Int J Pharm 1998; 165(2): 129-68.
[http://dx.doi.org/10.1016/S0378-5173(98)00006-4]
[84] Fonseca-Santos B, Gremião MP, Chorilli M. Nanotechnology-
based drug delivery systems for the treatment of Alzheimer’s dis-
ease. Int J Nanomedicine 2015; 10: 4981-5003.
[http://dx.doi.org/10.2147/IJN.S87148] [PMID: 26345528]
[85] Fernandes C, Soni U, Patravale V. Nano-interventions for neurode-
generative disorders. Pharmacol Res 2010; 62(2): 166-78.
[http://dx.doi.org/10.1016/j.phrs.2010.02.004] [PMID: 20153429]
[86] Silva AC, González-Mira E, Lobo JM, Amaral MH. Current pro-
gresses on nanodelivery systems for the treatment of neuropsy-
chiatric diseases: Alzheimer’s and schizophrenia. Curr Pharm Des
2013; 19(41): 7185-95.
[http://dx.doi.org/10.2174/138161281941131219123329] [PMID:
23489198]
[87] Pathak K, Pattnaik S, Swain K. Nanoemulsions; SM Jafari. Aca-
demic Press 2018; pp. 415-33.
[http://dx.doi.org/10.1016/B978-0-12-811838-2.00013-8]
[88] Salomone S, Caraci F, Leggio GM, Fedotova J, Drago F. New
pharmacological strategies for treatment of Alzheimer’s disease:
focus on disease modifying drug s. Br J Clin Pharm acol 2 012;
73(4): 504-17.
[http://dx.doi.org/10.1111/j.1365-2125.2011.04134.x] [PMID:
22035455]
[89] Cummings JL. Defining and labeling disease-modifying treatments
for Alzheimer’s disease. Alzheimers Dement 2009; 5(5): 406-18.
[http://dx.doi.org/10.1016/j.jalz.2008.12.003] [PMID: 19751920]
[90] Nazem A, Mansoori GA. Nanotechnology solutions for Alz-
heimer’s disease: advances in research tools, diagnostic methods
and therapeutic agents. J Alzheimers Dis 2008; 13(2): 199-223.
[http://dx.doi.org/10.3233/JAD-2008-13210] [PMID: 18376062]
[91] Songjiang Z, Lixiang W. Amyloid-beta associated with chitosan
nano-carrier has favorable immunogenicity and permeates the
BBB. AAPS PharmSciTech 2009; 10(3): 900-5.
[http://dx.doi.org/10.1208/s12249-009-9279-1] [PMID: 19609682]
[92] Mohammed MA, Syeda JTM, Wasan KM, Wasan EK. An Over-
view of Chitosan Nanoparticles and Its Application in Non-
Parenteral Drug Delivery. Pharmaceutics 2017; 9(4)E53
[http://dx.doi.org/10.3390/pharmaceutics9040053] [PMID:
29156634]
[93] Permanne B, Adessi C, Saborio GP, et al. Reduction of amyloid
load and cerebral damage in a transgenic mouse model of Alz-
heimer’s disease by treatment with a beta-sheet breaker peptide.
FASEB J 2002; 16(8): 860-2.
[http://dx.doi.org/10.1096/fj.01-0841fje] [PMID: 11967228]
[94] Loureiro JA, Gomes B, Fricker G, Coelho MAN, Rocha S, Pereira
MC. Cellular uptake of PLGA nanoparticles targeted with anti-
amyloid and anti-transferrin receptor antibodies for Alzheimer’s
disease treatment. Colloids Surf B Biointerfaces 2016; 145: 8-13.
[http://dx.doi.org/10.1016/j.colsurfb.2016.04.041] [PMID:
27131092]
[95] Bala I, Hariharan S, Kumar MN. PLGA nanoparticles in drug de-
livery: the state of the art. Crit Rev Ther Drug Carrier Syst 2004;
21(5): 387-422.
[http://dx.doi.org/10.1615/CritRevTherDrugCarrierSyst.v21.i5.20]
[PMID: 15719481]
[96] Fahrenkrug J. Vasoactive intestinal polypeptide: measurement,
distribution and putative neurotransmitter function. Digestion 1979;
19(3): 149-69.
[http://dx.doi.org/10.1159/000198339] [PMID: 39014]
[97] Brenneman DE, Glazner G, Hill JM, Hauser J, Davidson A, Gozes
I. VIP neurotrophism in the central nervous system: multiple effec-
tors and identification of a femtomolar-acting neuroprotective pep-
tide. Ann N Y Acad Sci 1998; 865: 207-12.
[http://dx.doi.org/10.1111/j.1749-6632.1998.tb11180.x] [PMID:
9928014]
[98] Gozes I, Divinsky I, Pilzer I, Fridkin M, Brenneman DE, Spier AD.
From vasoactive intestinal peptide (VIP) through activity-
dependent neuroprotective protein (ADNP) to NAP: a view of neu-
roprotection and cell division. J Mol Neurosci 2003; 20(3): 315-22.
[http://dx.doi.org/10.1385/JMN:20:3:315] [PMID: 14501014]
[99] Gao X, Wu B, Zhang Q, et al. Brain delivery of vasoactive intesti-
nal peptide enhanced with the nanoparticles conjugated with wheat
germ agglutinin following intranasal administration. J Control Re-
lease 2007; 121(3): 156-67.
[http://dx.doi.org/10.1016/j.jconrel.2007.05.026] [PMID:
17628165]
[100] Kanwar JR, Kanwar RK, Mahidhara G. Recent Advances in
Nanoneurology for Drug Delivery to the Brain. Curr Nanosci 2009;
5(4): 441-7.
[http://dx.doi.org/10.2174/157341309789378014]
[101] Rubinow DR, Schmidt PJ. Gonadal steroids, brain, and behavior:
role of context. Dialogues Clin Neurosci 2002; 4(2): 123-37.
[PMID: 22033695]
[102] Amtul Z, Wang L, Westaway D, Rozmahel RF. Neuroprotective
mechanism conferred by 17beta-estradiol on the biochemical basis
of Alzheimer’s disease. Neuroscience 2010; 169(2): 781-6.
[http://dx.doi.org/10.1016/j.neuroscience.2010.05.031] [PMID:
20493928]
[103] Wang X, Chi N, Tang X. Preparation of estradiol chitosan nanopar-
ticles for improving nasal absorption and brain targeting. Eur J
Pharm Biopharm 2008; 70(3): 735-40.
[http://dx.doi.org/10.1016/j.ejpb.2008.07.005] [PMID: 18684400]
[104] Mittal G, Sahana DK, Bhardwaj V, Ravi Kumar MN. Estradiol
loaded PLGA nanoparticles for oral administration: effect of poly-
mer molecular weight and copolymer composition on release be-
havior in vitro and in vivo. J Control Release 2007; 119(1): 77-85.
[http://dx.doi.org/10.1016/j.jconrel.2007.01.016] [PMID:
17349712]
[105] Mittal G, Carswell H, Brett R, Currie S, Kumar MN. Development
and evaluation of polymer nanoparticles for oral delivery of estra-
diol to rat brain in a model of Alzheimer’s pathology. J Control Re-
lease 2011; 150(2): 220-8.
[http://dx.doi.org/10.1016/j.jconrel.2010.11.013] [PMID:
21111014]
[106] Chen Q, Du Y, Zhang K, et al. Tau-Targeted Multifunctional
Nanocomposite for Combinational Therapy of Alzheimer’s Dis-
ease. ACS Nano 2018; 12(2): 1321-38.
[http://dx.doi.org/10.1021/acsnano.7b07625] [PMID: 29364648]
[107] Karthivashan G, Ganesan P, Park SY, Kim JS, Choi DK. Therapeu-
644 Current Drug Targets, 2020, Vol. 21, No. 7 Altinoglu and Adali
tic strategies and nano-drug delivery applications in management of
ageing Alzheimer’s disease. Drug Deliv 2018; 25(1): 307-20.
[http://dx.doi.org/10.1080/10717544.2018.1428243] [PMID:
29350055]
[108] Jinwal UK, Groshev A, Zhang J, Grover A, Sutariya VB. Prepara-
tion and characterization of methylene blue nanoparticles for Alz-
heimer’s disease and other tauopathies. Curr Drug Deliv 2014;
11(4): 541-50.
[http://dx.doi.org/10.2174/1567201810666131113102037] [PMID:
24237400]
[109] Lu RC, Tan MS, Wang H, Xie AM, Yu JT, Tan L. Heat shock
protein 70 in Alzheimer’s disease. BioMed Res Int 2014;
2014435203
[http://dx.doi.org/10.1155/2014/435203] [PMID: 25431764]
[110] Koya K, Li Y, Wang H, et al. MKT-077, a novel rhodacyanine dye
in clinical trials, exhibits anticarcinoma activity in preclinical stud-
ies based on selective mitochondrial accumulation. Cancer Res
1996; 56(3): 538-43.
[PMID: 8564968]
[111] Jinwal UK, Grover A, Narayan M, Hirani A, Sutariya V. Prepara-
tion and Characterization of MKT-077 Nanoparticles for Treatment
of Alzheimer’s Disease and Other Tauopathies. Pharm Nanotech-
nol 2014; 2(4): 217-26.
[http://dx.doi.org/10.2174/2211738503666150328001726]
[112] Fricker RA, Green EL, Jenkins SI, Griffin SM. The Influence of
Nicotinamide on Health and Disease in the Central Nervous Sys-
tem. Int J Tryptophan Res 2018; 111178646918776658
[http://dx.doi.org/10.1177/1178646918776658] [PMID: 29844677]
[113] Fillit H, Friedman L, Hara Y, Koemeter-Cox A, McKeehan N.
Closing in on a cure - 2017 Alzheimer’s clinical trials report. New
York: Alzheimer Drug Discovery Foundation 2017.
[114] Vakilinezhad MA, Amini A, Akbari Javar H, Baha’addini Beigi
Zarandi BF, Montaseri H, Dinarvand R. Nicotinamide loaded func-
tionalized solid lipid nanoparticles improves cognition in Alz-
heimer’s disease animal model by reducing Tau hyperphosphoryla-
tion. Daru 2018; 26(2): 165-77.
[http://dx.doi.org/10.1007/s40199-018-0221-5] [PMID: 30386982]
[115] Francis PT. The interplay of neurotransmitters in Alzheimer’s
disease. CNS Spectr 2005; 10(11)(Suppl. 18): 6-9.
[http://dx.doi.org/10.1017/S1092852900014164] [PMID:
16273023]
[116] Ahmad J, Akhter S, Rizwanullah M, et al. Nanotechnology Based
Theranostic Approaches in Alzheimer’s Disease Management: Cur-
rent Status and Future Perspective. Curr Alzheimer Res 2017;
14(11): 1164-81.
[http://dx.doi.org/10.2174/1567205014666170508121031] [PMID:
28482786]
[117] Wilson B, Samanta MK, Santhi K, Kumar KP, Paramakrishnan N,
Suresh B. Poly(n-butylcyanoacrylate) nanoparticles coated with
polysorbate 80 for the targeted delivery of rivastigmine into the
brain to treat Alzheimer’s disease. Brain Res 2008; 1200: 159-68.
[http://dx.doi.org/10.1016/j.brainres.2008.01.039] [PMID:
18291351]
[118] Joshi SA, Chavhan SS, Sawant KK. Rivastigmine-loaded PLGA
and PBCA nanoparticles: preparation, optimization, characteriza-
tion, in vitro and pharmacodynamic studies. Eur J Pharm Biopharm
2010; 76(2): 189-99.
[http://dx.doi.org/10.1016/j.ejpb.2010.07.007] [PMID: 20637869]
[119] Hanafy AS, Farid RM, ElGamal SS. Complexation as an approach
to entrap cationic drugs into cationic nanoparticles administered in-
tranasally for Alzheimer’s disease management: preparation and
detection in rat brain. Drug Dev Ind Pharm 2015; 41(12): 2055-68.
[http://dx.doi.org/10.3109/03639045.2015.1062897] [PMID:
26133084]
[120] Fazil M, Md S, Haque S, et al. Development and evaluation of
rivastigmine loaded chitosan nanoparticles for brain targeting. Eur
J Pharm Sci 2012; 47(1): 6-15.
[http://dx.doi.org/10.1016/j.ejps.2012.04.013] [PMID: 22561106]
[121] Bhavna Shadab M, Ali M, Bhatnagar A, Baboota S, Sahni JK, Ali
J. Design, Development, optimization and characterization of
donepezil loaded chitosan nanoparticles for brain targeting to treat
Alzheimer’s disease. Sci Adv Mater 2014; 6(4): 720-35.
[http://dx.doi.org/10.1166/sam.2014.1761]
[122] Alam S, Khan ZI, Mustafa G, et al. Development and evaluation of
thymoquinone-encapsulated chitosan nanoparticles for nose-to-
brain targeting: a pharmacoscintigraphic study. Int J Nanomedicine
2012; 7: 5705-18.
[http://dx.doi.org/10.2147/IJN.S35329] [PMID: 23180965]
[123] Tumiatti V, Minarini A, Bolognesi ML, Milelli A, Rosini M,
Melchiorre C. Tacrine derivatives and Alzheimer’s disease. Curr
Med Chem 2010; 17(17): 1825-38.
[http://dx.doi.org/10.2174/092986710791111206] [PMID:
20345341]
[124] Wilson B, Samanta MK, Santhi K, Kumar KP, Paramakrishnan N,
Suresh B. Targeted delivery of tacrine into the brain with polysor-
bate 80-coated poly(n-butylcyanoacrylate) nanoparticles. Eur J
Pharm Biopharm 2008; 70(1): 75-84.
[http://dx.doi.org/10.1016/j.ejpb.2008.03.009] [PMID: 18472255]
[125] Wilson B, Samanta MK, Santhi K, Kumar KP, Ramasamy M,
Suresh B. Chitosan nanoparticles as a new delivery system for the
anti-Alzheimer drug tacrine. Nanomedicine (Lond) 2010; 6(1):
144-52.
[http://dx.doi.org/10.1016/j.nano.2009.04.001] [PMID: 19446656]
[126] Sun W, Xie C, Wang H, Hu Y. Specific role of polysorbate 80
coating on the targeting of nanoparticles to the brain. Biomaterials
2004; 25(15): 3065-71.
[http://dx.doi.org/10.1016/j.biomaterials.2003.09.087] [PMID:
14967540]
[127] Sunena, Singh SK, Mishra DN. Nose to Brain Delivery of Galan-
tamine Loaded Nanoparticles: In-vivo Pharmacodynamic and Bio-
chemical Study in Mice. Curr Drug Deliv 2019; 16(1): 51-8.
[http://dx.doi.org/10.2174/1567201815666181004094707] [PMID:
30289074]
[128] Fan L, Wang J, Meng F, et al. Delivering the Acetylcholine Neuro-
transmitter by Nanodrugs as an Effective Treatment for Alz-
heimer’s Disease. J Biomed Nanotechnol 2018; 14(12): 2066-76.
[http://dx.doi.org/10.1166/jbn.2018.2649] [PMID: 30305214]
[129] Huang HC, Chang P, Dai XL, Jiang ZF. Protective effects of cur-
cumin on amyloid-β-induced neuronal oxidative damage. Neuro-
chem Res 2012; 37(7): 1584-97.
[http://dx.doi.org/10.1007/s11064-012-0754-9] [PMID: 22476982]
[130] Zhao LN, Chiu SW, Benoit J, Chew LY, Mu Y. The effect of cur-
cumin on the stability of Aβ dimers. J Phys Chem B 2012; 116(25):
7428-35.
[http://dx.doi.org/10.1021/jp3034209] [PMID: 22690789]
[131] Baum L, Ng A. Curcumin interaction with copper and iron suggests
one possible mechanism of action in Alzheimer’s disease animal
models. J Alzheimers Dis 2004; 6(4): 367-77.
[http://dx.doi.org/10.3233/JAD-2004-6403] [PMID: 15345806]
[132] Daniel S, Limson JL, Dairam A, Watkins GM, Daya S. Through
metal binding, curcumin protects against lead- and cadmium-
induced lipid peroxidation in rat brain homogenates and against
lead-induced tissue damage in rat brain. J Inorg Biochem 2004;
98(2): 266-75.
[http://dx.doi.org/10.1016/j.jinorgbio.2003.10.014] [PMID:
14729307]
[133] Sood S, Jain K, Gowthamarajan K. Curcumin-donepezilloaded
nanostructured lipid carriers for intranasal delivery in an Alz-
heimer’s disease model. Alzheimers Dement 2013; 9(4): 299.
[http://dx.doi.org/10.1016/j.jalz.2013.05.609]
[134] Kakkar V, Muppu SK, Chopra K, Kaur IP. Curcumin loaded solid
lipid nanoparticles: an efficient formulation approach for cerebral
ischemic reperfusion injury in rats. Eur J Pharm Biopharm 2013;
85(3 Pt A): 339-45.
[http://dx.doi.org/10.1016/j.ejpb.2013.02.005] [PMID: 23454202]
[135] Sun M, Gao Y, Guo C, et al. Enhancement of transport of curcumin
to brain in mice by poly(n-butylcyanoacrylate) nanoparticle. J
Nanopart Res 2010; 12(8): 3111-22.
[http://dx.doi.org/10.1007/s11051-010-9907-4]
[136] Mulik R, Mahadik K, Paradkar A. Development of curcuminoids
loaded poly(butyl) cyanoacrylate nanoparticles: Physicochemical
characterization and stability study. Eur J Pharm Sci 2009; 37(3-4):
395-404.
[http://dx.doi.org/10.1016/j.ejps.2009.03.009] [PMID: 19491031]
[137] Markus MA, Morris BJ. Resveratrol in prevention and treatment of
common clinical conditions of aging. Clin Interv Aging 2008; 3(2):
331-9.
[PMID: 18686754]
[138] Soleas GJ, Diamandis EP, Goldberg DM. Resveratrol: a molecule
whose time has come? And gone? Clin Biochem 1997; 30(2): 91-
113.
[http://dx.doi.org/10.1016/S0009-9120(96)00155-5] [PMID:
Alzheimer’s Disease Targeted Nano-Based Drug Delivery Systems Current Drug Targets, 2020, Vol. 21, No. 7 645
9127691]
[139] Jang JH, Surh YJ. Protective effect of resveratrol on beta-amyloid-
induced oxidative PC12 cell death. Free Radic Biol Med 2003;
34(8): 1100-10.
[http://dx.doi.org/10.1016/S0891-5849(03)00062-5] [PMID:
12684095]
[140] Frozza RL, Bernardi A, Paese K, et al. Characterization of trans-
resveratrol-loaded lipid-core nanocapsules and tissue distribution
studies in rats. J Biomed Nanotechnol 2010; 6(6): 694-703.
[http://dx.doi.org/10.1166/jbn.2010.1161] [PMID: 21361135]
[141] Lu X, Ji C, Xu H, et al. Resveratrol-loaded polymeric micelles
protect cells from Abeta-induced oxidative stress. Int J Pharm
2009; 375(1-2): 89-96.
[http://dx.doi.org/10.1016/j.ijpharm.2009.03.021] [PMID:
19481694]
[142] Bondi ML, Montana G, Craparo EF, et al. Ferulic acid-loaded lipid
nanostructures as drug delivery systems for Alzheimer’s disease:
preparation, characterization and cytotoxicity studies. Curr Nanosci
2009; 5(1): 26-32.
[http://dx.doi.org/10.2174/157341309787314656]
[143] Landsiedel R, Fabian E, Ma-Hock L, et al. Toxico-/biokinetics of
nanomaterials. Arch Toxicol 2012; 86(7): 1021-60.
[http://dx.doi.org/10.1007/s00204-012-0858-7] [PMID: 22576463]
[144] Kim JS, Yoon TJ, Yu KN, et al. Toxicity and tissue distribution of
magnetic nanoparticles in mice. Toxicol Sci 2006; 89(1): 338-47.
[http://dx.doi.org/10.1093/toxsci/kfj027] [PMID: 16237191]
[145] Kashiwada S. Distribution of nanoparticles in the see-through
medaka (Oryzias latipes). Environ Health Perspect 2006; 114(11):
1697-702.
[http://dx.doi.org/10.1289/ehp.9209] [PMID: 17107855]
[146] Li W, Luo R, Lin X, et al. Remote modulation of neural activities
via near-infrared triggered release of biomolecules. Biomaterials
2015; 65: 76-85.
[http://dx.doi.org/10.1016/j.biomaterials.2015.06.041] [PMID:
26142778]
[147] Masserini M. Nanoparticles for brain drug delivery. ISRN Biochem
2013; 2013238428
[http://dx.doi.org/10.1155/2013/238428] [PMID: 25937958]
[148] Pietroiusti A, Campagnolo L, Fadeel B. Interactions of engineered
nanoparticles with organs protected by internal biological barriers.
Small 2013; 9(9-10): 1557-72.
[http://dx.doi.org/10.1002/smll.201201463] [PMID: 23097249]
[149] Martin L, Latypova X, Wilson CM, et al. Tau protein kinases:
involvement in Alzheimer’s disease. Ageing Res Rev 2013; 12(1):
289-309.
[http://dx.doi.org/10.1016/j.arr.2012.06.003] [PMID: 22742992]
[150] Report WA. World Alzheimer Report The state of the art of de-
mentia research: New frontiers London: Alzheimer’s Disease In-
ternational (ADI). 2018.
[151] Khemariya RP, Khemariya PS. New-tangled approach in the man-
agement of Alzheimer by formulation of polysorbate 80 coated chi-
tosan nanoparticles of rivastigmine for brain delivery and their in
vivo evaluation. IJRMS 2016; 2(2): 18-29.
[152] Fornaguera C, Feiner-Gracia N, Calderó G, García-Celma MJ,
Solans C. Galantamine-loaded PLGA nanoparticles, from nano-
emulsion templating, as novel advanced drug delivery systems to
treat neurodegenerative diseases. Nanoscale 2015; 7(28): 12076-84.
[http://dx.doi.org/10.1039/C5NR03474D] [PMID: 26118655]
[153] Md S, Ali M, Baboota S, Sahni JK, Bhatnagar A, Ali J. Prepara-
tion, characterization, in vivo biodistribution and pharmacokinetic
studies of donepezil-loaded PLGA nanoparticles for brain target-
ing. Drug Dev Ind Pharm 2014; 40(2): 278-87.
[http://dx.doi.org/10.3109/03639045.2012.758130] [PMID:
23369094]
[154] Baysal I, Ucar G, Gultekinoglu M, Ulubayram K, Yabanoglu-Ciftci
S. Donepezil loaded PLGA-b-PEG nanoparticles: their ability to
induce destabilization of amyloid fibrils and to cross blood brain
barrier in vitro. J Neural Transm (Vienna) 2017; 124(1): 33-45.
[http://dx.doi.org/10.1007/s00702-016-1527-4] [PMID: 26911385]
[155] Agyare EK, Curran GL, Ramakrishnan M, Yu CC, Poduslo JF,
Kandimalla KK. Development of a smart nano-vehicle to target
cerebrovascular amyloid deposits and brain parenchymal plaques
observed in Alzheimer’s disease and cerebral amyloid angiopathy.
Pharm Res 2008; 25(11): 2674-84.
[http://dx.doi.org/10.1007/s11095-008-9688-y] [PMID: 18712585]
[156] Majzik A, Fülöp L, Csapó E, et al. Functionalization of gold
nanoparticles with amino acid, beta-amyloid peptides and frag-
ment. Colloids Surf B Biointerfaces 2010; 81(1): 235-41.
[http://dx.doi.org/10.1016/j.colsurfb.2010.07.011] [PMID:
20674288]
[157] Wang ZH, Wang ZY, Sun CS, Wang CY, Jiang TY, Wang SL.
Trimethylated chitosan-conjugated PLGA nanoparticles for the
delivery of drugs to the brain. Biomaterials 2010; 31(5): 908-15.
[http://dx.doi.org/10.1016/j.biomaterials.2009.09.104] [PMID:
19853292]
[158] Silva-Abreu M, Calpena AC, Andrés-Benito P, et al. PPARγ ago-
nist-loaded PLGA-PEG nanocarriers as a potential treatment for
Alzheimer’s disease: in vitro and in vivo studies. Int J Nanomedi-
cine 2018; 13: 5577-90.
[http://dx.doi.org/10.2147/IJN.S171490] [PMID: 30271148]
[159] Zhang C, Chen J, Feng C, et al. Intranasal nanoparticles of basic
fibroblast growth factor for brain delivery to treat Alzheimer’s dis-
ease. Int J Pharm 2014; 461(1-2): 192-202.
[http://dx.doi.org/10.1016/j.ijpharm.2013.11.049] [PMID:
24300213]
[160] Kurakhmaeva KB, Djindjikhashvili IA, Petrov VE, et al. Brain
targeting of nerve growth factor using poly(butyl cyanoacrylate)
nanoparticles. J Drug Target 2009; 17(8): 564-74.
[http://dx.doi.org/10.1080/10611860903112842] [PMID:
19694610]
[161] Liu Z, Gao X, Kang T, et al. B6 peptide-modified PEG-PLA
nanoparticles for enhanced brain delivery of neuroprotective pep-
tide. Bioconjug Chem 2013; 24(6): 997-1007.
[http://dx.doi.org/10.1021/bc400055h] [PMID: 23718945]
[162] Schaffazick SR, Pohlmann AR, de Cordova CA, Creczynski-Pasa
TB, Guterres SS. Protective properties of melatonin-loaded
nanoparticles against lipid peroxidation. Int J Pharm 2005; 289(1-
2): 209-13.
[http://dx.doi.org/10.1016/j.ijpharm.2004.11.003] [PMID:
15652213]
[163] He W, Horn SW, Hussain MD. Improved bioavailability of orally
administered mifepristone from PLGA nanoparticles. Int J Pharm
2007; 334(1-2): 173-8.
[http://dx.doi.org/10.1016/j.ijpharm.2006.10.025] [PMID:
17101249]
[164] Anand P, Nair HB, Sung B, et al. Design of curcumin-loaded
PLGA nanoparticles formulation with enhanced cellular uptake,
and increased bioactivity in vitro and superior bioavailability in
vivo. Biochem Pharmacol 2010; 79(3): 330-8.
[http://dx.doi.org/10.1016/j.bcp.2009.09.003] [PMID: 19735646]
[165] Mulik RS, Mönkkönen J, Juvonen RO, Mahadik KR, Paradkar AR.
ApoE3 mediated poly(butyl) cyanoacrylate nanoparticles contain-
ing curcumin: study of enhanced activity of curcumin against beta
amyloid induced cytotoxicity using in vitro cell culture model. Mol
Pharm 2010; 7(3): 815-25.
[http://dx.doi.org/10.1021/mp900306x] [PMID: 20230014]
[166] Dube A, Nicolazzo JA, Larson I. Chitosan nanoparticles enhance
the intestinal absorption of the green tea catechins (+)-catechin and
(-)-epigallocatechin gallate. Eur J Pharm Sci 2010; 41(2): 219-25.
[http://dx.doi.org/10.1016/j.ejps.2010.06.010] [PMID: 20600878]
[167] Elnaggar YS, Etman SM, Abdelmonsif DA, Abdallah OY. Intrana-
sal piperine-loaded chitosan nanoparticles as brain-targeted therapy
in Alzheimer’s disease: optimization, biological efficacy, and po-
tential toxicity. J Pharm Sci 2015; 104(10): 3544-56.
[http://dx.doi.org/10.1002/jps.24557]
[168] Liu G, Men P, Kudo W, Perry G, Smith MA. Nanoparticle-chelator
conjugates as inhibitors of amyloid-beta aggregation and neurotox-
icity: a novel therapeutic approach for Alzheimer disease. Neurosci
Lett 2009; 455(3): 187-90.
[http://dx.doi.org/10.1016/j.neulet.2009.03.064] [PMID: 19429118]
[169] Cui Z, Lockman PR, Atwood CS, et al. Novel D-penicillamine
carrying nanoparticles for metal chelation therapy in Alzheimer’s
and other CNS diseases. Eur J Pharm Biopharm 2005; 59(2): 263-
72.
[http://dx.doi.org/10.1016/j.ejpb.2004.07.009] [PMID: 15661498]
[170] Misra S, Chopra K, Sinha VR, Medhi B. Galantamine-loaded solid-
lipid nanoparticles for enhanced brain delivery: preparation, char-
acterization, in vitro and in vivo evaluations. Drug Deliv 2016;
23(4): 1434-43.
[http://dx.doi.org/10.3109/10717544.2015.1089956] [PMID:
26405825]
[171] Shah B, Khunt D, Bhatt H, Misra M, Padh H. Application of qual-
646 Current Drug Targets, 2020, Vol. 21, No. 7 Altinoglu and Adali
ity by design approach for intranasal delivery of rivastigmine
loaded solid lipid nanoparticles: Effect on formulation and charac-
terization parameters. Eur J Pharm Sci 2015; 78: 54-66.
[http://dx.doi.org/10.1016/j.ejps.2015.07.002] [PMID: 26143262]
[172] Picone P, Bondi ML, Montana G, et al. Ferulic acid inhibits oxida-
tive stress and cell death induced by Ab oligomers: improved de-
livery by solid lipid nanoparticles. Free Radic Res 2009; 43(11):
1133-45.
[http://dx.doi.org/10.1080/10715760903214454] [PMID:
19863373]
[173] Smith A, Giunta B, Bickford PC, Fountain M, Tan J, Shytle RD.
Nanolipidic particles improve the bioavailability and alpha-
secretase inducing ability of epigallocatechin-3-gallate (EGCG) for
the treatment of Alzheimer’s disease. Int J Pharm 2010; 389(1-2):
207-12.
[http://dx.doi.org/10.1016/j.ijpharm.2010.01.012] [PMID:
20083179]
[174] Kuo YC, Rajesh R. Nerve growth factor-loaded heparinized cati-
onic solid lipid nanoparticles for regulating membrane charge of
induced pluripotent stem cells during differentiation. Mater Sci Eng
C 2017; 77: 680-9.
[http://dx.doi.org/10.1016/j.msec.2017.03.303] [PMID: 28532079]
[175] Li W, Zhou Y, Zhao N, Hao B, Wang X, Kong P. Pharmacokinetic
behavior and efficiency of acetylcholinesterase inhibition in rat
brain after intranasal administration of galanthamine hydrobromide
loaded flexible liposomes. Environ Toxicol Pharmacol 2012; 34(2):
272-9.
[http://dx.doi.org/10.1016/j.etap.2012.04.012] [PMID: 22613079]
[176] Yang ZZ, Zhang YQ, Wang ZZ, Wu K, Lou JN, Qi XR. Enhanced
brain distribution and pharmacodynamics of rivastigmine by
liposomes following intranasal administration. Int J Pharm 2013;
452(1-2): 344-54.
[http://dx.doi.org/10.1016/j.ijpharm.2013.05.009] [PMID:
23680731]
[177] Mutlu NB, Değim Z, Yilmaz Ş, Eşsiz D, Nacar A. New perspective
for the treatment of Alzheimer diseases: liposomal rivastigmine
formulations. Drug Dev Ind Pharm 2011; 37(7): 775-89.
[http://dx.doi.org/10.3109/03639045.2010.541262] [PMID:
21231901]
[178] Arumugam K, Subramanian GS, Mallayasamy SR, Averineni RK,
Reddy MS, Udupa N. A study of rivastigmine liposomes for deliv-
ery into the brain through intranasal route. Acta Pharm 2008; 58(3):
287-97.
[http://dx.doi.org/10.2478/v10007-008-0014-3] [PMID: 19103565]
[179] Al Asmari AK, Ullah Z, Tariq M, Fatani A. Preparation, charac-
terization, and in vivo evaluation of intranasally administered
liposomal formulation of donepezil. Drug Des Devel Ther 2016;
10: 205-15.
[PMID: 26834457]
[180] Spuch C, Navarro C. Liposomes for targeted delivery of active
agents against neurodegenerative diseases (Alzheimer’s disease and
Parkinson’s disease). J Drug Deliv 2011; 2011469679
[http://dx.doi.org/10.1155/2011/469679] [PMID: 22203906]
[181] Mourtas S, Canovi M, Zona C, et al. Curcumin-decorated
nanoliposomes with very high affinity for amyloid-β1-42 peptide.
Biomaterials 2011; 32(6): 1635-45.
[http://dx.doi.org/10.1016/j.biomaterials.2010.10.027] [PMID:
21131044]
[182] Patel PA, Patil SC, Kalaria DR, Kalia YN, Patravale VB. Compara-
tive in vitro and in vivo evaluation of lipid based nanocarriers of
Huperzine A. Int J Pharm 2013; 446(1-2): 16-23.
[http://dx.doi.org/10.1016/j.ijpharm.2013.02.014] [PMID:
23410989]
[183] Jin Y, Wen J, Garg S, et al. Development of a novel niosomal
system for oral delivery of Ginkgo biloba extract. Int J Nanomedi-
cine 2013; 8: 421-30.
[http://dx.doi.org/10.2147/IJN.S37984] [PMID: 23378764]
[184] Chaiyana W, Rades T, Okonogi S. Characterization and in vitro
permeation study of microemulsions and liquid crystalline systems
containing the anticholinesterase alkaloidal extract from Taber-
naemontana divaricata. Int J Pharm 2013; 452(1-2): 201-10.
[http://dx.doi.org/10.1016/j.ijpharm.2013.05.005] [PMID:
23680734]
[185] Zhang LK, Xu RX, Jiang M, et al. Evaluation of brain-targeting of
β-asarone microemulsion by intranasal administration. Chin Tradit
Herbal Drugs 2014; 45(1): 86-9.
DISCLAIMER: The above article has been published in Epub (ahead of print) on the basis of the materials provided by the author. The Edito-
rial Department reserves the right to make minor modifications for further improvement of the manuscript.
... In this section, we will look at some of the preclinical research investigations that used nanocarriers and other gene-targeting strategies to successfully boost drug bioavailability in the brain. However, neuronal transmission impairment can enhance patients' cognitive abilities without being able to stop or reverse AD progression [98]. The functionalized NPs for symptomatic treatments and hallmark targeting are administered to diseased neurons in the AD brain. ...
... Represent the primary target of nanocarrier-based drug delivery and the role of the relevant nanocarrier in treating significant impairment induced by a specific target gene in the AD [97,98]. The interest in nanoparticles, namely polymeric NP, liposome NP, and MSNs for drug administration, is examined in this review study due to a number of issues. ...
Article
Full-text available
Alzheimer’s disease (AD), is the major type of dementia and most progressive, irreversible widespread neurodegenerative disorder affecting the elderly worldwide. The prime hallmarks of Alzheimer’s disease (AD) are beta-amyloid plaques (Aβ) and neurofibrillary tangles (NFT). In spite of recent advances and developments in targeting the hallmarks of AD, symptomatic medications that promise neuroprotective activity against AD are currently unable to treat degenerating brain clinically or therapeutically and show little efficacy. The extensive progress of AD therapies over time has resulted in the advent of disease-modifying medications with the potential to alleviate AD. However, due to the presence of a defensive connection between the vascular system and the neural tissues known as the blood–brain barrier (BBB), directing these medications to the site of action in the degenerating brain is the key problem. BBB acts as a highly selective semipermeable membrane that prevents any type of foreign substance from entering the microenvironment of neurons. To overcome this limitation, the revolutionary approach of nanoparticle(NP)/nanocarrier-mediated drug delivery system has marked the era with its unique property to cross, avoid, or disrupt the defensive BBB efficiently and release the modified drug at the target site of action. After comprehensive data mining, this review focuses on the detailed understanding of different types of nanoparticle(NP)/nanocarrier-mediated drug delivery system like liposomes, micelles, gold nanoparticles(NP), polymeric NPs, etc. which have promising potential in carrying the desired drug(cargo) to the location in the degenerated brain thus mitigating the Alzheimer’s disease.
... Because of their favorability to the brain, increased stability, biocompatibility, and biodegradability, protection from enzymatic degradation, increased half-life, improved bioavailability, and controlled release, nanomedicines may be more advantageous than other conventional methods of drug delivery to the brain for the treatment of AD [19]. The conjugation or encapsulation of drugs, together with their protection and prolonged blood circulation, are ensured by the fundamental core structures of NP-functionalized nanomedicines [20; 21]. ...
... At the same time, nanocapsules can reduce dosage and frequency while improving patient adherence. 17 Irrespective of some clinical problems, nanomedicines are beneficial in terms of stability, biodegradability, biocompatibility, safety, prevention from degradation enzymes, bioavailability, and sustained release of drugs to treat AD. ...
Article
The incidence of Alzheimer’s disease (AD) is increasing day by day worldwide, which results in a poor quality of life. Early diagnosis and treatment of AD is necessary to suppress the progression of the disease. Conventional treatments have several limitations due to the protective blood-brain barrier. In this review, we described a nanoparticle-based approach to crossing the blood-brain barrier for AD detection and treatment. Nanoparticles encapsulate the anti-AD drug and are directed to the target tissues where controlled release of the drug takes place. There are various types of nanoparticles that are used to encapsulate drugs, including solid-based nanoparticles, liposomes, nanoemulsions, iron NPs, cerium NPs, selenium NPs, and gold NPs. In this review, we have described the use of different nanoparticles as nanomedicine. Nanoparticles are also coated with proteins and antibodies for efficient release of drugs. This review aims to provide clinical insights and the importance of nanotechnology in theranostics and describes how nanomedicine has revolutionized the drug delivery approach for AD treatment
... In this regard, this review aims to discuss the most recently developed DDSs for Food and Drug Administration (FDA)-approved Alzheimer's drugs, emphasizing there in vitro and in vivo performance. Due to the relevance of this topic to the scientific community, several review papers aiming at the use of NPs as DDSs for AD management have been published in recent years [15][16][17][18][19][20]. Despite the extensive information provided, those works only addressed the use of NPs. ...
Article
Full-text available
Alzheimer’s disease (AD) is the most common form of dementia, with a high impact worldwide, accounting for more than 46 million cases. The continuous increase of AD demands the fast development of preventive and curative therapeutic strategies that are truly effective. The drugs approved for AD treatment are classified into acetylcholinesterase inhibitors and N-methyl-D-aspartate receptor antagonists. The therapeutic effectiveness of those drugs is hindered by their restricted access to the brain due to the blood–brain barrier, low bioavailability, and poor pharmacokinetic properties. In addition, the drugs are reported to have undesirable side effects. Several drug delivery systems (DDSs) have been widely exploited to address these issues. DDSs serve as drug carriers, combining the ability to deliver drugs locally and in a targeted manner with the ability to release them in a controlled and sustained manner. As a result, the pharmacological therapeutic effectiveness is raised, while the unwanted side effects induced by the unspecific distribution decrease. This article reviews the recently developed DDSs to increase the efficacy of Food and Drug Administration-approved AD drugs.
Chapter
The book summarizes the role of multiple enzyme targets and strategies to design and develop novel drug candidates for Alzheimer's disease (AD). Insights from researchers across the globe from diverse fields are presented in a thematic volume. The chapters highlight current information scientists have unraveled about the origin, pathogenesis and prevention of AD. The contributions consider both established and emerging drug targets viz. Tau proteins, TREM, and microglia. Topics covered in the book include multi-target anti-Alzheimer's agents, epigenetic modifications, and the role of specific proteins like TMP21 and Tau in AD. A section dedicated to pharmacological treatments discusses the significance of tubulin-modifying enzymes, memantine, and glutamate antagonists. Enzymatic targets for drug discovery are thoroughly examined, focusing on cholinesterase, secretases, and other enzymes. Additionally, the book explores innovative nano-carrier-based drug delivery methods, emphasizing the crucial role of nanotechnology in effective Alzheimer's treatment. The book aims to inform students and researchers in the field of neuroscience, medicine and pharmacology about current research and biochemical nuances of AD pathogenesis and enzymatic drug targeting strategies.
Chapter
Full-text available
Alzheimer's disease (AzD) is the most common type of dementia among older adults, with a progressive disorder of brain that affect thinking ability, behavior, and memorizing capability. It is the most common cause of dementia in older adults, accounting for approximately 60%–80% of all cases of dementia. The exact cause of AzD is yet not fully understood, however several reports indicate that a combinations of genetic, environmental, and lifestyle factors contribute to the development and progression of AzD. The risk factors associated with AzD are age, family history, head injuries, high blood pressure, and high cholesterol. Moreover, the abnormal deposition of protein (β-amyloid plaques and tau tangles) in brain lead to the mortality of brain cells and gradual decline of cognitive functions. Currently, there is no complete cure for AzD available, however advancement in drug delivery system developed several treatment strategies that can help manage symptoms and slow the progression of the disease. The management strategies include medications to improve cognitive function, behavioral and environmental modifications, and support for caregivers and family members. Recently nanotechnology have gained significant attention among researcher in management of AzD although the drug diffusion to blood brain barriers presents several difficulty. Carrier-based drug delivery presents promising alternative to deliver medicament to the brain. Several carrier-based drug delivery reported such as liposomes, nanocrystals, nanoemulsions, dendrimers, polymer-protein/drug conjugates, nanoparticles, antibody conjugated drugs, nanotubes, nanogels, and micelles are included in this chapter for the management AzD. While the use of nanotechnology in AzD is still in early stages of research, it holds great promise for the fabrication of pharmaceuticals in management of this devastating disease.
Article
Full-text available
Alzheimer's disease (AD) is a well-known chronic neurodegenerative disorder that leads to the progressive death of brain cells, resulting in memory loss and the loss of other critical body functions. In March 2019, one of the major pharmaceutical companies and its partners announced that currently, there is no drug to cure AD, and all clinical trials of the new ones have been cancelled , leaving many people without hope. However, despite the clear message and startling reality, the research continued. Finally, in the last two years, the Food and Drug Administration (FDA) approved the first-ever medications to treat Alzheimer's, aducanumab and lecanemab. Despite researchers' support of this decision, there are serious concerns about their effectiveness and safety. The validation of aducanumab by the Centers for Medicare and Medicaid Services is still pending, and lecanemab was authorized without considering data from the phase III trials. Furthermore, numerous reports suggest that patients have died when undergoing extended treatment. While there is evidence that aducanumab and lecanemab may provide some relief to those suffering from AD, their impact remains a topic of ongoing research and debate within the medical community. The fact is that even though there are considerable efforts regarding pharmacological treatment, no definitive cure for AD has been found yet. Nevertheless, it is strongly believed that modern nanotechnology holds promising solutions and effective clinical strategies for the development of diagnostic tools and treatments for AD. This review summarizes the major hallmarks of AD, its etiological mechanisms, and challenges. It explores existing diagnostic and therapeutic methods and the potential of nanotechnology-based approaches for recognizing and monitoring patients at risk of irreversible neuronal degeneration. Overall, it provides a broad overview for those interested in the evolving areas of clinical neuroscience, AD, and related nanotechnology. With further research and development, nanotechnology-based approaches may offer new solutions and hope for millions of people affected by this devastating disease.
Article
Background Alzheimer's disease (AD) is the most common neurological disorder, affecting more than 50 million individuals worldwide and causing gradual but progressive cognitive decline. The rising cost of medical treatment is mostly attributable to AD. There are now mainly a few slightly symptomatic therapeutic options accessible. Although this is not the primary reason, the failure to develop effective treatments for AD is often attributed to the disease's complicated pathophysiology and the wide range of underlying ideas. Objective Studies undertaken over the past decade have aimed to find novel methods of overcoming these barriers and effectively delivering drugs to the central nervous system. As a result, nanotechnology provides a promising alternative to the standard means of administering anti-amyloidosis drugs, enhancing expectations for a successful treatment of Alzheimer's disease. These therapeutic implications of using nanoparticle-based approaches for the treatment of Alzheimer's disease are discussed in this paper. Methodology Published articles from PubMed, SciFinder, Google Scholar, ClinicalTrials.org, and the Alzheimer Association reports were carefully examined to compile information on the various strategies for combating AD. That has been studied to summarize the recent advancements and clinical studies for the treatment of Alzheimer's disease (AD). Statistics is the study and manipulation of data, including ways to gather, review, analyze, and draw conclusions from data. Conclusion The biology of the BBB and its processes of penetration must be carefully taken into account while creating DDSs. If we have a better grasp of the disease's mechanism, we might be able to overcome the shortcomings of current treatments for AD. Different DDSs show interesting properties for delivering medication tailored to the brain. This review paper examines the recent applications of DDSs in diverse domains. By selecting the best targeting vectors and optimizing the combination of carriers, multifunctionalized DDS may be produced, and these DDS have a significant impact on AD therapy potential. To develop DDSs with the best therapeutic efficacy and manageable side effects, experts from a variety of fields may need to contribute their efforts. Currently, the therapeutic use of nanotechnology-based DDSs appears to be a promising prospect for AD therapy, and as the pathophysiology of AD is better understood, this strategy will develop over time.
Article
Neurodegenerative diseases are posing pressing health issues due to the high prevalence among aging populations in the 21st century. They are evidenced by the progressive loss of neuronal function, often associated with neuronal necrosis and many related devastating complications. Nevertheless, effective therapeutical strategies to treat neurodegenerative diseases remain a tremendous challenge due to the multisystemic nature and limited drug delivery to the central nervous system. As a result, there is a pressing need to develop effective alternative therapeutics to manage the progression of neurodegenerative diseases. By utilizing the functional reconstructive materials and technologies with specific targeting ability at the nanoscale level, nanotechnology‐empowered medicines can transform the therapeutic paradigms of neurodegenerative diseases with minimal systemic side effects. This review outlines the current applications and progresses of the nanotechnology‐enabled drug delivery systems to enhance the therapeutic efficacy in treating neurodegenerative diseases. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies
Article
Full-text available
Background: Nicotinamide is considered to be effective in halting the Alzheimer's disease progression. The body could absorb a limited amount of nicotinamide at a time, requiring multiple doses through a day. To overcome such an obstacle which reduces the patient compliance, a sustained/controlled delivery system could be useful. Method: Nicotinamide loaded solid lipid nanoparticles (SLN) were prepared and functionalized with polysorbate 80 (S80), phosphatidylserine (PS) or phosphatidic acid (PA). The acquired particles were characterized and evaluated in respect of their cytotoxicity, biodistribution, and in vivo effectiveness through the different routes of administration. Results: The optimum sizes of 112 ± 1.6 nm, 124 ± 0.8 nm, and 137 ± 1.05 nm were acquired for S80-, PS-, and PA-functionalized SLNs, respectively. The in vitro cytotoxicity on SH-SY5Y cell line showed the safety of formulations except for S80-functionalized SLNs. Biodistribution study of SLNs has proved the benefits of functionalization in improving the brain delivery. The results of spatial and memory test, i.e. Morris water maze, and also histopathology and biochemical tests demonstrated the effectiveness of i.p. injection of PS -functionalized SLNs in improving the cognition, preserving the neuronal cells and reducing tau hyperphosphorylation in a rat model of Alzheimer's disease. Conclusion: The acquired PS-functionalized SLN could be a potential brain delivery system. Loaded with nicotinamide, an HDAC inhibitor, it could ameliorate the cognition impairment of rats more effectively than the conventional administration of nicotinamide, i.e. oral, in the early stage of Alzheimer's disease. Graphical abstract ᅟ.
Article
Full-text available
Objective The first aim of this study was to develop a nanocarrier that could transport the peroxisome proliferator-activated receptor agonist, pioglitazone (PGZ) across brain endothelium and examine the mechanism of nanoparticle transcytosis. The second aim was to determine whether these nanocarriers could successfully treat a mouse model of Alzheimer’s disease (AD). Methods PGZ-loaded nanoparticles (PGZ-NPs) were synthesized by the solvent displacement technique, following a factorial design using poly (lactic-co-glycolic acid) polyethylene glycol (PLGA-PEG). The transport of the carriers was assessed in vitro, using a human brain endothelial cell line, cytotoxicity assays, fluorescence-tagged nanocarriers, fluorescence-activated cell sorting, confocal and transmission electron microscopy. The effectiveness of the treatment was assessed in APP/PS1 mice in a behavioral assay and by measuring the cortical deposition of β-amyloid. Results Incorporation of PGZ into the carriers promoted a 50x greater uptake into brain endothelium compared with the free drug and the carriers showed a delayed release profile of PGZ in vitro. In the doses used, the nanocarriers were not toxic for the endothelial cells, nor did they alter the permeability of the blood–brain barrier model. Electron microscopy indicated that the nanocarriers were transported from the apical to the basal surface of the endothelium by vesicular transcytosis. An efficacy test carried out in APP/PS1 transgenic mice showed a reduction of memory deficit in mice chronically treated with PGZ-NPs. Deposition of β-amyloid in the cerebral cortex, measured by immunohistochemistry and image analysis, was correspondingly reduced. Conclusion PLGA-PEG nanocarriers cross brain endothelium by transcytosis and can be loaded with a pharmaceutical agent to effectively treat a mouse model of AD.
Article
Full-text available
Nicotinamide, the amide form of vitamin B3 (niacin), has long been associated with neuronal development, survival, and function in the central nervous system (CNS), being implicated in both neuronal death and neuroprotection. Here, we summarise a body of research investigating the role of nicotinamide in neuronal health within the CNS, with a focus on studies that have shown a neuroprotective effect. Nicotinamide appears to play a role in protecting neurons from traumatic injury, ischaemia, and stroke, as well as being implicated in 3 key neurodegenerative conditions: Alzheimer’s, Parkinson’s, and Huntington’s diseases. A key factor is the bioavailability of nicotinamide, with low concentrations leading to neurological deficits and dementia and high levels potentially causing neurotoxicity. Finally, nicotinamide’s potential mechanisms of action are discussed, including the general maintenance of cellular energy levels and the more specific inhibition of molecules such as the nicotinamide adenine dinucleotide-dependent deacetylase, sirtuin 1 (SIRT1).
Article
Full-text available
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder and the most common cause for dementia. There are many hypotheses about AD, including abnormal deposit of amyloid β (Aβ) protein in the extracellular spaces of neurons, formation of twisted fibers of tau proteins inside neurons, cholinergic neuron damage, inflammation, oxidative stress, etc., and many anti-AD drugs based on these hypotheses have been developed. In this review, we will discuss the existing and emerging hypothesis and related therapies.
Article
Full-text available
The so-called amyloid hypothesis, that the accumulation and deposition of oligomeric or fibrillar amyloid β (Aβ) peptide is the primary cause of Alzheimer's disease (AD), has been the mainstream concept underlying AD research for over 20 years. However, all attempts to develop Aβ-targeting drugs to treat AD have ended in failure. Here, we review recent findings indicating that the main factor underlying the development and progression of AD is tau, not Aβ, and we describe the deficiencies of the amyloid hypothesis that have supported the emergence of this idea.
Article
Full-text available
In recent years, the incidental rate of neurodegenerative disorders has increased proportionately with the aging population. Alzheimer’s disease (AD) is one of the most commonly reported neurodegenerative disorders, and it is estimated to increase by roughly 30% among the aged population. In spite of screening numerous drug candidates against various molecular targets of AD, only a few candidates – such as acetylcholinesterase inhibitors are currently utilized as an effective clinical therapy. However, targeted drug delivery of these drugs to the central nervous system (CNS) exhibits several limitations including meager solubility, low bioavailability, and reduced efficiency due to the impediments of the blood-brain barrier (BBB). Current advances in nanotechnology present opportunities to overcome such limitations in delivering active drug candidates. Nanodrug delivery systems are promising in targeting several therapeutic moieties by easing the penetration of drug molecules across the CNS and improving their bioavailability. Recently, a wide range of nano-carriers, such as polymers, emulsions, lipo-carriers, solid lipid carriers, carbon nanotubes, metal based carriers etc., have been adapted to develop successful therapeutics with sustained release and improved efficacy. Here, we discuss few recently updated nano-drug delivery applications that have been adapted in the field of AD therapeutics, and future prospects on potential molecular targets for nano-drug delivery systems.
Article
The current clinical symptomatic therapy for Alzheimer's disease involves increasing acetylcholine levels in the brain by inhibiting acetylcholinesterase. However, the effectiveness of acetylcholinesterase inhibitors decreases as the disease progresses, leading to many side effects including over-inhibition of other enzymes and hepatic injury. Herein, we investigate the effects of the direct delivery of a low-dose of acetylcholine via human serum albumin nanoparticles to brain. This novel nanodrug improved both spatial learning and memory capability, whereas it reduced oxidative damage in mice. More importantly, damage to the liver or interference with the inherent neurotransmitter generation due to supplementation were almost absent. Our study is the first to demonstrate that supplementation of acetylcholine-loaded nanoparticles might offer a better therapeutic option in the ease of Alzheimer's disease.
Article
Background: Presence of blood brain barrier is one of the major hurdle in drug delivery to brain for the treatment of neurological diseases. Alternative and more effective drug delivery approaches have been investigated for the drug targeting to brain in therapeutic range. Objective: The present investigation was carried out to improve the galantamine bioavailability in brain by intranasal drug delivery through thiolated chitosan nanoparticles and compared to nasal and oral delivery of its solution using pharmacodynamic activity as well as biochemical estimation. Method: Thiolated chitosan (modified) nanoparticles were fabricated using modified ionic gelation method and intranasal delivery is evaluated by reversal of scopolamine induced amnesia and biochemical estimation of acetylcholinesterase activity in Swiss albino mice brain. Scopolamine (0.4 mg/kg, i.p.) was used to induce amnesia. Piracetam (400mg/kg, i.p.) was used as positive control. Mice were treated with galantamine solution (4mg/kg) by oral and nasal route and formulated galantamine nanoparticles (equivalent to 4mg/kg) by intranasal administrationfor 7 successive days and the results were compared statistically. Results: Intranasal delivery of galantamine loaded thiolated chitosan nanoparticles was found significant (p<0.05) as compared oral and nasal administration of its solution, by pharmacodynamic study and biochemical estimation of acetylcholinesterase activity in Swiss albino mice brain. Conclusion: Significant recovery in amnesia induced mice model by intranasal administration of galantamine loaded thiolated chitosan nanoparticles establish the relevance of nose to brain delivery over the conventional oral therapies for the treatment of alzheimer's disease.
Chapter
Nanoemulsions are being increasingly utilized as drug delivery systems for the effective administration of pharmaceuticals because of their potential advantages over other approaches. Nanoemulsions can be used to design delivery systems that have increased drug loading, enhanced drug solubility, increased bioavailability, controlled drug release, and enhanced protection against chemical or enzymatic degradation. Moreover, nanoemulsions have better stability to flocculation, sedimentation, and creaming than conventional emulsions. Their small droplet dimensions and large droplet surface area positively influence drug transport and delivery, along with allowing targeting to specific sites. This chapter focuses on recent applications of nanoemulsions in the area of drug delivery.
Article
Alzheimer’s disease (AD) remains to be an incurable disease and lacks efficient diagnostic methods. Most AD treatments have focused on amyloid-β (Aβ) targeted therapy, however, it is time to consider the alternative theranostics due to accumulated findings of weak correlation between Aβ deposition and cognition, as well as the failures of Phase III clinical trial on Aβ targeted therapy. Recent researches have shown that tau pathway is closely associated with clinical development of AD symptoms, which might be a potential therapeutic target. We herein construct a methylene blue (MB, a tau aggregation inhibitor) loaded nanocomposite (CeNC/IONC/MSN-T807), which not only possesses high binding affinity to hyperphosphorylated tau, but also inhibits multiple key pathways of tau-associated AD pathogenesis. We demonstrate that these nanocomposites can relieve the AD symptoms by mitigating mitochondrial oxidative stress, suppressing tau hyperphosphorylation and protecting neuronal death both in vitro and in vivo. The memory deficits of AD rats are significantly rescued upon treatment with MB loaded CeNC/IONC/MSN-T807. Our results indicate that hyperphosphorylated tau-targeted multifunctional nanocomposites would be a promising therapeutic candidate for Alzheimer’s disease.