Alzheimer Disease PW
Alzheimer Disease PW
BACHELORS OF PHARMACY
By
ABHISHEK
This is to certify that Mr. Abhishek S/O Mr. Surender Kumar a student of B.Pharmacy Final
Year in this college has worked on the project entitled
He has worked hard, meticulously and methodically. I wish him every success in future
endeavors and recommended that project work to be forwarded for evaluation
SUPERVISOR PRINCIPAL
PHARMACY PHARMACY
2
Affectionately
Dedicated
To
GOD,
TEACHERS
and
my
FAMILY
3
Saraswati Mantra...
4
Acknowledgment
work on this project has been an inspiring often exciting, sometimes challenging, The but
always interesting and an enjoyable experience.
Thankfully, I've had the opportunity to work with a great many people who are far more
talented, dedicated and experienced than me at every step of the way. I know I can be quite a
handful at times, but many of my mentors have been generous enough with their time and trust
to permit me to pursue my own paths.
First of all, I would like to thank my supervisor Ms Anjali Chhikara, South Point College of
Pharmacy, who guided this project and helped whenever I was in need.
He has always had time for discussion and continually shown great interest in my research.
Among his absolutely best sides is her ability to stimulate one's imagination and creativity.
I owe a huge debt of gratitude to Mr. Dilbag Singh Khatri, Chairman, SouthPoint College of
Pharmacy for providing me all the facilities and encouragement for the successful completion
of my project work.
I would also like to thank Dr. Mamta Sachdeva, Director, South Point College of Pharmacy for
her outstanding advice and valuable contribution for my project work.
A Special thanks to the faculty of South Point College of Pharmacy, for their help and guidance
during the research.
I am very thankful to library In charge, for their help and guidance during the research.
I am thankful to my friends Sunny, Pawan, Himanshu for their constant support and
encouragement.
Words are inadequate to reflect the extent of my feeling for my parents whose bounteous
support, love, motivation, encouragement and sacrifice at various stages of my life has
provided the stepping stones for me to reach the milestone where I am today. My worthy
parents believed in me before I believed in myself.
ABHISHEK
5
INDEX
1. INTRODUCTION 8-10
6
LIST OF FIGURE
7. Cholinesterase Inhibitors 28
7
ABSTRACT
Owing to the important progress in the field of pathophysiology in the last couple of years,
new therapeutic targets are available that should render the underlying disease process to be
tackled directly. Understanding the extent of Alzheimer disease related knowledge can assist
disease management that result in improved disease management and reduced care costs. This
article attempts to focus on some of the important recent developments in understanding and
management of Alzheimer disease.
8
1. INTRODUCTION
Alzheimer's disease (AD) is the most common cause of dementia and is clinically
characterized by a progression from episodic memory problems to a slow general decline of
cognitive function. [1] In 2013, ~44 million of the world-wide population was estimated to be
affected by dementia and a steep rise to ~136 million has been predicted by 2050 2. To date,
there are no treatments with proven disease-modifying effects and AD remains the largest
unmet medical need in neurology. [1] AD pathology presents a complex interplay between
several biochemical alterations, including changes in amyloid precursor protein metabolism,
phosphorylation of the tau protein, oxidative stress, impaired energetics, mitochondrial
dysfunction, inflammation, membrane lipid dysregulation and neurotransmitter pathway
disruption.[3] Most of these pathological features can be directly linked to metabolic
abnormalities and it is now clear that metabolic dysfunction is an important factor in AD.[4]
For example, impaired cerebral glucose uptake occurs decades prior to the onset of cognitive
dysfunction and is an invariant feature of AD.[5]
Figure-1. The physiological structure of the brain and neurons in (a) healthy brain and (b)
Alzheimer’s disease (AD) brain
9
Aβ42 and mitochondrial enzymes leads to increased release of reactive oxygen species (ROS),
affecting glycolysis, the TCA cycle and mitochondrial respiratory-chain activity through the
accumulation of deleterious intermediate metabolites in the mitochondria.[6-7 ]
1.1Dementia
2.EPIDEMIOLOGY OF ALZHEIMER’DISEASE
AD is a critical public health issue in the United States and many other countries around the
world, with a significant health, social, and financial burden on society. An estimated 5 million
Americans have AD, with a new diagnosis being made every 68 sec.8 In the United States, AD
is the fifth leading cause of death among older adults, and about $200 billion are spent annually
on direct care of individuals living with dementia. Worldwide, it is estimated that 35 million
people have AD or other types of dementia, and about 65 million people are expected to have
dementia by 2030 (115 million by 2050).
AD is a multi-factorial disease, with no single cause known, and several modifiable and
nonmodifiable risk factors are associated with its development and progression. Age is the
greatest risk factor for the development of AD. The likelihood of developing AD increases
exponentially with age, approximately doubling every 5 years after age 65. [10-11] The vast
majority ofindividuals suffering from AD are aged 65 or older andhave ‘late-onset’ or
10
‘sporadic’ AD (95% of all cases).Rare genetic mutations are associated with the developmentof
AD before age 65, which is known as ‘earlyonset’or ‘familial’ AD (B5% of all cases). [12]
People with familial forms of AD have an autosomal dominant mutation in either one of the
presenilin genes located on chromosomes 1 and 14 or in the amyloid precursor protein (APP)
gene located on chromosome 21
. In addition, individuals with Down’s syndrome (trisomy 21) have an increased risk of
developing early-onset AD. The genetics of sporadic AD are more complex and less well
understood. It is known that the epsilon four allele of the apolipoprotein E (APOE) gene located
on chromosome 19 is a risk factor for the development of sporadic AD. [13] The prevalence of
AD is higher among females, reflecting the longer life expectancy of women. [14] Lower
educational attainment has been associated with increased risk of AD dementia,10 consistent
with the idea that education serves to increase a person’s cognitive reserve and resilience to
AD pathology. [15] A large body of evidence suggests that cerebrovascular risk factors play a
significant role in both the development progression of AD; people with a history of diabetes,
hypertension, obesity, and smoking have a substantially elevated risk of AD. [16] Family
history of AD in first-degree relatives and a history of head injury with loss of consciousness
are also risk factors for the development of A.[17]
2.1 History - Alois Alzheimer and Auguste D The German psychiatrist and neuropathologist
Dr Alois Alzheimer is credited with describing for the first time a dementing condition which
later became known as AD. In his landmark 1906 conference lecture and a subsequent 1907
article, Alzheimer described the case of Auguste D, a 51-year-old woman with a ‘peculiar
disease of the cerebral cortex,’ who had presented with progressive memory and language
impairment, disorientation, behavioural symptoms (hallucinations, delusions, paranoia), and
psychosocial impairment. [18-20]
Alzheimer later published his descriptions of several similar patients in 1909 and Kraepelin
included Ms. Deter's case in the 1910 edition of his widely respected psychiatry textbook. It
was Kraepelin who named this dementia after his junior colleague.
Auguste Deter was not an elderly woman at the onset of her illness, and Alzheimer's disease
(AD) was therefore regarded as a "presenile dementia" to distinguish it from the familiar "senile
dementia" thought to result from aging-related vascular disease. Further investigation,
11
however, showed that plaques and tangles were present in the brains of the majority of older
adults with symptoms of dementia.
In the late 1960's, the British psychiatrists Tomlinson and Roth described the importance of
these plaques in older adults, and in 1970 Dr.. Roth questioned the meaningfulness of the age
criterion that distinguished AD from "senile dementia of the Alzheimer's type."
12
changes, including
depression or anxiety may
react unusually angrily or
aggressively on occasion.
Behaviour changes may Change in mobility, may be
include wandering, repeated unable to walk or be confined
questioning, calling out, to a wheelchair or bed
clinging, disturbed sleeping,
hallucinations (seeing or
hearing things which are not
there)
May display inappropriate Behaviour changes, may
behaviour in the home or in escalate and include
the community (e.g. aggression towards carer,
disinhibition, aggression). nonverbal agitation (kicking,
hitting, screaming or
moaning)
Source: World Alzheimer’s Report 2009. [22]
Researchers believe that many risk factors play a role in causing Alzheimer's and other
dementias, including genetics, behaviors and habits. While some risk factors, such as age and
family history, may be set in their influence, there are many risk factors that can be changed to
potentially reduce a person's risk of cognitive decline[23]
5. Genetics (heredity)
4.1.Age
The greatest known risk factor for Alzheimer’s and other dementias is increasing age, but these
disorders are not a normal part of aging. While age increases risk, it is not a direct cause of
13
Alzheimer's.
Most individuals with the disease are 65 and older. After age 65, the risk of Alzheimer's doubles
every five years. After age 85, the risk reaches nearly one-third. [24-25]
Another strong risk factor for Alzheimer's is family history. According to research, those who
have a parent, brother or sister with Alzheimer’s are more likely to develop the disease. The
risk increases if more than one family member has the illness. Modifiable risk factors such as
sleep, smoking habits, hypertension or diabetes can further increase the risk.
Scientists know genes are involved in Alzheimer’s. Two categories of genes influence whether
a person develops a disease: risk genes and deterministic genes. Alzheimer's genes have been
found in both categories. It is estimated that less than 1% of Alzheimer’s cases are caused by
deterministic genes (genes that cause a disease, rather than increase the risk of developing a
disease).
4.4.Head Injury
There is a link between head injury and future risk of dementia. Protect your brain by buckling
your seat belt, wearing your helmet when participating in sports, and “fall-proofing” your home
Some of the strongest evidence links brain health to heart health. This connection makes sense,
because the brain is nourished by one of the body’s richest networks of blood vessels, and the
heart is responsible for pumping blood through these blood vessels to the brain.
The risk of developing dementia appears to be increased by many conditions that also are not
good for our hearts, including high blood pressure and diabetes. Work with your doctor to
monitor your heart health and treat any problems that arise [26].
5.1.1. Aging
14
The most important risk factor in AD is aging. Younger individuals rarely have this disease,
and most AD cases have a late onset that starts after 65 years of age [27]. Aging is a complex
and irreversible process that occurs through multiple organs and cell systems with a reduction
in the brain volume and weight, a loss of synapses, and ventricles’ enlargement in specific areas
accompanied by SP deposition and NFT. Moreover, several conditions might emerge during
aging such as glucose hypometabolism, cholesterol dyshomeostasis, mitochondria dysfunction,
depression, and cognitive decline. These changes also appear in normal aging, which makes it
difficult to distinguish the cases in early AD [28-29]. AD can be divided based on age of onset
into early-onset AD (EOAD), the rare form with around 1–6% of cases, in which most of them
are familial AD characterized by having more than one member in more than one generation
with AD, and ranges from 30–60 or 65 years. The second type is the late-onset AD (LOAD),
which is more common with age of onset above 65 years. Both types may occur in people who
have a family with a positive history of AD and families with a late-onset disease [30].
5.1.2. Genetics
Genetic factors were discovered over the years and were found to play a major role in the
development of AD. 70% of the AD cases were related to genetic factors: most cases of EOAD
are inherited in an autosomal dominant pattern and mutations in the dominant genes such as
Amyloid precursor protein (APP), Presenilin-1 (PSEN-1), Presenilin-2 (PSEN-2), and
apolipoprotein E (ApoE) are associated with AD [31-32]. Herein, we discuss the strong genetic
risk factors in AD.
APP is a type I transmembrane protein cleaved by α-, β-, and γ-secretase to release Aβ and
other proteins and is encoded by the APP gene on chromosome 21. Thirty mutations have been
found in the APP gene in which twenty-five of them are related to AD and cause an
accumulation of Aβ with elevated amounts. Meanwhile, there is one protective mutation,
A673T, which protects Molecules 2020, 25, 5789 7 of 28 against AD by decreasing Aβ, Aβ40,
and Aβ42 secretion [33-34]. All mutations surround the secretase cleavage site, for example,
the KM670/671NL mutation in mouse models has shown an increasing level of amyloid
plaques in the hippocampus and cortex with no NFTs. A673V, D678H, D678N, E682K, and
K687N mutations have shown cortical atrophy, whereas E682K has shown hippocampal
atrophy. Neuropathological reports for the A673V mutation demonstrated a presence of NFTs
and Aβ, activation of microglia and astrocytes, and neuronal loss, compared to the rest of the
15
mentioned mutations, which show no change in the intracellular Aβ according to
neuropathological reports [33,35]. Other mutations such as T714I, V715A, V715M, V717I,
V717L, L723P, K724N, and I716V affect the γ-secretase cleavage site and cause an increase
in the Aβ42/Aβ40 ratio, while E693G, E693K, D694N, and A692G mutations affect the α-
secretase cleavage site and cause polymorphic aggregates with the ability to disrupt bilayer
integrity. Also, the E693delta is a deletion mutation that enhances the formation of synaptotoxic
Aβ [36-37].
• Apolipoprotein E (ApoE)
ApoE protein is a glycoprotein expressed highly in the liver and brain astrocytes and some
microglia and serves as a receptor-mediated endocytosis ligand for lipoprotein particles like
cholesterol, which is essential for myelin production and normal brain function. The ApoE
gene located on chromosome 19 has three isoforms, ApoE2, ApoE3, and ApoE4, due to single-
nucleotide polymorphisms (SNPs) which cause changes in the coding sequence. The ApoEε4
allele is a strong risk factor for both EOAD and LOAD compared to ApoEε2 and ApoEε3
alleles that
are associated with a lower risk and protective effect, respectively [38]. ApoEε4 plays an
important role in Aβ deposition as a senile plaque and causes cerebral amyloid angiopathy
(CAA), which is known as a marker for AD [39]. ApoEε4 was also shown to be associated with
vascular damage in the brain, which leads to AD pathogenesis [40].
• Other Genes
Other genes’ polymorphism associated with increasing the risk of AD include vitamin D
receptor (VDR) gene polymorphism, which affects the affinity of vitamin D to its receptor and
may cause neurodegenerative diseases and neuronal damage [41]. Moreover, epigenetic factors
like DNA methylation, histone, and chromatin modifications were demonstrated to be involved
in AD [42].
Aging and genetic risk factors cannot explain all cases of AD. Environmental risk factors
including air pollution, diet, metals, infections, and many others may induce oxidative stress
and inflammation and increase the risk for developing AD. Herein, we report the most
important environmental factors and their relationships with AD [43-44]
16
5.2.2. Air Pollution
The air pollution is characterized by modifying the nature of the atmosphere through the
introduction of chemical, physical, or biological pollutants. It is associated with respiratory and
cardiovascular diseases and recently, its association with AD was documented. Six air
pollutants have been defined by National Ambient Air Quality Standards (NAAQSs) in the
USA as a threat to human health, including ozone (O3), nitrogen oxides (NOx), carbon
monoxide (CO), particulate matter (PM), sulfur dioxide (SO2), and lead. Studies on animals
and cellular models have shown that an exposure to high levels of air pollution can result in a
damage to the olfactory mucosa and bulb, in addition to the frontal cortex region, similar to
that observed in AD. In individuals exposed to air pollutants, there is a link between oxidative
stress, neuroinflammation, and neurodegeneration, with the presence of hyper-phosphorylated
tau and Aβ plaques in the frontal cortex. The air pollution can cause an increase in Aβ42
formation, accumulation, and impaired cognitive function [45-46].
5.2.3. Diet
In recent years, the number of studies on the role of nutrition in AD have been increased.
Several dietary supplements such as antioxidants, vitamins, polyphenols, and fish were
reported to decrease the risk of AD, whereas
saturated fatty acids and high-calorie intake were associated with increasing the risk of AD
[47]. The food processing causes degradation of heat-sensitive micronutrients (e.g., vitamin C
and folates), loss of large amounts of water, and formation of toxic secondary products
(advanced glycation end products, AGEs) from non-enzymatic glycation of free amino groups
in proteins, lipids, and nucleic acids. The toxic effect of AGEs is referred to as their ability to
induce oxidative stress and inflammation by modifying the structure and function of the cell
surface receptors and body proteins.
5.2.4. Metals
Metals are found in nature and biological systems and can be divided into bio-metals that have
a physiological function in living organisms (e.g., copper, zinc, and iron), and toxicological
metals which do not possess any biological function (e.g., aluminium and lead) [48].
Aluminium is used significantly in the industries such as processed foods, cosmetics, medical
preparations, medicines, and others. In the body, aluminium is bound to plasma transferrin and
to citrate molecules that can mediate the transfer of aluminium to the brain. Studies
17
demonstrated that Al accumulates in the cortex, hippocampus, and cerebellum areas, where it
interacts with proteins and causes misfolding, aggregation, and phosphorylation of highly
phosphorylated proteins like tau protein, characteristic of AD []. Lead competes with the
binding site of bio-metals like calcium and can cross the blood–brain barrier (BBB) rapidly,
where it can modify neural differentiation and synaptogenesis and cause severe damage. [50]
5.2.5. Infections
Chronic infections to the central nervous system (CNS) can cause an accumulation of Aβ
plaques and NFT, therefore, they are included among the risk factors in AD. Studies by Dr.
Itzhaki showed that the DNA of herpes simplex virus (HSV-1) was found in patients with
ApoE-ε4 allele carriers, which explains the high risk for developing AD. HSV-1 can replicate
in the brain, which can result in the activation of the inflammatory response and an increase in
Aβ deposition, resulting in damage to neurons and gradual development of AD[51-53]
18
6. Pathophysiology
Since its discovery and classification, the pathophysiology of Alzheimer’s disease has been
studied extensively, leading to confirmation of the two prominent neuropathological motifs that
Alois Alzheimer (54) and Oskar Fischer (55) first identified in the early 1900s: plaques and
neurofibrillary tangles (56). The first, senile plaques, are defined as extracellular aggregates of
beta-amyloid protein (Aβ), which is produced through proteolytic cleavage of a critical
membrane glycoprotein, amyloid precursor protein (APP) (57). While APP can
be differentially cleaved by β-secretase and γ-secretase to produce Aβ peptides of various
lengths, it is the 42 amino acid form that is predominantly involved in plaque formation (58),
as a result of its decreased solubility and increased propensity for fibril assembly. These fibrils,
while largely implicated in Aβ plaque formation, represent only one of the potential polymeric
forms of Aβ. Because a variety of studies have shown that Aβ plaque formation is not correlated
with the incidence or severity of Alzheimer’s (59), attention has largely turned to the oligomeric
form of Aβ, which is soluble and capable of spreading throughout the brain via the
cerebrospinal fluid (CSF) (60). These oligomers have the potential to bind to a number of
extracellular receptors (61), at least one of which (PrPC) is demonstrated to recognize and bind
Aβ fibrils and oligomers, preventing their elongation and contributing to the formation of short
and highly neurotoxic Aβ polymers (62). Upon binding, their cytotoxic effects seem to
be mediated by disrupted Ca2+ signaling, oxidative stress, and mitochondrial dysfunction
(63). Notably, a 2002 paper published in Nature showed that Aβ oligomers, in the absence of
monomers and fibrils, significantly inhibit long-term potentiation in the hippocampus of rats
(64). Many studies have attempted to determine the mechanism for this neuronal damage; for
example, a 2020 study found that incubation with soluble Aβ oligomers led to sensitization of
Toll-like Receptor 4 (TLR4) and increased production of Tumor Necrosis Factor-ɑ (TNF-ɑ) in
murine microglia and astrocytes (65). However, research into the role of Aβ in Alzheimer’s is
far from complete—in the light oF
news that data from a critical 2006 study demonstrating the role of 56kDa Aβ oligomers in
murine impairment (66) was likely falsified, new findings are of critical importance in
affirming the validity of existing research
19
FIGURE.3- Graphical representation of the three primary pathophysiological manifestations
of Alzheimer’s disease. (A) The amyloid plaque in the cerebral cortex is formed as a result of
APP cleavage to form the Amyloid-beta monomer, which can subsequently form soluble
oligomers. These oligomers can exert neurotoxic effects as they are, but may also assemble
into insoluble protofibrils and later fibrils, which compose the plaques. (B) Depicted within a
pyramidal neuron of the hippocampus, the neurofibrillary tangle (NFT) is localized to the cell
body and results from the hyperphosphorylation of tau protein, which aggregates into paired
helical filaments (PHFs). (C) Death of basal forebrain cholinergic neurons is a prominent
marker of Alzheimer’s disease.
20
Serine/Threonine residues alters its chemical and physical properties such that it can no longer
fulfill its biological function (69). Specifically, it has been found that hyperphosphorylated tau
protein is unable to bind tubulin (which is critical for its role in microtubule assembly), but
readily binds to normal tau protein and other microtubule-associated proteins (70), contributing
to the loss of cytoskeletal microtubules (71) and increased intracellular tau aggregation
observed in the Alzheimer’s brain. Further, tau hyperphosphorylation contributes to
intracellular tau mislocalization, including to the dendritic spine where it contributes to
synaptic dysfunction (72). Further, the degeneration of cholinergic neurons in the nucleus
basalis has been widely documented in the brains of patients with Alzheimer’s (73), eliciting
an alternative, cholinergic hypothesis forthe cognitive deficits observed in AD
21
Amyloid ẞ precursor protein (APP)
ẞand y secretases
Amyloid ẞ peptide
Accumulation on neurons
Neurotransmitter deficit
22
7. Diagnosis of Alzheimer Disease-
There is no single test that can determine if a person is living with Alzheimer’s or another
dementia. Physicians use diagnostic tools combined with medical history and other
information, including neurological exams, cognitive and functional assessments, brain
imaging (MRI, CT, PET) and cerebrospinal fluid or blood tests to make an accurate
diagnosis.[74]
1. Medical history
During the medical workup, the health care provider will review the person's medical
history, including psychiatric history and history of cognitive and behaviourl changes. He
or she will want to know about any current and past medical problems and concerns, as
well as any medications the person is taking. The doctor will also ask about key medical
conditions affecting other family members, including whether they may have had
Alzheimer's disease or other dementias.
• Review all medications. (Bring a list or the containers of all medicines currently
being taken, including over-the-counter drugs and supplements.)
23
• Collect blood or urine samples for laboratory testing.
Information from a physical exam and laboratory tests can help identify health issues
that can cause symptoms of dementia. [75]
3. Neurological Exam
During a neurological exam, the physician will closely evaluate the person for problems
that may signal brain disorders other than Alzheimer's. The doctor will look for signs of
stroke, Parkinson's disease, brain tumors, buildup of fluid in the brain, and other
conditions that may impair memory or thinking
• Reflexes.
• Eye movement.
• Speech.
• Sensation.
4. cognitive ,functional and behavioural test
Cognitive, functional and behavioral tests evaluate memory, thinking and simple
problem-solving abilities, and may quickly assess changes in behaviors and symptoms.
Some tests are brief, while others can be more time intensive and complex. More
comprehensive cognitive, functional and behavioral tests are often given by a
neuropsychologist to evaluate executive function, judgment, attention and language.
Such tests may give an overall sense of whether a person is experiencing cognitive
symptoms that affect activities of daily living and function and is aware of these
symptoms; knows the date, time and where he or she is; and can remember a short list of
words, follow instructions and perform simple calculations.[76]
5.Depression Screen
24
In addition to assessing mental status, the doctor will evaluate a person's sense of well-
being to detect depression or other mood disorders that can cause memory problems, loss
of interest in life, and other symptoms that can overlap with dementia.
6. Brain Imaging
6.1 Fluorodeoxyglucose (FDG) PET imaging scans show areas of the brain in which
nutrients are poorly metabolized. Finding patterns in the areas of low metabolism can help
distinguish between Alzheimer's disease and other types of dementia.
25
6.2 Amyloid PET imaging can measure the burden of amyloid deposits in the brain. This
test is mainly used in research but may be used if a person has unusual or very early onset of
dementia symptoms.
6.3 Tau PET imaging, which measures the tangles in the brain, is generally used in the
research setting.
7. CSF test
CSF is a clear fluid that bathes and cushions the brain and spinal cord. Adults have about
1 pint of CSF, which physicians can sample through a minimally invasive procedure
called a lumbar puncture, or spinal tap. Research suggests that Alzheimer's disease in
early stages may cause changes in CSF levels of multiple markers such as tau and beta-
amyloid, two markers that form abnormal brain deposits strongly linked to Alzheimer's.
Another potential marker is neurofilament light (NfL), an increased level of which has
been found in neurodegenerative diseases such as Alzheimer’s.
There are a few new CSF tests — Lumipulse® and Elecsys® — that have received FDA
approval. These diagnostic tools can be used by clinicians to detect beta-amyloid and tau
markers in CSF, which can be predictive of amyloid changes in the brain.[77]
8. Blood test
Researchers are investigating whether consistent and measurable changes in blood levels
of specific markers may be reliably associated with Alzheimer’s related changes. These
markers may include tau, beta-amyloid or other biomarkers the could be measured
before and after symptoms appear.
An urgent need exists for simple, inexpensive, non-invasive and easily available diagnostic
tools such as blood tests to diagnose the disease. These testing technologies would support
drug development by helping to identify and follow treatment effectiveness in clinical trial
participants and to increase the possibility of early detection, diagnosis and intervention.
A blood test would also enable interpretation and understanding of the progression of
Alzheimer’s in larger and more diverse populations.[78]
Currently, Alzheimer’s disease cases worldwide are reported to be around 24 million, and in
2050, the total number of people with dementia is estimated to increase 4 times. Even though
26
AD is a public health issue, as of now, there is only two classes of drugs approved to treat AD,
including inhibitors to cholinesterase enzyme (naturally derived, synthetic and hybrid
analogues) and antagonists to N-methyl d-aspartate (NMDA). Several physiological processes
in AD destroy Ach-producing cells which reduce cholinergic transmission through the brain.
Acetylcholinesterase inhibitors (AChEIs), which are classified as reversible, irreversible, and
pseudo-reversible, act by blocking cholinesterase enzymes (AChE and butyrylcholinesterase
(BChE)) from breaking down ACh, which results in increasing ACh levels in the synaptic cleft
. On the other hand, overactivation of NMDAR leads to increasing levels of influxed Ca2+,
which promotes cell death and synaptic dysfunction. NMDAR antagonist prevents
overactivation of NMDAR glutamate receptor and hence, Ca2+ influx, and restores its normal
activity. Despite the therapeutic effect of these two classes, they are effective only in treating
the symptoms of AD, but do not cure or prevent the disease .
27
FIGURE.7-Cholinesterase inhibitors
(A) Donepezil
(B) Rivastigmine
28
functions and daily life activities. Oral administration of the drug is associated with adverse
effects such as nausea, vomiting, dyspepsia, asthenia, anorexia, and weight loss. In many cases,
these they can be settled down in time and consequently, the drug becomes more tolerated.
Rivastigmine can be delivered by transdermal patches for controlled and continuous delivery
of the drug through the skin, with enhanced tolerability and caregiver satisfaction. Also, the
patches can deliver a lower dosage compared to pills, which results in reduced side effects.
Most AD patients suffer from memory loss and swallowing problems which affect their
compliance in administering oral drugs at regular intervals. Therefore, the use of transdermal
patches is the most appropriate method for delivering the drug in AD patients [83-87]
(C )Galantamine (GAL)
Galantamine is considered a standard first-line drug for mild to moderate AD cases. GAL is a
selective tertiary isoquinoline alkaloid with a dual mechanism of action in which it acts as a
competitive inhibitor of AChE and can bind allosterically to the α-subunit of nicotinic
acetylcholine receptors and activate them. GAL can improve behavioral symptoms, daily life
activities, and cognitive performance with good efficacy and tolerability, similar to other AChE
inhibitors. Several delivery systems were developed to improve the drug delivery to the brain:
Wahba et al. attached GAL to ceria-containing hydroxyapatite particles for selective delivery
of the drug to the affected regions in the brain. Misra et al. and Fornaguera et al. used solid-
lipid nanoparticles and nano-emulsification approaches respectively, to carry GAL
hydrobromide. The results of these studies demonstrated a promising strategy for safe delivery
of the drug. Hanafy et al. developed nasal GAL hydrobromide/chitosan complex nanoparticles
which showed good pharmacological efficacy, while Woo et al. utilized the patch system as a
carrier for a controlled release dosage form of the drug [87-91]
29
dysfunction, neuronal cell death, and a decline in cognitive functions. Several NMDAR
uncompetitive antagonists have been developed and entered clinical trials, however, most of
them failed due to low efficacy and side effects. Memantine (5, Figure 4) is the only approved
drug in this category to treat moderate to severe AD; in addition, other NMDAR uncompetitive
antagonist compounds are being developed, such as RL-208 (3,4,8,9-tetramethyltetracyclo
[4.4.0.03,9.04,8]dec-1-yl)methylamine hydrochloride), a polycyclic amine compound that may
possess a promising therapeutic effect in age-related cognitive problems and AD [93-94].
30
31
[96-98]
32
9. Potential Role of Nanoparticles in Treating the Accumulation of Amyloid-
Beta Peptide in Alzheimer’s Patients-
Because the BBB level with nanoparticles is difficult to cross, particles will cross the BBB
within the 50–100 nm range without complex adaptation. If there is a limit to the crossing of
the BBB [102], a therapeutic solution is recommended for each nanoparticle derivative. The
extra iron dramatically accelerates AD in the brain, and the cells are destroyed by excess iron.
On AD plates and in situ-considered encounters because of pathological iron dysfunction,
magnet nanoparticles able to catalyze the formation of reactive oxygen species [103] are
present. To avoid and minimize the growth of Aß, combination therapy is required to
simultaneously manage the imbalance of acetylcholine as a potential cure for AD. Clioquinol
(metal-ion chelating agent) and donepezil (acetylcholinesterase (AChE) inhibitor) co-
encapsulated human serum albumin (HSA) nanoparticles (dcHGT NPs) [104]. A central
limiting factor in brain supply is the BBB. The intention was not to concentrate on
pharmaceuticals using traditional medicinal products. The most promising mechanism in the
delivery of nasal drugs was an improvement in brain medicine [105]. Pharmaceutical products
availability. Lower solubility, low blood–brain barrier and low dryness of anti-AD products
decrease therapeutic efficacy.
In this connection, large and small molecular medicine for the treatment of AD seems to be
promising. Nasal supplies are a popular road. This promising trip results in bad neighborhood
systemic results, greater bioavailability, and therapeutic efficacy [106] on the olfactory route
33
to the brain. The primary Aβ fibrillation engine is the Lys-Leu-Val-Phe-Phe (KLVFF) series.
Aβ is also used for attack, and Aβ aggregation can be avoided [107]. Based on significant
nanoscience and nanotechnology advancement, biosensor advances have made significant
progress in recognizing essential AD biomarkers. The special and special features of
nanomaterials improve the electrochemical and optical activity of the transducer to immobilize
biological components of detection [108]. The aim of this review is to shed light on the use of
nanotechnology in treating Alzheimer’s disease using nanoparticles. To clarify the effective
role of these nanoparticles, they were divided into three sections, and they are polymeric, lipid
and gold nanoparticles. The next sections will deal with more details about them. A comparison
is made between the characteristics of each and the volumes used in each of the treatment
methodologies
Diseases to cure multiple diseases and to resolve treatment hurdles promise to be treated easily
with multifunction. The main enzyme in Aβ formation has been reduced by the supply of non-
34
coding plasmid ribonucleic acid (RNA). Concurrent delivery of therapeutic peptides to the
brain helps to reduce neurofibrillary entanglements [109]. It has been extensively explored on
the routes of nano-sufficiency, such as liposomes, polymeric nanoparticles, micelles,
conjugates, peptide carriers, cyclodextrins, stable dispersions, lipid nanoparticles, and
emulsions. The effect of molybdenum disulfide inhibition is known as molybdenum disulfide
(MoS2). A laser-pulsed removal method was used in polyvinylpyrrolidone functional MoS2
NPs.
Using AuNPs@POMD-pep, the use of BBB to deal with drawbacks of small molecular anti-
AD medication [113] is being used to cross BBBs. Approved expression and successful
clearing in microglia and liver cells of the Aß low-density lipoprotein receptor (LDLR) α-
mangostin, which is in vivo is decreased due to hydrophobia, low solubility and aqueous
environ-mental stability, hence low bioavailability and objective aggregation of bacteria. PEG-
PLA was encapsulated to overcome this limitation. To overcome this limit, poly-metals were
encapsulated [114]
35
curcumin nanoformulation (Se NPs). It has been examined using analytical instrument
techniques to determine the moral structures of the polymer.
Both metabolites given intravenously can be quickly metabolized in the liver and the intestine
into two dangerous metabolites. One easy way to remove toxins is by using combined subunits.
That is not like an antidepressant as the substance cannot pass through the blood–brain barrier.
The particles of surfactant were prepared by a double emulsion evaporation technique.
Glaucoma is a disease in which the patient’s vision slowly reduces over the course of days or
weeks, and it eventually goes completely. The two situations are often linked for many reasons.
The particles also help protect the nerve, and especially the brain, by promoting the delivery of
nanotechnology. Luca Technologies is one of the primary companies that master the field of
drug delivery. It is possible that the medicines that are causing the disorder are made stronger
36
through the help of nanoparticles [115]. The brain supply system Beta-Secretase 1 (BACE1)
with small interfering RNA (siRNA) is optimal and functional. A short peptide extract from
the chimeric rabies virus glycoprotein fragment (RVG-9R) glycoprotein virus enhances
transcellular trajectory in neuronal cells. The perfect molar relationship between siRNA and
BACE1 was thus demonstrated. The installation between them was screened. The nasal
delivery system was proposed for olfactory and trigeminal pathways. The coating process
affects the loading and protection of nanoparticles [116].
The Alzheimer's Association is the world's largest nonprofit funder of Alzheimer's research.
Since 1982, we have awarded over $350 million to more than 2,300 research investigations
worldwide.
When Dr. Alois Alzheimer first described the disease in 1906, a person in the United States
lived, an average of about 50 years. Few people reached the age of greatest risk. As a result,
the disease was considered rare and attracted little scientific interest. That attitude changed
asthe average life span increased and scientists began to realize how often Alzheimer's strikes
37
people in their 70s and 80s. The Centers for Disease Control and Prevention recently estimated
the average life expectancy to be 78.8 years.
Today, Alzheimer's is at the forefront of biomedical research, with 90 percent of what we know
discovered in the last 20 years. Some of the most remarkable progress has shed light on how
Alzheimer's affects the brain. Better understanding of the disease's impact may lead to better
treatments.
Scientists are constantly working to advance our understanding of Alzheimer's. But without
clinical research and the help of human volunteers, we cannot treat, prevent or cure
Alzheimer's. Clinical trials test new interventions or drugs to prevent, detect or treat disease
for safety and effectiveness. Clinical studies are any type of clinical research involving. people
and those that look at other aspects of care, such as improving quality of life. Every clinicaltrial
or study contributes valuable knowledge, regardless if favorable results are achieved.
One promising target is beta-amyloid. This protein fragment builds up into the plaques
considered to be one hallmark of Alzheimer's disease. Researchers have developed several
ways to clear beta-amyloid from the brain or prevent it from clumping together into plaques.
Experimental drugs that zero in on beta-amyloid are now being tested. Many other new
approaches to treatment are also under investigation worldwide. We don't yet know which of
these strategies may work, but scientists say that with the necessary funding, the outlook is
good for developing treatments that slow or stop Alzheimer'.
. Eating a diet low in saturated fats and rich in fruits and vegetables, exercising regularly, and
staying mentally and socially active may all help protect the brain.
11. CONCLUSION
Alzheimer's disease is the most common form of dementia in older adults. The disease is
extremely complex, with causes not fully understood. Patients progress through degenerative,
irreversible phases of the disease, while cognitive functioning (memory, thinking and
reasoning) and behavioural abilities are affected, up to the point where the person must depend
completely on others for basic activities of daily living. Current pharmacological treatment
used in the management of AD may ease the symptoms for a period of time, but donot stop the
38
persistent downward progression of the disease. Patients may benefit from non-
pharmacological strategies tailored to the needs of the individual person. Ongoing research
indicated that specific precautionary measures, i.e., higher education and improved heart
health, are most likely to delay the onset of AD. The urgent need for drug development is
echoed by the words of Margaret Chan, the Director-General, World Health Organization "I
can think of no other disease where innovation, including breakthrough discoveries to developa
cure, is so badly needed."
As medical care becomes more complex and people live longer, the share of the population
over 60 continues to grow, and the incidence of Alzheimer's disease and other dementias
increases, many groups in society feel the effects and pressures. For the healthcare industry
there are pressures both financial- and personnel-related.
No doubt for healthcare providers it can be daunting to take on yet more responsibility but, in
the long term, improving support to caregivers of those with Alzheimer's and other dementias
(and to all family caregivers) stands to save everyone individuals, communities, state and
federal governments-time and money by reducing the need for emergency visits, hospital
readmissions, and related services, and improving the quality of life for care recipients,
caregivers, and all those who support them.
12.REFERNCES
1. M. Citron, Alzheimer's disease: strategies for disease modification, Nat. Rev. Drug Discov.
2010; (9): 387-398
39
5. Z. Chen, C. Zhong, Decoding Alzheimer's disease from perturbed cerebral glucose
metabolism: implications for diagnostic and therapeutic strategies, Prog. Neurobiol. 2013;
(108): 21–43.
6. C. Caspersen, N. Wang, J. Yao, A. Sosunov, X. Chen, J.W. Lustbader, H.W. Xu, D. Stern, G.
McKhann, S.D. Yan, Mitochondrial abeta: a potential focal point for neuronal metabolic
dysfunction in Alzheimer's disease, FASEB J. 2005; (19): 2040–2041.
8. Baddeley, A.D. (1978), 'The trouble with levels: a re-examination of Craik and Lockhart's
framework for memory research', Psychological Review. 1978; 85: 139-152
11. World Alzheimer Report 2009. London, Alzheimer Disease International, 2009.
Neurological disorders: public health challenges. Geneva, World Health Organization, 2006.
12. Ott A, Breteler MM, van Harskamp F, Claus JJ, van der Cammen TJ, Grobbee DE, et al.
Prevalence of Alzheimer’s disease and vascular dementia: association with education. The
Rotterdam study. BMJ 1995; 310: 970-3.
13. Querfurth HW, LaFerla FM. Alzheimer’s disease. N Engl J Med 2010; 362: 329-44.
14. Holtzman DM, Morris JC, Goate AM. Alzheimer’s disease: the challenge of the second
century. SciTransl Med 2011; 3: 77sr1.
15. Reiman EM, Chen K, Alexander GE, Caselli RJ, Bandy D, Osborne D, et al. Correlations
between apolipoprotein E epsilon4 gene dose and brain-imaging measurements of regional
hypometabolism. ProcNatlAcadSci U S A 2005; 102: 8299-302
16. Hebert LE, Scherr PA, McCann JJ, Beckett LA, Evans DA. Is the risk of developing
Alzheimer’s disease greater for women than for men? Am J Epidemiol 2001; 153: 132-6.
17. Stern Y. Cognitive reserve in ageing and Alzheimer’s disease. Lancet Neurol 2012; 11:
1006-12.
20. Maurer K, Volk S, Gerbaldo H. Auguste D and Alzheimer’s disease. Lancet 1997;
349(9064): 1546-9.
21. World Health Organization. WHO Mental Health Gap Action Programme (mhGAP) 2008.
http://www.who.int/mental_health/mhgap/en/
22. International statistical classification of diseases and related health problems, 10th
Revision. Geneva, World Health Organization, 1992.
23. Armstrong, R.A. Risk factors for Alzheimer’s disease. Folia Neuropathol. 2019, 57, 87–
105. [CrossRef] [PubMed]
24. Anand, P.; Singh, B. A review on cholinesterase inhibitors for Alzheimer’s disease. Arch.
Pharmacal Res. 2013, 36, 375–399. [CrossRef]
26. Ferreira-Vieira, T.H.; Guimaraes, I.M.; Silva, F.R.; Ribeiro, F.M. Alzheimer’s disease:
Targeting the Cholinergic System. Curr. Neuropharmacol. 2016, 14, 101–115. [CrossRef]
27. Guerreiro, R.; Bras, J. The age factor in Alzheimer’s disease. Genome Med. 2015, 7, 106.
[CrossRef] [PubMed]
28. Riedel, B.C.; Thompson, P.M.; Brinton, R.D. Age, APOE and sex: Triad of risk of
Alzheimer’s disease. J. Steroid Biochem. Mol. Biol. 2016, 160, 134–147. [CrossRef] [PubMed]
29. Hou, Y.; Dan, X.; Babbar, M.; Wei, Y.; Hasselbalch, S.G.; Croteau, D.L.; Bohr, V.A. Ageing
as a risk factor for neurodegenerative disease. Nat. Rev. Neurol. 2019, 15, 565–581. [CrossRef]
[PubMed]
30. Bekris, L.M.; Yu, C.E.; Bird, T.D.; Tsuang, D.W. Genetics of Alzheimer disease. J. Geriatr.
Psychiatry Neurol. 2010, 23, 213–227. [CrossRef] [PubMed]
31. Van Cauwenberghe, C.; Van Broeckhoven, C.; Sleegers, K. The genetic landscape of
Alzheimer disease: Clinical implications and perspectives. Genet. Med. Off. J. Am. Coll. Med
Genet. 2016, 18, 421–430. [CrossRef]
41
32. Khanahmadi, M.; Farhud, D.D.; Malmir, M. Genetic of Alzheimer’s disease: A narrative
review article. Iran. J. Public Health 2015, 44, 892–901.
33. Li, N.M.; Liu, K.F.; Qiu, Y.J.; Zhang, H.H.; Nakanishi, H.; Qing, H. Mutations of beta-
amyloid precursor protein alter the consequence of Alzheimer’s disease pathogenesis. Neural
Regen. Res. 2019, 14, 658–665. [CrossRef]
34. Tcw, J.; Goate, A.M. Genetics of beta-Amyloid precursor protein in Alzheimer’s disease.
Cold Spring Harb. Perspect. Med. 2017, 7, a024539. [CrossRef]
35. Bi, C.; Bi, S.; Li, B. Processing of mutant beta-amyloid precursor protein and the
clinicopathological features of familial Alzheimer’s disease. Aging Dis. 2019, 10, 383–403.
[CrossRef]
36. Dai, M.H.; Zheng, H.; Zeng, L.D.; Zhang, Y. The genes associated with early-onset
Alzheimer’s disease. Oncotarget 2018, 9, 15132–15143. [CrossRef]
37. Zhao, J.; Liu, X.; Xia, W.; Zhang, Y.; Wang, C. Targeting amyloidogenic processing of APP
in Alzheimer’s disease. Front. Mol. Neurosci. 2020, 13, 137. [CrossRef] [PubMed]
38. Kim, J.; Basak, J.M.; Holtzman, D.M. The role of apolipoprotein E in Alzheimer’s disease.
Neuron 2009, 63, 287–303. [CrossRef]
39. Liu, C.C.; Liu, C.C.; Kanekiyo, T.; Xu, H.; Bu, G. Apolipoprotein E and Alzheimer disease:
Risk, mechanisms and therapy. Nat. Rev. Neurol. 2013, 9, 106–118. [CrossRef]
40. Giau, V.V.; Bagyinszky, E.; An, S.S.; Kim, S.Y. Role of apolipoprotein E in
neurodegenerative diseases. Neuropsychiatr. Dis. Treat. 2015, 11, 1723–1737. [CrossRef]
41. Khorram Khorshid, H.R.; Gozalpour, E.; Saliminejad, K.; Karimloo, M.; Ohadi, M.;
Kamali, K. Vitamin D Receptor (VDR) polymorphisms and late-onset Alzheimer’s disease: An
association study. Iran. J. Public Health 2013, 42, 1253–1258. [PubMed]
42. Liu, X.; Jiao, B.; Shen, L. The epigenetics of Alzheimer’s Disease: Factors and therapeutic
implications. Front. Genet. 2018, 9, 579. [CrossRef] [PubMed]
43. Wainaina, M.N.; Chen, Z.; Zhong, C. Environmental factors in the development and
progression of late-onset Alzheimer’s disease. Neurosci. Bull. 2014, 30, 253–270. [CrossRef]
[PubMed]
42
44. Grant, W.B.; Campbell, A.; Itzhaki, R.F.; Savory, J. The significance of environmental
factors in the etiology of Alzheimer’s disease. J. Alzheimer’s Dis. Jad 2002, 4, 179–189.
[CrossRef] [PubMed] Molecules 2020, 25, 5789 25 of 28
45. Moulton, P.V.; Yang, W. Air pollution, oxidative stress, and Alzheimer’s disease. J.
Environ. Public Health 2012, 2012, 472751. [CrossRef] [PubMed]
46. Croze, M.L.; Zimmer, L. Ozone atmospheric pollution and Alzheimer’s disease: From
epidemiological facts to molecular mechanisms. J. Alzheimer’s Dis. Jad 2018, 62, 503–522.
[CrossRef]
47. Hu, N.; Yu, J.T.; Tan, L.; Wang, Y.L.; Sun, L.; Tan, L. Nutrition and the risk of Alzheimer’s
disease. Biomed. Res. Int. 2013, 2013, 524820. [CrossRef]
48. Adlard, P.A.; Bush, A.I. Metals and Alzheimer’s disease. J. Alzheimer’s Dis. Jad 2006, 10,
145–163. [CrossRef] [PubMed]
49. Colomina, M.T.; Peris-Sampedro, F. Aluminum and Alzheimer’s disease. Adv. Neurobiol.
2017, 18, 183–197. [CrossRef] [PubMed]
50. Huat, T.J.; Camats-Perna, J.; Newcombe, E.A.; Valmas, N.; Kitazawa, M.; Medeiros, R.
Metal toxicity links to Alzheimer’s disease and neuroinflammation. J. Mol. Biol. 2019, 431,
1843–1868. [CrossRef] [PubMed]
51. Sochocka, M.; Zwolinska, K.; Leszek, J. The infectious etiology of Alzheimer’s disease.
Curr. Neuropharmacol. 2017, 15, 996–1009. [CrossRef]
52. Fulop, T.; Itzhaki, R.F.; Balin, B.J.; Miklossy, J.; Barron, A.E. Role of microbes in the
development of Alzheimer’s disease: State of the art—An international symposium presented
at the 2017 IAGG congress in San Francisco. Front. Genet. 2018, 9, 362. [CrossRef]
53. Muzambi, R.; Bhaskaran, K.; Brayne, C.; Smeeth, L.; Warren-Gash, C. Common bacterial
infections and risk of incident cognitive decline or dementia: A systematic review protocol.
BMJ Open 2019, 9, e030874. [CrossRef]
54. Hippius H, Neundörfer G. The discovery of Alzheimer's disease. Dialogues Clin Neurosci.
(2003) 5:101–8. doi: 10.31887/DCNS.2003.5.1/hhippius
55. Goedert M. Oskar Fischer and the study of dementia. Brain. (2009) 132:1102–11. doi:
10.1093/brain/awn256
43
56. Alzheimer A, Stelzmann RA, Schnitzlein HN, Murtagh FR. An English translation of
Alzheimer's 1907 paper, "Uber eine eigenartige Erkankung der Hirnrinde". Clin Anat. (1995)
8:429–31. doi: 10.1002/ca.980080612
58. Roher AE, Lowenson JD, Clarke S, Woods AS, Cotter RJ, Gowing E, et al. betaamyloid-
(1-42) is a major component of cerebrovascular amyloid deposits: implications for the
pathology of Alzheimer disease. Proc Natl Acad Sci U S A. (1993) 90:10836–40. doi:
10.1073/pnas.90.22.10836
59. Ricciarelli R, Fedele E. The amyloid Cascade hypothesis in Alzheimer's disease: It's time
to change our mind. Curr Neuropharmacol. (2017) 15:926–35. doi: 10.2174/157015
9X15666170116143743
60. Haass C, Selkoe DJ. Soluble protein oligomers in neurodegeneration: lessons from the
Alzheimer's amyloid beta-peptide. Nat Rev Mol Cell Biol. (2007) 8:101–12. doi:
10.1038/nrm2101
62. Amin L, Harris DA. Aβ receptors specifically recognize molecular features displayed by
fibril ends and neurotoxic oligomers. Nat Commun. (2021) 12:3451. doi: 10.1038/s41467-
02123507-z 10. Canevari L, Abramov AY, Duchen MR. Toxicity of amyloid beta peptide: tales
of calcium, mitochondria, and oxidative stress. Neurochem Res. (2004) 29:637–50. Doi
63.1023/b:nere.0000014834.06405.af
64. Walsh DM, Klyubin I, Fadeeva JV, Cullen WK, Anwyl R, Wolfe MS, et al. Naturally
secreted oligomers of amyloid beta protein potently inhibit hippocampal long-term potentiation
in vivo. Nature. (2002) 416:535–9. doi: 10.1038/416535a
65. Hughes C, Choi ML, Yi JH, Kim SC, Drews A, George-Hyslop PS, et al. Beta amyloid
aggregates induce sensitised TLR4 signalling causing long-term potentiation deficit and rat
neuronal cell death. Commun Biol. (2020) 3. doi: 10.1038/ s42003-020-0792-9
44
66. Lesné S, Koh MT, Kotilinek L, Kayed R, Glabe CG, Yang A, et al. A specific amyloidbeta
protein assembly in the brain impairs memory. Nature. (2006) 440:352–7. doi:
10.1038/nature04533
67. Crowther RA. Straight and paired helical filaments in Alzheimer disease have a common
structural unit. Proc Natl Acad Sci U S A. (1991) 88:2288–92. doi: 10.1073/ pnas.88.6.2288
68. Grundke-Iqbal I, Iqbal K, Tung YC, Quinlan M, Wisniewski HM, Binder LI. Abnormal
phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal
pathology. Proc Natl Acad Sci U S A. (1986) 83:4913–7. doi: 10.1073/ pnas.83.13.4913
70. Alonso AC, Zaidi T, Grundke-Iqbal I, Iqbal K. Role of abnormally phosphorylated tau in
the breakdown of microtubules in Alzheimer disease. Proc Natl Acad Sci U S A. (1994)
91:5562–6. doi: 10.1073/pnas.91.12.5562
71. Bamburg JR, Bloom GS. Cytoskeletal pathologies of Alzheimer disease. Cell Motil
Cytoskeleton. (2009) 66:635–49. doi: 10.1002/cm.20388
72.Hoover BR, Reed MN, Su J, Penrod RD, Kotilinek LA, Grant MK, et al. Tau mislocalization
to dendritic spines mediates synaptic dysfunction independently of neurodegeneration. Neuron.
(2010) 68:1067–81. doi: 10.1016/j. neuron.2010.11.030
73. Ferreira-Vieira TH, Guimaraes IM, Silva FR, Ribeiro FM. Alzheimer's disease: targeting
the cholinergic system. Curr Neuropharmacol. (2016) 14:101–15. doi: 10.217
4/1570159x13666150716165726
74. Barnes DE, Yaffe K. The projected effect of risk factor reduction on Alzheimer’s disease
prevalence. Lancet Neurol 2011; 10: 819-28.
75. Gilman S. Oxford American handbook of neurology. Oxford University Press: Oxford,
UK; 2010
76. McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM. Clinical diagnosis
of Alzheimer’s disease: report of the NINCDS-ADRDA Work Group under the
45
77. McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR Jr, Kawas CH, et al. The
diagnosis of dementia due to Alzheimer's disease: recommendations from the National Institute
on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's
disease. Alzheimers Dement. (2011) 7:263–9. doi: 10.1016/j. jalz.2011.03.005
78. Hampel H, Bürger K, Teipel SJ, Bokde AL, Zetterberg H, Blennow K. Core candidate
neurochemical and imaging biomarkers of Alzheimer's disease. Alzheimers Dement. (2008)
4:38–48. doi: 10.1016/j.jalz.2007.08.006
79. Dubois B, Villain N, Frisoni GB, Rabinovici GD, Sabbagh M, Cappa S, et al. Clinical
diagnosis of Alzheimer's disease: recommendations of the international working group. Lancet
Neurol. (2021) 20:484–96. doi: 10.1016/S1474-4422(21)00066-1
80. Crismon, M.L. Tacrine: First drug approved for Alzheimer’s disease. Ann. Pharmacother.
1994, 28, 744–751. [CrossRef]
81. Qizilbash, N.; Birks, J.; Lopez Arrieta, J.; Lewington, S.; Szeto, S. Tacrine for Alzheimer’s
disease. Cochrane Database Syst. Rev. 2000, CD000202. [CrossRef]
83. Kumar, A.; Sharma, S. Donepezil. In StatPearls; StatPearls Publishing: Treasure Island, FL,
USA, 2020. Available online: https://www.ncbi.nlm.nih.gov/books/NBK513257/ (accessed on
8 December 2020).
84. Dooley, M.; Lamb, H.M. Donepezil: A review of its use in Alzheimer’s disease. Drugs
Aging 2000, 16, 199–226. [CrossRef] [PubMed]
85. Annicchiarico, R.; Federici, A.; Pettenati, C.; Caltagirone, C. Rivastigmine in Alzheimer’s
disease: Cognitive function and quality of life. Ther. Clin. Risk Manag. 2007, 3, 1113–1123.
[PubMed]
87. Khoury, R.; Rajamanickam, J.; Grossberg, G.T. An update on the safety of current therapies
for Alzheimer’s disease: Focus on rivastigmine. Ther. Adv. Drug Saf. 2018, 9, 171–178.
[CrossRef]
46
88. Birks, J.; Grimley Evans, J.; Iakovidou, V.; Tsolaki, M.; Holt, F.E. Rivastigmine for
Alzheimer’s disease. Cochrane Database Syst. Rev. 2009, CD001191. [CrossRef]
89. Scott, L.J.; Goa, K.L. Galantamine: A review of its use in Alzheimer’s disease. Drugs 2000,
60, 1095–1122. [CrossRef]
90. Prvulovic, D.; Hampel, H.; Pantel, J. Galantamine for Alzheimer’s disease. Expert Opin.
Drug Metab. Toxicol. 2010, 6, 345–354. [CrossRef]
91. Kim, J.K.; Park, S.U. Pharmacological aspects of galantamine for the treatment of
Alzheimer’s disease. Excli J. 2017, 16, 35–39. [CrossRef]
92. Wahba, S.M.; Darwish, A.S.; Kamal, S.M. Ceria-containing uncoated and coated
hydroxyapatite-based galantamine nanocomposites for formidable treatment of Alzheimer’s
disease in ovariectomized albino-rat model. Mater. Sci. Eng. C Mater. Biol. Appl. 2016, 65,
151–163. [CrossRef]
93. Liu, J.; Chang, L.; Song, Y.; Li, H.; Wu, Y. The role of NMDA receptors in Alzheimer’s
disease. Front. Neurosci. 2019, 13, 43. [CrossRef] [PubMed]
94. Huang, Y.J.; Lin, C.H.; Lane, H.Y.; Tsai, G.E. NMDA Neurotransmission dysfunction in
behavioral and psychological symptoms of Alzheimer’s disease. Curr. Neuropharmacol. 2012,
10, 272–285. [CrossRef] [PubMed]
119. Companys-Alemany, J.; Turcu, A.L.; Bellver-Sanchis, A.; Loza, M.I.; Brea, J.M.;
Canudas, A.M.; Leiva, R.; Vazquez, S.; Pallas, M.; Grinan-Ferre, C. A novel NMDA receptor
antagonist protects against cognitive decline presented by senescent mice. Pharmaceutics 2020,
12, 284. [CrossRef] [PubMed]
95. Folch, J.; Busquets, O.; Ettcheto, M.; Sanchez-Lopez, E.; Castro-Torres, R.D.; Verdaguer,
E.; Garcia, M.L.; Olloquequi, J.; Casadesus, G.; Beas-Zarate, C.; et al. Memantine for the
treatment of dementia:
96.American Psychiatric Association. Practice Guideline for the Treatment of Patients with
Alzheimer's Disease and Other Dementias. 2nd ed. Arlington, Va.: American Psychiatric
Association; 2007.
97.Knight Alzheimer's Disease Research Center. Clinical Dementia Rating.
http://alzheimer.wustl.edu/cdr/AboutCDR/aboutcdr.htm. Accessed November 29, 2010.
47
98.Caltagirone C, Bianchetti A, Di Luca M, et al. Guidelines for the treatment of Alzheimer's
disease from the Italian Association of Psychogeriatrics. Drugs Aging. 2005;22(suppl 1):1-26.
99. Gupta J., Fatima M., Islam Z., Khan R., Uversky V., Salahuddin P. Nanoparticle
formulations in the diagnosis and therapy of Alzheimer’s disease. Int. J. Biol.
Macromol. 2019;130:515–526. doi: 10.1016/j.ijbiomac.2019.02.156. [PubMed]
[CrossRef] [Google Scholar]
100. Cai J., Dao P., Chen H., Yan L., Li Y., Zhang W., Li L., Du Z., Dong C., Meunier B.
Ultrasmall superparamagnetic iron oxide nanoparticles bound NIR dyes: Novel theranostic
agents for Alzheimer’s disease. Dye. Pigment. 2020;173:107968.
doi: 10.1016/j.dyepig.2019.107968. [CrossRef] [Google Scholar]
101. Luo S., Ma C., Zhu M., Ju W., Yang Y., Wang X. Application of Iron Oxide Nanoparticles
in the Diagnosis and Treatment of Neurodegenerative Diseases With Emphasis on Alzheimer’s
Disease. Front. Cell. Neurosci. 2020;14 doi: 10.3389/fnagi.2020.00014. [PMC free
article] [PubMed] [CrossRef] [Google Scholar]
103. Maher B.A. Airborne Magnetite- and Iron-Rich Pollution Nanoparticles: Potential
Neurotoxicants and Environmental Risk Factors for Neurodegenerative Disease, Including
Alzheimer’s Disease. J. Alzheimer’s Dis. Jad. 2019 doi: 10.3233/JAD-190204. [PubMed]
[CrossRef] [Google Scholar]
104. Yang H., Mu W., Wei D., Zhang Y., Duan Y., Gao J., Gong X., Wang H., Wu X., Tao H.,
et al. A Novel Targeted and High-Efficiency Nanosystem for Combinational Therapy for
Alzheimer’s Disease. Adv. Sci. 2020;7 doi: 10.1002/advs.201902906. [PMC free
article] [PubMed] [CrossRef] [Google Scholar]
105. Bahadur S., Sachan N., Harwansh R.K., Deshmukh R. Nanoparticlized System: Promising
Approach for the Management of Alzheimer’s Disease through Intranasal Delivery. Curr.
Pharm. Des. 2020 doi: 10.2174/1381612826666200311131658. [PubMed]
[CrossRef] [Google Scholar]
48
106. Pandey M., Choudhury H., Verma R.K., Chawla V., Bhattamisra S., Gorain B., Raja
M.A., Amjad M. Nanoparticles Based Intranasal Delivery of Drug to Treat Alzheimer’s
Disease: A Recent Update. CNS Neurol. Disord. Drug Targets. 2020
doi: 10.2174/1871527319999200819095620. [PubMed] [CrossRef] [Google Scholar]
107. Liu D., Fu D., Zhang L., Sun L. Detection of amyloid-beta by Fmoc-KLVFF self-
assembled fluorescent nanoparticles for Alzheimer’s disease diagnosis. Chin. Chem. Lett. 2020
doi: 10.1016/j.cclet.2020.09.009. [CrossRef] [Google Scholar]
108. Carneiro P., Morais S., Pereira M.C. Nanomaterials towards Biosensing of Alzheimer’s
Disease Biomarkers. Nanomaterials. 2019;9:1663. doi: 10.3390/nano9121663. [PMC free
article] [PubMed] [CrossRef] [Google Scholar]
109. Liu Y., An S., Li J., Kuang Y., He X., Guo Y., Ma H., Zhang Y., Ji B., Jiang C. Brain-
targeted co-delivery of therapeutic gene and peptide by multifunctional nanoparticles in
Alzheimer’s disease mice. Biomaterials. 2016;80:33–45.
doi: 10.1016/j.biomaterials.2015.11.060. [PubMed] [CrossRef] [Google Scholar]
110. Han Q., Cai S., Yang L., Wang X., Qi C., Yang R., Wang C. Molybdenum Disulfide
Nanoparticles as Multifunctional Inhibitors against Alzheimer’s Disease. ACS Appl. Mater.
Interfaces. 2017;9:21116–21123.doi: 10.1021/acsami.7b03816. [PubMed]
[CrossRef] [Google Scholar]
111. Faustino C.M., Rijo P., Reis C. Nanotechnological strategies for nerve growth factor
delivery: Therapeutic implications in Alzheimer’s disease. Pharmacol. Res. 2017;120:68–87.
doi: 10.1016/j.phrs.2017.03.020. [PubMed] [CrossRef] [Google Scholar]
112. Moore K.A., Pate K.M., Soto-Ortega D.D., Lohse S.E., Munnik N.P., Lim M., Jackson
K.S., Lyles V.D., Jones L., Glassgow N., et al. Influence of gold nanoparticle surface chemistry
and diameter upon Alzheimer’s disease amyloid-β protein aggregation. J. Biol.
Eng. 2017;11 doi: 10.1186/s13036-017-0047-6. [PMC free article] [PubMed]
[CrossRef] [Google Scholar]
113. Gao N., Sun H., Dong K., Ren J., Qu X. Gold-nanoparticle-based multifunctional
amyloid-β inhibitor against Alzheimer’s disease. Chemistry. 2015;2:829–835.
doi: 10.1002/chem.201404562. [PubMed] [CrossRef] [Google Scholar]
49
114. Yao L., Gu X., Song Q., Wang X., Huang M., Hu M., Hou L., Kang T., Chen J., Chen H.,
et al. Nanoformulated alpha-mangostin ameliorates Alzheimer’s disease neuropathology by
elevating LDLR expression and accelerating amyloid-beta clearance. J. Control. Release Off.
J. Control. Release Soc. 2016;226:1–14. doi: 10.1016/j.jconrel.2016.01.055. [PubMed]
[CrossRef] [Google Scholar]
115. López E.H., Machado A.L., Vidal L.B., Pizarro R.G., Silva A.M., Souto E.B. Lipid
nanoparticles as carriers for the treatment of neurodegeneration associated with Alzheimer’s
disease and glaucoma: Present and future challenges. Curr. Pharm. Des. 2020;26:1235–1250.
doi: 10.2174/1381612826666200218101231. [PubMed] [CrossRef] [Google Scholar]
116. Rassu G., Soddu E., Posadino A.M., Pintus G., Sarmento B., Giunchedi P., Gavini E.
Noseto-brain delivery of BACE1 siRNA loaded in solid lipid nanoparticles for Alzheimer’s
therapy. Colloids Surf. B Biointerfaces. 2017;152:296–301. doi:
10.1016/j.colsurfb.2017.01.031. [PubMed] [CrossRef] [Google Scholar
50