Recent Advances In Tacrine-Based Anti-Alzheimer’s Drug Design
Page: 1-26 (26)
Author: Atukuri Dorababu*
DOI: 10.2174/9789815136142123010004
PDF Price: $15
Abstract
Alzheimer’s has become a common disease in aged people that leads to
cognitive impairment and finally results in dementia and death. As the disease has a
complicated etiology, it can hardly be prevented and cured. Hence, it turned out to be
one of the menacing neurodegenerative diseases. The important concerning factor
about Alzheimer’s is its unaffordable treatment cost. Also, there are only a few
efficient anti-Alzheimer drugs. Now, it is a very urgent need to discover the most
efficient and cost-effective anti-Alzheimer’s drugs. Nowadays, research reveals drugs
based on heterocyclic scaffolds that have attributed to potent pharmacology.
Quinoline-containing molecule, tacrine was recommended as an acetylcholinesterase
inhibitor. However, its use has been withdrawn because of its toxicity. While research
is going on designing derivatives of tacrine. Fortunately, some tacrine derivatives
showed the most potent anti-Alzheimer properties. In view of this, here, anti-Alzheimer
properties of recently reported tacrine-based Alzheimer’s agents are discussed and
evaluated. The structure-activity relationship has been helpful in identifying potent
molecules in a series of derivatives.
Epigenetics of Alzheimer’s Disease: Past, Present and Future
Page: 27-72 (46)
Author: Divya Adiga, Sangavi Eswaran, S. Sriharikrishnaa, Nadeem G. Khan, Shama Prasada Kabekkodu* and Dileep Kumar
DOI: 10.2174/9789815136142123010005
PDF Price: $15
Abstract
Alzheimer’s disease (AD) exemplifies a looming epidemic lacking effective
treatment and manifests with the accumulation of neurofibrillary tangles, amyloid-β
plaques, neuroinflammation, behavioral changes, and acute cognitive impairments. It is
a complex, multifactorial disorder that arises from the intricate interaction between
environment and genetic factors, restrained via epigenetic machinery. Though the
research progress has improved the understanding of clinical manifestations and
disease advancement, the causal mechanism of detrimental consequences remains
undefined. Despite the substantial improvement in recent diagnostic modalities, it is
challenging to distinguish AD from other forms of dementia. Accurate diagnosis is a
major glitch in AD as it banks on the symptoms and clinical criteria. Several studies are
underway in exploring novel and reliable biomarkers for AD. In this direction,
epigenetic alterations have transpired as key modulators in AD pathogenesis with the
impeding inferences for the management of this neurological disorder. The present
chapter aims to discuss the significance of epigenetic modifications reported in the
pathophysiology of AD such as DNA methylation, hydroxy-methylation, methylation
of mtDNA, histone modifications, and noncoding RNAs. Additionally, the chapter also
describes the possible therapeutic avenues that target epigenetic modifications in AD.
TMP21 in Alzheimer’s Disease: An Important Target For Effective Treatment Approach
Page: 73-92 (20)
Author: Dipanjan Karati and Dileep Kumar*
DOI: 10.2174/9789815136142123010006
PDF Price: $15
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia, and it is
considered a dynamic cognitive decline. Neurofibrillary tangles and nerve cell injury
are important neuropharmacological symptoms for one AD brain. TMP21 is an
important molecule in cellular protein trafficking. TMP21, a protein involved in the
production of neurotic plaques, appears to be dysregulated in AD. As a result, we want
to look into TMP21 dysregulation in Alzheimer's disease, as well as the involvement of
TMP21 in neurotic plaque development and the underlying mechanisms. TMP21's
significance in the creation of neurofibrillary tangles, synaptic disbalance, and nerve
cell death is also explored. It will shed light on the therapeutic potential of regulating
TMP21 as a treatment for AD.
Tubulin Modifying Enzymes as Target for the Treatment of Alzheimer's Disease: Old Perspective With A New Angle
Page: 93-110 (18)
Author: Shweta Shrivastava*, Ayush Kumar, Manish Kumar Jeengar and Chandraprabha Sahu
DOI: 10.2174/9789815136142123010007
PDF Price: $15
Abstract
Alzheimer's disease (AD) is a major cause of mental disability in the elderly,
accounting for 50-60% of all dementia. While β-amyloid plaques as well as
neurofibrillary tangles are neuropathological markers, inflammation plays a critical role
in AD development. The aberrant detachment of microtubules (MTs) from axon MTs,
cellular mislocalization, and hyperphosphorylation of tau are major factors in
neurodegeneration death. Tau's ability to aggregate as well as form NFTs is assumed to
be regulated by post-translational changes, which are regarded to be an essential
regulatory mechanism. So far, drugs that target tau phosphorylation as well as
aggregation have not shown therapeutic impact. It is now clear that tubulin PTMs cause
tau dysfunction. High glutamylation and detyrosination levels in the neurons affect MT
surface physicochemical characteristics. Further evidence for the relevance of such an
enzymatic machinery in neurobiology comes from the recent discovery of harmful
mutations in enzymes involved in surface MT modification. In this chapter, we
discussed that targeting tubulin-modifying enzymes pharmacologically may be useful
in treating neurodegenerative disorders.
Memantine and Glutamate Antagonists in the Treatment of Alzheimer's Disease: Current Updates
Page: 111-120 (10)
Author: Rakesh Kore, Priya Tiwari, Vijay K Patel, Ekta Shirbhate, Ravichandran Veerasamy, Achal Mishra and Harish Rajak*
DOI: 10.2174/9789815136142123010008
PDF Price: $15
Abstract
Alzheimer’s disease (AD) is the most important cause of dementia and a
complex chronic neurodegenerative disease. Many of the currently marketed drugs are
used to treat this disease condition, but a major issue with these drugs is their
neurotoxicity. Alzheimer's treatment with the FDA approval of memantine resolves the
neurotoxicity issue. Memantine acts on glutamate and its receptors in the treatment of
AD. Recent studies show that NMDA receptor-acting drugs are doing well in the
healing of Alzheimer's patients, because of their selectivity on receptor and
neuroprotective activity. The present work is an attempt to collect updated information
about memantine and glutamate antagonists used for the treatment of AD.
Enzymatic Targets for Drug Discovery Against Alzheimer's Disease
Page: 121-158 (38)
Author: Ahmet Ozan Ozgen and Ozan Emre Eyupoglu*
DOI: 10.2174/9789815136142123010009
PDF Price: $15
Abstract
Alzheimer’s Disease (AD) is a neurodegenerative disease. The disease itself
is progressive and full recovery from it isn’t achievable yet. There are several
hypotheses asserted (Cholinergic hypothesis, Amyloid hypothesis etc.) to explain the
mechanisms behind the disease. Also, many targets have been identified for possible
therapeutics and from these targets, numerous drug candidates have been evaluated in
clinical trials. Unfortunately, most of these trials failed due to the enigmatic nature of
this disease. Currently, there are 7103 targets associated with Alzheimer's disease listed
in the Open Targets platform where 1240 of them are enzyme-related. In this chapter,
enzymatic targets of the AD have been reviewed, and those claimed to have disease modifying effects were selected and presented according to their clinical significance.
Tau Protein: Targets And Development Against Alzheimer’s Disease
Page: 159-180 (22)
Author: Sonal Dubey* and Mahesh AR
DOI: 10.2174/9789815136142123010010
PDF Price: $15
Abstract
The clinical manifestations of Alzheimer's disease (AD) and associated
human tauopathies are driven by tau neuronal and glial abnormalities. Tau, a
microtubule-associated protein is inherently disordered due to its lack of a stable
structure and great flexibility. Intracellular inclusions of fibrillar tau with a sheet shape
accumulate in the brains of individuals with AD and other tauopathies. As a result, tau
separation from microtubules and tau transition from a disordered state to an
inappropriately aggregated state are critical steps before the start of tau-related
illnesses. Many studies have demonstrated that this shift is triggered by post translational changes such as hyperphosphorylation and acetylation. Before the
development of tau inclusions, the misfolded tau self-assembles and forms a tau
oligomer. Animal and clinical research utilising human samples has shown that tau
oligomer development contributes to neuronal death. During tauopathies, tau seeds are
released from cells and absorbed into neighbouring cells, resulting in the spread of
abnormal tau aggregation. Thus, Tau has become both a physiological and pathological
target for AD treatments during the last decade. Evidence reveals many potential
techniques for preventing tau-mediated toxicity: (1) direct suppression of pathological
tau aggregation; (2) inhibition of tau post-translational changes that occur before
pathological tau aggregation; (3) inhibition of tau propagation; and (4) microtubule
stabilisation. Aside from traditional low-molecular-weight compounds, newer drug
discovery approaches, such as the development of medium-molecular-weight drugs
(peptide- or oligonucleotide-based drugs) and high-molecular-weight drugs (antibody based drugs), provide alternative pathways to preventing the formation of abnormal
tau. Suppression of protein kinases or protein-3-O-(N-acetyl-beta-D-glucosaminl)-L-serine/threonine hydrolase, inhibition of tau aggregation, active and passive
immunotherapies, and tau silencing using antisense oligonucleotides; in several animal
models, have shown the capacity to prevent or minimise tau lesions and treat either
cognitive or motor impairment. Immunotherapy, which has already reached the clinical
stage of drug development, is the most advanced technique for treating human
tauopathies. Tau vaccines or humanised antibodies are designed to target a range of tauspecies in both intracellular and extracellular environments. Some of them recognise
the amino- or carboxy-terminus, while others have proline-rich areas or microtubule binding domains that they can attach to. In this review, we examine various clinical
targets for the treatment of tauopathies as well as the various molecules researched as
tau inhibitors that can be used in AD. Furthermore, we explore the efficacy of some of
the prominent molecules in clinical studies for tau-targeted therapies research.
Promising Nano-Carriers-Based Targeted Drug Delivery Approaches for the Effective Treatment of Alzheimer’s Disease
Page: 181-204 (24)
Author: Yogita Kumari, Khushboo Raj and Pankaj Kumar Singh*
DOI: 10.2174/9789815136142123010011
PDF Price: $15
Abstract
Alzheimer’s disease (AD) is an attained disorder of cognitive and behavioral
impingement with progressive symptoms over time. It is mostly witnessed in elderly
people, and as per the World Health Organization (WHO), it has affected more than 35
million people worldwide, and this figure is presumed to double by the year 2050. The
most commonly believed cause of AD is the accumulation of beta-amyloid, which
forms extracellular plaques. Presently conventional therapy for treating cognitive
impairments in AD relies on a neurotransmitter or enzyme modulation strategy.
Conventional approved drugs, such as acetylcholinesterase inhibitors (memantine,
tacrine), are widely available for the treatment of mild to moderate AD, but due to their
lower bioavailability, poor solubility, and ineffective capability to surpass the blood brain barrier (BBB), they often fail to produce the desired effect. The potency of
conventional AD drugs is highly dependent on various physiological aspects such as
BBB; blood-cerebrospinal fluid barrier and drug efflux by P-glycoprotein, which all
hampers the capabilities of AD drugs to grasp the central nervous system (CNS). So, in
order to conquer the hurdle and these existing limitations faced by CNS drugs to cross
the BBB, innovative pathways in drug development have become the need of the hour.
Various nanocarriers based approaches profitably meet this demand by improving the
efficacy as well as facilitating the sustained release of the entrapped AD drug via
targeted drug delivery. The blood-brain barrier offers protection to the central nervous
system and also limits the entry of therapeutic molecules to the CNS. On the other
hand, nanotechnology offers the possibility to deliver small molecules against CNS
disorders across BBB due to their enormous properties, such as small surface area,
controllable physicochemical properties, higher drug payload, and better drug
circulation time. Plenty of nanocarriers and nanoparticle prodrugs have been reported
to have inconsequential cytotoxicity in preclinical studies, and these advancements
have proclaimed a new juncture for the development of new classes of nano carriers’
based potent drug formulations for the treatment of AD. A plethora of nanotechnology-based approaches such as polymers, emulsions, lipo-carriers, solid lipid carriers, carbon
nanotubes, and metal-based carriers have been redefined over time, and they have been successfully focusing on both neuroprotective and neurogenerative techniques for
treating AD. Many researchers also reported that nanotechnological-based techniques
can improve the early diagnosis of AD and enhance the therapeutic efficacy and
bioavailability of drugs.
Effects of Carbonic Anhydrase Inhibitors on Mitochondrial Dysfunction and Consequently on Alzheimer’s Disease
Page: 205-220 (16)
Author: Devyani Bhatnagar, Shreya Ladhe and Dileep Kumar*
DOI: 10.2174/9789815136142123010012
PDF Price: $15
Abstract
With the discovery of Carbonic Anhydrase (CA) and its isoenzymes in
various Alzheimer’s disease (AD) models and the brain of AD patients, the role of CA
in AD pathology has become of keen interest among scholars around the world.
Several experiments were performed to investigate the same, albeit they didn’t provide
us with the exact mechanism through which CAs are involved in AD progression, but
they gave us an important insight into the beneficial outcomes of CA inhibition.
Carbonic Anhydrase Inhibitor (CAI) administration showed a significant reduction in
the release of the proapoptotic factor- Cytochrome C (cyt C) from the challenged
mitochondria (under oxidative stress). Thus, a link between ageing, oxidative stress,
mitochondria dysfunction and pathogenesis of Alzheimer’s disease was established.
Treatment with CAI indirectly lowers neuronal loss and, thus, cognitive impairment,
which are characteristic features of AD. Though, the precise functions of CA in
exaggerating or mediating AD still remain hazy, with the support of various
scholarships globally, the use of CAII (an isoenzyme of CA) as a potential biomarker
for AD can be proposed.
Novel Treatment for Alzheimer’s Disease: Tapping the Somatostatin-evoked Aβ Catabolism via α-endosulfine-KATP Channel Pathway
Page: 221-243 (23)
Author: Ryan Varghese, Gargi Digholkar, Abha Deshpande and Dileep Kumar*
DOI: 10.2174/9789815136142123010013
PDF Price: $15
Abstract
Alzheimer's disease (AD) is a debilitating neurological disease that is known to worsen as people age. As a chronic illness, it has a negative impact on the health and financial well-being of patients and their families. Despite decades of research into new medications and therapy regimens, the therapeutic choices for these conditions are still limited. Although currently available medications for AD do not prevent or stop disease progression, they are used to treat symptoms and provide brief comfort to patients. The development of medications and other therapy modalities to address the unmet medical need has sparked a surge of interest in understanding the mechanism of AD in recent years. Growing bodies of evidence direct towards the treatment of AD by intercepting the Somatostatin-evoked Aβ catabolism in the brain, via the α-endosulfin-KATP channel pathway. The latter can be achieved through the repurposing or repositioning of drugs previously approved by the regulatory authorities and indicated in other diseases. With the advent of technology in the healthcare sector, these could be corroborated through various in-silico and in-vitro techniques. This article aims to explore the various aspects of the byzantine α-endosulfine-KATP channel pathway while providing information and future prospects for the development of new therapies to combat AD.
Diagnosis and Potential Strategies to Discover New Drugs for the Treatment of Alzheimer’s Disease (AD)
Page: 244-265 (22)
Author: Kavya Manjunath*, Arvinder Kaur, Deepa Bagur Parmesh and Shilpa Murthy
DOI: 10.2174/9789815136142123010014
PDF Price: $15
Abstract
Alzheimer disease (AD) is most common cause of dementia, which is
characterized by impaired cognitive and behavioural charateristics. Deposition of Aβ
plaques and neurofibrillary tangs (NFTs) are the hallmark of AD. Generally it is a
chronic disease where neurodegeneration, and loss of neuronal function arise earlier
before it is diagnosed. Early detection of AD is important as it reduces the severity of
the disease. In this regard, an effective tools/methods are available including CSF
biomarkers, Magnetic Resonance imaging (MRI), Positron emission tomography (PET)
but all these methods are painful and often cannot be afforded by the patients.
Therapy of AD includes inhibitors of choline esterases, and antagonists at NMDA
receptors. From the studies it is shown that these drugs just offer relief from symptoms
rather than alleviating the progression of disease. Multiple pathological processes
contribute for AD, like oxidative stress, dysregulation of neurotransmitters,
inflammation of neurons, aggregation β-amyloid, phosphorylation of tau protein. It is
essential to target multiple causes for an effective outcome in the treatment of AD.
Early diagnosis is also crucial as it reduces disease progression thereby cost involved in
AD therapy.
This review focuses on non-invasive, patient affordable diagnosis methods and also
potential targets to discover new drugs beyond conventional and available drugs.
Subject Index
Page: 266-271 (6)
Author: Dileep Kumar and Prashant Tiwari
DOI: 10.2174/9789815136142123010015
Introduction
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.