Open access peer-reviewed chapter

Alzheimer’s Disease: An Insightful Review on the Future Trends of the Effective Therapeutics

Written By

Afreen Hashmi, Vivek Srivastava, Syed Abul Kalam and Devesh Kumar Mishra

Submitted: 16 September 2021 Reviewed: 19 January 2022 Published: 15 March 2022

DOI: 10.5772/intechopen.102762

From the Edited Volume

Alzheimer's Disease

Edited by Montasir Elahi

Chapter metrics overview

315 Chapter Downloads

View Full Metrics

Abstract

Alzheimer’s disease (AD) is a disorder of brain which progressively weakens the cognitive function. It is occur due to formation of β-amyloid plaques, neurofibrillary tangles, and degeneration of cholinergic neurotransmitter. There is no effective treatment capable of slowing down disease progression, current pharmacotherapy for AD only provides symptomatic relief and limited improvement in cognitive functions. Many molecules have been explored that show promising outcomes in AD therapy and can regulate cellular survival through different pathways. Present study involves current directions in the search for novel, potentially effective agents for the treatment of AD, as well as selected promising treatment strategies. These include agents acting upon the β-amyloid, such as vaccines, antibodies and inhibitors or modulators of γ- and β-secretase; agents directed against the tau protein. Current clinical trials with Aβ antibodies (solanezumab, bapineuzumab, and crenezumab) seem to be promising, while vaccines against the tau protein (AADvac1) are now in primary-stage trials. Most phase II clinical trials ending with a positive result do not succeed in phase III, often due to serious side effects or lack of therapeutic efficacy but Abucanumab (marketed as Aduhelm) now approved by USFDA in 2021 for the treatment of AD.

Keywords

  • neurodegeneration
  • novel strategies
  • clinical trials
  • medicinal plants

1. Introduction

Alzheimer’s disease (AD) is a brain disorder described in 1906 by Aloes Alzheimer, a German physician [1]. It is a progressive and neurodegenerative disorder which mainly occur in old aged people of over 65 years of age [1, 2, 3]. For progression and development of disease various pathways are involved such as formation of plaque, inflammatory cascade, cholinergic deficit, oxidative stress, and many more. Senile plaques formation and neurofibrillary tangles persist significant neuro-pathological symbols of this disease. Senile plaques are the main component of amyloid beta (Aβ) peptide that are covered by dystrophic neurites and activated microglia. Accumulation of Aβ results changed process of proteolytic amyloid precursor protein (APP) through beta and gamma secretase. The β-amyloid peptide, with 39–42 amino acid residues (BAP), perform vital role in development of AD. There are mainly two types of AD, familial AD which affects the people who have age less than 65. The other type of AD is sporadic AD which affect the people older than 65. At present there is no cure for Alzheimer’s disease but it could me managed to some level by using available medications (Table 1) [6, 7].

FactorsDementiaAlzheimer’s diseaseNormal aging
DefinitionCNS disorder due to disease or any other pathological condition.Common form of dementia.Condition occur due to programmed cell death with time (gene therapy) and causes various disability.
CauseAD, stroke, thyroid issues, vitamin deficiency, etc.Deposition of beta amyloid protein in brain.May cause biological systems to fail (DNA oxidation, DNA methylation, and apoptosis).
Duration and agePermanent damage and 65 years and olders.Average 8–20 years and 65 year but can occur as early as 30s.Gradual and progressive condition until death.
SymptomsIssues with memory, poor judgment, less focus and attention.Difficulty to remembering newly learned information.Bone break more easily, decrease overall energy, greater risk of heart stroke or hypothermia.

Table 1.

Alzheimer’s disease versus dementia and normal aging [4, 5].

1.1 Epidemiology

In 2020 approx. 50 million individual dealing with dementia worldwide. In India more than four million of people suffering from AD and dementia while in USA approx. 5.8 million living with dementia and AD. It is estimated that it is the fifth main source of death in USA and the number of death increased 146% between 2000 and 2018. It is predicted the causality will increase to 13.8 million which number of patient increases to 13.5 million by 2050. Elderly persons are more prone to younger one [8].

1.2 Etiology

In maximum case genetic lifestyle choices aging stress and environmental factors induces AD [9].

1.2.1 Age

Researchers have claimed that older adults have more risk of having AD. Scientists are still learning, how age-related changes in the brain may harm neurons and contribute to Alzheimer’s [10].

1.2.2 Genetic factors

1.2.2.1 Early onset

It occur due to mutation in chromosome 1, 14, and 21. The changes on chromosome 1 produces PRESENILIN-2 (PSEN2) named protein while chromosome 14 produces PRESENILIN-1 (PSEN1). These PSEN 1 and PSEN 2 directly and indirectly both trigger/encode for membrane protein convoluted for amyloid precursor protein. These mutations reduce the effectiveness of γ-secretase, an enzyme which is responsible for formation of beta amyloid peptide (βAP) [11]. Amyloid precursor protein is coded on chromosome 21 and this mutation results in overproduction of beta amyloid peptide. Mutation on chromosome 1, 14, and 21 results in early onset AD [12].

1.2.2.2 Late onset

Apo-lipoprotein E (APOE) gene is responsible for late onset AD. APOE gene is lipid metabolism regulator which have an affinity for beta amyloid protein and increases the risk of AD. Chromosome 19 produces APOE gene. The inheritance of APOEe4 allele own genetic risk in sporadic AD. APOEe4 allele, age elevate the risk for development of late AD by two to three folds and two copies of five folds [13].

Variations in gene for receptor sortilin, SORT1, that is important for transferring APP from surface of cell to Golgi-endoplasmic reticulum complex, have been found in familial and sporadic types of AD [14].

1.2.2.3 Environmental factors

Conditions such as heart disease, stroke, high blood pressure, diabetes, and obesity are also linked as risk factors for AD [15].

Advertisement

2. Pathogenesis and clinical findings

The real origin of this disease is not well known but problems are linked with brain protein that work abnormally and cause malfunction. As a result neurons were damaged then fail to connect other neuron as a result they die. Initially the degradation starts within the region of brain which control memory ultimately dementia occur (Figure 1) [17].

Figure 1.

Clinical findings in Alzheimer’s disease [16].

2.1 Beta-amyloid protein aggregation and deposition

In the initial stage of AD amyloid proteins works abnormally and cause overproduction of beta amyloid secretase named enzyme split amyloid processor protein and due to deviation from this process, especially sudden change in gamma and beta secretases leads to unnatural production of amyloid beta [18].

2.2 Neurofibrillary hypothesis

Tau protein is known for its stabilizing property. It is useful in the transportation of nutrients and others essential matter within the neurons while in AD. Tau protein cause mutation and changes its structure which is known as neurofibrillary tangles [18, 19].

2.3 Cholinergic hypothesis

It is observed in the patient of AD there is deficiency of ACh due to abnormal functioning of choline acetyl transferase. This will treat as a clinical hallmark to support cholinergic hypothesis there is also a possible treatment of AD by increasing the level of ACh by reducing the activity of AChE cholinergic depletion observed after neurodegenerative cascade various cholinesterase inhibitors currently used in the treatment of AD [20].

2.4 Excitotoxicity

It is defined as the excess interaction of neurotransmitter glutamate and other excitatory neurotransmitter which may act as a potent neurotoxins for Alzheimer [21].

2.5 Vascular diseases and high cholesterol

Apo-lipoprotein E play important role in the cholesterol transportation and catabolism of triglyceride lipoprotein. Cholesterol also alter the clearance of amyloid beta and generation of NFT in neuronal membrane APOE4 also enhance the deposition of beta amyloid protein. High level of cholesterol in brain there by alter the member functioning this leads to plaque formation resulting AD [22].

2.6 Oxidative stress

Oxidative stress is generated due to imbalance of ROS generation and its quenching. Brain is more prone for oxidative stress due to high consumption of O2. High level of polyunsaturated fatty acid. Low level of antioxidants and high level of redox transition metal ions. These all factors facilitate the production of reactive oxygen species like superoxide, hydrogen peroxide, etc. These ROS interact with surroundings proteins nucleic acids, etc. and cause cellular dysfunction [23]. There is also a close relationship between amyloid beta and oxidative stress because amyloid beta elevate the formation of ROS and initiate mitochondrial damage. This will also cause oxidative damage. These effects can also be observed in brain of triple transgenic mouse model of AD where tocopherol and GSH level decrease while lipid peroxidation is increased [24]. However this was observed before any plaque formation. While in another model dual mutant APP was expressed, oxidative stress and inflammation was induced by thiamine deficiency provoke plaque formation and enhance the level of amyloid [25].

2.7 Mitochondrial dysfunction

It is observed in the marphotric analysis of AD patients brain showed significant deficiency of mitochondria while its DNA and protein concentration elevate in cytoplasm and in the vacuoles associated with lipofuscin [26]. These mitochondria may be damaged due to autophagy and oxidative stress. Mitochondrial cytochrome oxidase activity also reduced in cortical region of AD brain. Due to this deficiency mitochondrial dysfunction occur and ROS generated and energy stores were decreased and ultimately neurodegeneration occur [27].

2.8 Inflammatory mediators

Amyloid deposition in brain also associated with local inflammation and immunologic alleviations [28]. This association induces the release of NO3, cytokines which cause neural damage and cause inflammation [29, 30].

Advertisement

3. Role of sex hormone in Alzheimer’s

Evidence from animal and human studies support functional roles of sex hormones like estrogens, progesterone, and androgens in behavior and cognition. With several neuroprotective activity involved, age reduces level of sex hormones were connected with greater possibilities of cognitive degeneration and AD. For example, in females development of AD is associated with decreased exposure to estrogens across the lifetime, while in males age related degeneration in both levels of peripheral and brain testosterone is linked with greater susceptibilities of AD development. Also, alterations in receptors of sex hormone and downstream signaling pathways during aging have been stated. For example, the nonfunctional splicing estrogen variants receptor alpha in the hippocampus was enhanced throughout aging and AD, with advanced levels in female old age subjects in comparison to males. Moreover, studies recognized polymorphisms of estrogen receptors related with intellectual decay and AD development in females, especially in APOE ε4 (APOE4) transporters. These information recommended diminished responsiveness of brain to sex hormones during aging and disease development. However, clinical trial outcomes of sex hormone therapy in AD are rather contentious. Despite prior studies associating protective activity of estrogen replacement against AD in females, huge clinical studies failed to exhibit any useful possessions. It was suggested that replacement of hormone initiation in the serious window of perimenopause may diminish the risks of dementia, while it might raise the risks if started a very long time after menopause. Moreover treatment timing, reduced responsiveness at receptors of brain and downstream signaling pathways might add to the uselessness of hormonal therapy. Together, these investigations recommend the complication of sex hormones association in AD [31].

Advertisement

4. Strategies used in the treatment of Alzheimer’s disease: A clinical data

4.1 Conventional approaches

Currently there is no cure for this disease, the objective of several medicine is used to reduce symptoms linked with disease and to reduce disease progression (Table 2) [32, 33, 34, 35, 36].

Drug nameIndicationMode of actionAdverse effect
DonepezilMinor to chronicIt stops the breakdown of ACh by preventing the function of acetyl cholinesteraseFatigue, abnormal dreams, hallucinations, confusion, hypertension, abdominal pain
Treats intellectual indication of AD
GalantamineMinor to mediumStops the breakdown of Ach and stimulates receptors to discharge extra AChSomnolence, bradycardia, insomnia, urinary tract infection, anorexia, syncope
Treats intellectual indication of AD
RivastigmineMinor to mediumStops the breakdown of Ach by preventing the enzymes that abolish AChDizziness, diarrhea, anxiety, vertigo, asthenia, tachycardia
Also used to treat dementia from Parkinson’s diseaseTreats intellectual indication of AD
MemantineMedium to severeBlocks glutamatergic (NMDA) receptors and controls the action of glutamateHeadache, constipation, vomiting, backache
Treats intellectual indication of AD
Donepezil/memantineMedium to severeit binds to NMDA receptor-operated caption channels, and gives therapeutic effects by preventing persistent stimulation in CNSHallucination, headache, cough, fatigue, cramping, syncope, increased frequency of bowel movements

Table 2.

Currently used drug for the treatment of Alzheimer’s disease.

4.2 Current scenario

4.2.1 Antiamyloidogenic pathway and amyloidogenic route as approaches for development of therapeutic treatments adjusting the course of Alzheimer’s disease

From the previous eras, the pharmaceutical industry has decided to chiefly focused on the amyloidocentric method, dedicating significant possessions to form useful AD drugs. Nevertheless, numerous failures of drug candidates in clinical trials have led investigators to question the viability of this approach [10, 11, 12]. Possible cause for failure is a absence of biomarkers that could consistently recognize AD in comparatively initial phases. It is totally promising that the patients presently enrolled for phase III trials are in such advanced phases of AD that any attempted interference is possibly inadequate. In the meantime, there is still a number of new management under development, that focused the amyloidogenic route. In order to decrease generation of Aβ from the APP, inhibition of γ- and β-secretase and the potentiation of activity of α-secretase have been deliberated.

4.2.2 Inhibitors and modulators of β-secretase

β-secretase enzyme complex contributes in the primary phases of the amyloidogenic APP-processing pathway. The inhibitors of β-secretase development is a task because, besides the APP, this complex has several substrates. To give just one example, neuregulin-1, that included in the CNS axons myelination and synaptic elasticity, is a target β-secretase. Substrates wide range results to substantial adverse effects, even if the precise enzyme inhibition is reached. But, E2609 (clinical trial ID# NCT01600859), MK-8931 (NCT01739348), and LY2886721 (NCT01807026 and NCT01561430) have all exposed efficiency in decreasing the production of Aβ by up to 80–90% in the cerebrospinal fluid (CSF) in humans. None of inhibitors of β-secretase have touched the market so far [37, 38, 39, 40].

4.2.3 Inhibitors and modulators of γ-secretase

In the final stage of amyloidogenesis, γ-secretase complex is responsible for the production of Aβ(1–40) and Aβ(1–42). Inhibition of γ-secretase was firstly proposed strategy for the management of Alzheimer’s disease but the substrate promiscuity shows equal issues facing γ-secretase inhibitors. γ-secretase proposed to target the Notch protein which is responsible for the regulation of cell proliferation, development, differentiation and cellular communication but off target secondary effects are major concern [41, 42, 43].

Semagacestat (LY450139) named γ-secretase inhibitor reduces the Aβ level in the blood and in cerebrospinal fluid [44]. The results obtained from the clinical study conducted on 3000 patients shows the major adverse effects like decrease cognition abilities and difficulty in the carry out daily living activities and elevated skin cancer incidence and increased risk of infection and weight loss. Another γ-secretase named avagacestat discontinued in the development stage due to lack of efficacy (NCT00810147, NCT00890890, NCT00810147, NCT01079819, [45, 46, 47]).

Several nonsteroidal anti-inflammatory drugs like indomethacin, ibuprofen, flurbiprofen, sulindac also decreases the Aβ(1–42) peptide levels in in-vivo and in in-vitro studies. Ibuprofen is a cyclooxigenase inhibitor while R-flurbiprofen (tarenflurbil) is not, so the reduction of Aβ(1–42) peptide levels is not associated with the COX inhibition. Unfortunately, in clinical trials tarenflurbil and ibuprofen does not shows efficacy for the treatment of Alzheimer’s disease. The idea of long term use of NSAID’s for the treatments of Alzheimer’s disease as NSAIDS reduces the Aβ peptide level in blood but negative results reported in the clinical studies that’s why this hypothesis requires further investigations [48, 49].

Clinical studies with 8-hydroxiquinolines compounds like clioquinol and PBT2 also conducted for the treatment of Alzheimer’s disease. The mechanism of action is yet established, but the expected MOA suggested that the increased levels of oxidative stress is due to the copper ions binding to Aβ, leading to metal-mediated generation of ROS (reactive oxygen species). It is proposed that the 8-hydroxiquinolines may prevent Aβ aggregation and restoring homeostasis in the cellular levels of copper and zinc ions. But after in clinical development these compounds failed due to lack of efficacy [50, 51, 52].

4.2.4 Agents that stimulate the removal of amyloid deposits and aggregates

Another possible treatment choice that is involved on the amyloidogenic pathway is to stimulate the existing amyloid aggregates clearance. To achieve this, three different approaches have been assessed.

4.2.5 Activation of enzymes that destroy amyloid plaques

Amyloid plaques are destroyed by various proteases comprising neprilysin, IDE, plasmin, angiotensin converting enzyme, endothelin converting enzyme, and metalloproteinases. Levels of protein these enzymes reduces in AD, that promotes accumulation and formation of Aβ. Despite being an attractive approach for forming disease-modifying medicine, no compounds with this MOA have ever entered advanced clinical development because of lack of specificity.

4.2.6 Modulation of β-amyloid transport between the brain and the peripheral circulation

Transport of Aβ between the circulation of CNS and peripheral is controlled by: (i) apolipoproteins (e.g., Aβ might be transported from the blood to the brain when it is bound to APOE); (ii) low-density lipoprotein receptor-related protein (LRP-1), that enhances Aβ discharge from the brain to the blood; (iii) receptor for progressive glycation end products (RAGE), that enables the Aβ transport across the blood-brain barrier (BBB) [53, 54].

Any treatment goal, that is determined on this mechanism, is to decrease the load of cerebral amyloid by trying to control Aβ to the peripheral circulation. To this end, a different number of approaches have been suggested, particularly the administration of LRP-1 peripherally. Though, the only drug candidates that have entered the clinical phase are the RAGE inhibitors.

4.2.7 Antiamyloid immunotherapy

4.2.7.1 Active immunotherapy

Immunotherapy approach designed to stimulate clearance of Aβ with the aimed of decreasing load of amyloid load in AD. Active immunization (vaccination) with either Aβ(1–42) (main form found in senile plaques) or other synthetic fragments has been positively assessed in transgenic mouse models of AD. Human tests were primarily hopeful; though first-generation vaccine (AN1792) treatment has shown major adverse events which results to the phase II trials cessation. AN1792 contained of a synthetic full-length Aβ(1–42) peptide with a QS-21 adjuvant. Because of a T cell-mediated autoimmune response, 6% of patients have established inflammation in brain that ended up being aseptic meningoencephalitis [55].

Second-generation vaccines were planned utilizing a limited portion of Aβ(1–6) peptide in an try to inhibit nonspecific immune response seen with the full-length vaccine. Novartis designed CAD 106, was the first second-generation vaccine which moved to development phase. Newly finished phase II trial have exposed a Aβ-specific antibody response in 75% of treated patients, without producing any side effect. Janssen developed ACC-001, has freshly finished two-phase II trials (NCT01284387 and NCT00479557) with an additional phase II trial still continuing (NCT01227564). Though, the pharmaceutical industry has canceled the ideas for this vaccine development. Further vaccines, comprising tetra-palmitoylated Aβ(1–15) re-formed in a liposome (ACI-24), MER5101 and AF205 are now in different phases of preclinical progression [56, 57, 58].

4.2.7.2 Passive immunization

It is the monoclonal or polyclonal antibodies administration directed against Aβ. This treatment contains intravenous administration of anti-Aβ antibodies to the patient. The advantage of this approach is to match to active immunization is which the proinflammatory T cell-mediated immune response should not arise. Reports have shown that in transgenic animals passive immunization decreases the load of cerebral amyloid and recovers cognition, even when the amyloid plaque numbers are not suggestively decreased. This could be recognized to the soluble amyloid oligomers neutralization, that progressively identified to play an important role in the pathophysiology of AD.

Bapineuzumab and solanezumab are two monoclonal antibodies which are reach now present in advanced phase of development. Though, two phase III trials had failed in 2012 due to low effectiveness in patients with mild-to-moderate AD [59]. Both are humanized monoclonal antibodies against Aβ(1–6) and Aβ(12–28), respectively. In bapineuzumab, noteworthy decrease in brain amyloid plaques and phosphorylated Tau in cerebrospinal fluid was stated. Though, the treatment unsuccessful to give noteworthy developments of brain function. In a solanezumab trial, infusions of 400 mg of solanezumab or placebo were given for 80 weeks once a month in patients with mild-to-moderate AD. The outcomes recommended that solanezumab might recover cognition in mild AD; but statistical significance was not attained in study. Presently solanezumab present in phase III trials in patients with AD (NCT01127633 and NCT01900665) and in older persons who have common thinking and memory function but who might be at danger of AD developing in the future (NCT02008357, [60, 61]).

Crenezumab (MABT5102A) is a humanized monoclonal antibody that uses IgG4 backbone. In April 2014 a stage II trial to measure the safety and effectiveness in patients with mild-to-moderate AD (NCT01343966) was accomplished, while the outcomes are not yet openly accessible. The supreme stage II trial pointing to assess the safety and effectiveness of crenezumab in asymptomatic transporters of E280A autosomal-dominant mutation of PSEN1 initiated in November 2013 (NCT01998841).

Other monoclonal antibodies against Aβ established so far contain PF-04360365 (ponezumab) that targets the free carboxy terminal amino acids 33–40 of the Aβ peptide; MABT5102A, that binds to Aβ monomers, oligomers, and fibrils with similarly great affinity; GSK933776A, that is likewise to bapineuzumab in which it binds to the N-terminal Aβ(1–5). Additional, other passive immunotherapies typically in stage I clinical trial involve NI-101, SAR-228810, and BAN-2401 [58, 62].

4.2.8 Approaches focused on Tau proteins

In neurons Tau proteins are extremely soluble and abundant where they play a important role in stabilization of microtubule, mainly in axons [63]. Tau hyperphosphorylation resulting the insoluble paired helical filaments (PHF) development that form neurofibrillary tangles. The microtubule-binding capacity damage initiate destabilization of cytoskeleton, that ultimately develops neurodegeneration and neuronal death [64]. As a substitute to amyloidocentric strategies, this treatments goal to prevent the phosphorylation of Tau protein. Additional, microtubule-stabilizing drugs can be utilized as a disease-modifying approach in AD. In current years, immunomodulation was recommended as a feasible choice for stimulating operative Tau aggregates clearance [65].

4.2.9 Hyperphosphorylation of Tau inhibitors

All Tau proteins are a result of different splicing of a microtubule-associated protein Tau (MAPT) gene. Primary mechanism that controls Tau binding to microtubules is phosphorylation. The protein remains soluble under physiological circumstances; though, in this disease, pathological hyperphosphorylation of Tau compromises its regular functions [66, 67]. Imbalance between the catalytic activity of kinases and phosphatases occurs hyperphosphorylation. Enhanced expression of active forms of several kinases in the areas proximal to neurofibrillary tangles has been labeled in AD, comprising CDK5, GSK3β, Fyn, stress-activated protein kinases JNK and p38, and mitogen-activated protein kinases ERK1 and ERK2 [68]. Certain kinases promote continuation of tau phosphorylation in neurofibrillary tangles. Resulting, noteworthy research determinations have been dedicated to the kinase inhibitors development as a probable treatment approach for AD. For example, SP600125, a extensively utilized pan-JNK inhibitor, employs valuable effects on cognition and decreases neurodegeneration in an APP/PS1 transgenic mouse model of AD. It has been planned which precise inhibition of JNK3 can be adequate to carry comparable benefits as seen with SP600125 in rodent models. Human data in AD patients designate a positive correlation between the JNK3 and Aβ(1–42) levels in the brain. Moreover, JNK3 upregulation was distinguished in the CSF and was related with loss of memory. Consequently, inhibition of JNK3 remains a capable goal for future treatments [69, 70, 71].

4.2.10 Tau aggregation inhibitors

Tau hyperphosphorylation contribute to neurotoxicity detected in AD brain. Methylene blue dye derivatives have revealed certain potential Tau aggregates formation inhibition. Methylene blue disturbs the Tau aggregation, has the capability to prevent amyloid aggregation, recovers the effectiveness of mitochondrial electron transport chain, decreases oxidative stress, stops mitochondrial impairment, and is also an autophagy modulator. The first-generation molecule resulting from methylene blue (Rember) seemed to stabilize AD development in a clinical trial that continued 50 weeks. These outcomes encouraged investigators to form a next-generation form of methylene blue, TRx 0237. This agents is a purified derivative of methylene blue that not only prevents aggregation of Tau protein but also liquefies brain tau aggregates. Various trials are presently ongoing (NCT01626391, NCT01689233, NCT01689246, NCT01626378) to assess the possible effectiveness of this agent in AD [72, 73].

4.2.11 Stabilizers of microtubule

Stabilization of microtubule might possibly attain a comparable end-result as which seen with the Tau hyperphosphorylation inhibitors. Paclitaxel is a microtubule-stabilizing agents presently in utilize in the oncology arena. Inappropriately, this agents is unable of BBB crossing and its utilize is related with major adverse events, that limits its efficacy in AD. In addition to paclitaxel, other microtubule-stabilizing agents like TPI 287 have been measured as a probable AD remedy. TPI 287 is a derivative of taxane, also utilize in the treatment of cancer. TPI 287 alleviates the microtubules by binding to tubulin. NCT01966666 trial will estimate TPI-287 safety, pharmacokinetic possessions, and tolerability by intravenous infusion in mild-to-moderate AD.

Epothilone D is a microtubule-stabilizing agent that enhanced axonal transport, decrease axonal dystrophy, reduced Tau neuropathology, and decreased hippocampal loss of neuron; though, in 2013 drug development for AD was discontinued after an unsuccessful clinical trial. With respect to Tau, more research are essential in order to better understand the exact molecular mechanisms elaborate in neurotoxicity of Tau. Current research associating the neurotoxic profiles of different forms of Tau recommend which is a soluble form is probable the greatest toxic. Thus, future therapeutic approaches should be focused on aiming Tau soluble forms [74].

4.2.12 Anti-Tau immunotherapy

Just as with the immunotherapies aiming Aβ, both passive and active immunization approaches against Tau have been measured. It was established that decrease in formation of Tau aggregate and enhanced Tau oligomers clearance and insoluble aggregates could all be reached with either active or passive immunotherapies. In rodents, treatment with monoclonal antibodies directed against hyperphosphorylated Tau has results to improvements in cognition and was not connected with noteworthy side effects.

Axon neuroscience began a stage I trial in 2013 to estimate the safety and tolerability of AADvac-1, an active immunotherapy that contains synthetic peptide derived from the Tau sequence coupled to keyhole limpet hemocyanin; the precise molecular nature of the antigen has not been disclosed (NCT01850238 and NCT02031198). AADvac-1 uses aluminum hydroxide as an adjuvant. At the 2014 Alzheimer’s Association International Conference (AAIC) in Copenhagen, good preclinical safety profile was reported for the treatment period of up to 6 months in rats, rabbits, and dogs. These initial outcomes are hopeful and it remains to be seen whether AADvac-1 will prove satisfactory safety and efficiency in patients [75, 76].

4.2.13 The cholinergic hypothesis

The hippocampus, the chief region of brain elaborate in memory processing, is influenced by modulation of cholinergic neurotransmitter. One of the well categorized irregularities linked with neurotransmitter deviations is the cholinergic neurons degeneration in the nucleus basalis of Meynert and the cholinergic inputs loss to the neocortex and hippocampus. Various studies reported reduced in choline acetyltransferase (ChAT), acetylcholine (ACh) release, as well as decreases in nicotinic and muscarinic receptors in the cerebral cortex and hippocampus of postmortem AD brains. Acetylcholinesterase inhibitors (AChEI), one of the only two classes of compounds that presently accepted for AD treatment, act by stimulating ACh bioavailability at the synapse. Inappropriately, none of these agents are proficient of withdrawing the course of AD nor of even noticeably reducing down the degree of disease development. Their clinical effect is basically palliative; though, their possible utilize in combination therapy with other disease-modifying agents should not be omitted [77, 78].

4.2.14 Altering the perception: AD as a metabolic disorder

As revealed by clinical study data and research articles that diabetes is a one of the key factor that leads to AD pathology and unfolds the close connection between insulin-deficient diabetes and cerebral amyloidosis. These data also suggests about insulin signaling impairments (both peripheral and central) is possibly be existing in both diseases. Hence, considering insulin hormone at the core, “type 3 diabetes” hypothesis of AD was developed, observing metabolic phenotypes into a coherent framework [79].

The most anticipated mechanisms for the development of AD due to diabetes could be: glucose toxicity, insulin resistance, oxidative stress, elevated levels of advanced glycation end products, and cytokine-mediated neuroinflammation. Recently, Clarke and colleagues demonstrated that neuroinflammatory cascades can be initiated by the administration of soluble hypothalamic Aβ oligomers that ultimately causes disturbances in peripheral glucose homeostasis. Tumor necrosis factor α (TNFα) may have a significant role during this process [80].

Rosiglitazone and pioglitazone are used as antidiabetic drugs, which regulate glucose homeostasis by increasing insulin sensitivity, reducing blood glucose levels, and improving lipid metabolism. Both compounds have also been studied as potential therapeutics for AD treatment, with reported improvements in mitochondrial oxidative metabolism [81]. In animal models, pioglitazone modified various indices of brain aging but did not slow down the cognitive decline. Rosiglitazone and pioglitazone also induce the expression of peroxisome proliferator-activated receptor-γ co-activator 1 alpha (PGC-1α), a molecule that plays multiple roles in mitochondrial biogenesis, energy metabolism, and mitochondrial antioxidants expression. Previous studies have demonstrated that, in the human brain tissues, the expression of PGC-1α decreases with progression of AD dementia. Thus, PGC-1α upregulation may improve the mitochondrial energy metabolism and AD pathology [82, 83, 84, 85, 86].

In a small scale clinical trial on mild-to-moderate AD patients, it was found that pioglitazone enhances memory and cognition. On the other hand clinical trial (phase II) with larger group of patients (who did not possess an ApoE4 allele) were on treatment with rosiglitazone (6 months) shows improvement in memory retention and attention. However, similar study (phase III trial) using rosiglitazone failed to show efficacy in AD (NCT00550420). It is important to note that rosiglitazone was administered at much lower dosage than required to exert efficacious effects on AD pathophysiology in these trials, in rodent models of the disease. NCT00348140 recently completed clinical trial in which rosiglitazone was administrated in combination with AChEIs in patients with AD (mild-to-moderate) and until now no further outcome yet reported.

As a treatment possibility for AD, intranasal insulin have also been considered as it bypasses the BBB easily; adding the advantage of possibly minimum adverse events in peripheral tissues. Theoretically it is well established that direct delivery of insulin to the brain will activate cerebral insulin signaling leading to enhancements in memory processing resulting into neuroprotection. A recent ongoing clinical trial (with NCT017679090 is assessing long-term (12 months) efficacy of intranasal insulin (Humulin R U-100) among mild AD patients [87].

Also, it has been found that reduced plasma amylin concentrations may contribute in the progression of AD. As revealed by transgenic animal models of AD, amylin and pramlintide (amylin analog) reduced the brain Aβ levels and advances cognition. Interestingly, amylin inhibits β-secretase, whereas pramlintide did not [88].

Advertisement

5. Medicinal plants for the treatment of Alzheimer’s disease

Here is the number of herbal plants reported to might have anti-Alzheimer activity (Table 3).

Sr. No.Work donePlant usedCommon nameAuthorYearRef.
1.Effects of the hydroethanolic extract of Lycopodium selago L. on scopolamine-induced memory deficits in zebrafishL. selagoFir clubmossValu et al.2021[89]
2.Evaluation of traditional herb extract Salvia officinalis in treatment of Alzheimer’s diseaseS. officinalisSageDatta et al.2020[90]
3.Protective effects of tenuifolin isolated from Polygala tenuifolia Willd roots on neuronal apoptosis and learning and memory deficits in mice with Alzheimer’s diseaseP. tenuifoliaYuan zhiWang et al.2019[91]
4.Convolvulus pluricaulis (Shankhapushpi) ameliorates human microtubule-associated protein tau (hMAPτ) induced neurotoxicity in Alzheimer’s disease Drosophila modelC. pluricaulisShankhapushpiKizhakke et al.2019[92]
5.Malva parviflora extract ameliorates the deleterious effects of a high fat diet on the cognitive deficit in a mouse model of Alzheimer’s disease by restoring microglial function via a PPAR-γ-dependent mechanismM. parvifloraCheeseweedJiménez et al.2019[93]
6.Antioxidant, anti-Alzheimer and anti-parkinson activity of Artemisia nilagirica leaves with flowering topsA. nilagiricaIndian wormwoodPal and Pradeep2018[94]
7.Antioxidant and anti-acetylcholinesterase activities of essential oils from garlic (Allium sativum) BulbsA. sativumGarlicAkinyemi et al.2018[95]
8.Nootropic activity of ethanolic extract of Alangium salvifolium leaves on scopolamine mouse model of Alzheimer’s diseaseA. salvifoliumAnkolParameshwari et al.2018[96]
9.Moringa oleifera alleviates homocysteine-induced Alzheimer’s disease-like pathology and cognitive impairmentM. oleiferaDrumstick treeMahaman et al.2018[97]
10.Ameliorative effect of Cleome gynandra L. against scopolamine induced amnesia in miceC. gynandraShonna cabbageManasa et al.2017[98]
11.Evaluation of nootropic activity of green peas in micePisum sativumGreen peasKaura et al.2017[99]
12.Ameliorative effect of Apium graveolens Linn on scopolamine-induced amnesia miceA. graveolensCeleryPhetcharat et al.2017[100]
13.Evaluation of effect of alcoholic extract of Tinospora cordifolia on learning and memory in alprazolam induced amnesia in albino miceT. cordifoliaGuduchiJyothi et al.2016[101]
14Effect of Camellia sinensis on spatial memory in a rat model of Alzheimer’s diseaseC. sinensisGreen teaMahmoodzadeh et al.2016[102]
15.Evaluation of nootropic activity of Curcuma longa leaves in diazepam and scopolamine-induced amnesic mice and ratsC. longaTurmericReddy et al.2015[103]
16.Effect of ethanolic seed extract of Bauhinia purpurea linn on cognition in scopolamine induced Alzheimer’s disease rat’s modelB. purpureaOrchid treeNemalapalli et al.2015[104]
17.Mori fructus improves cognitive and neuronal dysfunction induced by beta-amyloid toxicity through the GSK-3β pathway in vitro and in vivoM. fructusMoraKim et al.2015[105]
18.Anticholinesterase and antioxidant properties of aqueous extract of Cola acuminate seed in vitroC. acuminateCola nutOboh et al.2014[106]
19.Antiamnesic effect of piracetam potentiated with Emblica officinalis and C. longa in aluminum induced neurotoxicity of Alzheimer’s diseaseE. officinalisAamlaRamachandran et al.2013[107]
20Antiamnesic activity of Syzygium cumini against scopolamine induced spatial memory impairments in ratsS. cuminiJamunAlikatte et al.2012[108]
21Acetylcholine and memory-enhancing activity of Ficus racemosa barkF. racemosaCluster figFaiyaz et al.2011[109]
22Protective effect of Morinda citrifolia fruits on beta-amyloid (25–35) induced cognitive dysfunction in mice: an experimental and biochemical studyM. citrifoliaNoniMuralidharan et al.2010[110]

Table 3.

Plants studied in Alzheimer’s disease.

Advertisement

6. Recent advances in the treatment of Alzheimer’s disease

In 2021 USFDA approved Aducanumab (marketed as Aduhelm) for the treatment of Alzheimer’s disease. It is an amyloid beta-directed antibody approved under the accelerated approval pathway based on reductioning amyloid β plaques observed in patients treated with this drug.

It was approved for medical use in the United States. Aducanumab has since been approved by the Ministry of Health and Prevention in the United Arab Emirates as of October 3, 2021, making it the second country in the world to approve the treatment.

Pharmacology-Mechanism of Action: Immunoglobulin gamma 1 (IgG1) monoclonal antibody directed against aggregated soluble and insoluble forms of amyloid beta. The buildup of beta amyloid plaques in brain is crucial pathophysiological hallmark of Alzheimer’s disease.

Dosage Form and Strength: Aduhelm is a clear to opalescent and colorless to yellow solution, accessible as: Injection: 170 mg/1.7 mL (100 mg/mL) in a single-dose vial and 300 mg/3 mL (100 mg/mL) in a single-dose vial [111].

Who should take this drug?

It is suggested for mild cognitive impairment (MCI) or mild dementia stage of Alzheimer’s disease [112, 113].

6.1 Novel compound under investigation

Here is the figure that shows the agents which is in developing stage involve in the trials for the management of Alzheimer’s disease. Most of agents in the trial target disease modification [114] (Figure 2).

Figure 2.

Drugs in clinical trials for treatment of Alzheimer’s disease in 2021. In which the shape of icons shows the population involve in trials; the outer ring shows drugs in Phase I; the middle rings shows drugs in Phase II; the inner most ring shows drugs in Phase III trials [16].

In which the shape of icons shows the population involve in trials; the outer ring shows drugs in phase I; the middle rings shows drugs in phase II; the inner most ring shows drugs in phase III trials [115].

Advertisement

7. Conclusion

Alzheimer’s disease is serious brain disorder, at present there is no cure for this disease but currently it can be controlled by using a drugs which symptomatically treat AD. AChE inhibitors are the first approved anti-AD drugs by the FDA, and they are also the first and the most useful drug used in the clinical treatment of AD. But now few of drugs also approved by USFDA in 2021 for the treatment of AD and few also in the trial phase. Results from clinical studies have shown different new drugs in pipeline and various novel approaches may also beneficial for treating AD. Interests in the utilization of different herbal products also increase day by day. This study provides the details about recent advancement the medicinal plants against the Alzheimer’s disease. Availability of these new medicinal plants for AD will further increase the treatment options and thus provide a significant benefit to patients who remain uncontrollable to existing therapy.

References

  1. 1. Knopman DS, Petersen RC, Jack CR. A brief history of “Alzheimer disease”: Multiple meanings separated by a common name. Neurology. 2019;92(22):1053-1059
  2. 2. Hippius H, Neundörfer G. The discovery of Alzheimer’s disease. Dialogues in Clinical Neuroscience. 2003;5(1):101-108
  3. 3. Alzheimer’s Disease. Mayo Clinic. Mayo Foundation for Medical Education and Research. 2021. Available from: https://www.mayoclinic.org/diseases-conditions/alzheimers-disease/symptoms-causes/syc-20350447 [Accessed: 27 August 2021]
  4. 4. Toepper M. Dissociating normal aging from Alzheimer’s disease: A view from cognitive neuroscience. Journal of Alzheimer’s Disease. 2017;57(2):331-352
  5. 5. Anonymous. Alzheimer’s association: Alzheimer’s disease facts and figures. Alzheimers Dement. 2020;16:391-460
  6. 6. Gairola N, Kothiyal P. Alzheimer’s disease: Current perspectives. World Journal of Pharmacy and Pharmaceutical Sciences. 2018;7(3):333-349
  7. 7. Piaceri I, Nacmias B, Sorbi S. Genetics of familial and sporadic Alzheimer’s disease. Frontiers in Bioscience. 2013;1(5):167-177
  8. 8. Patel HH. Alzheimer disease epidemiology. Available from: https://www.news-medical.net/health/Alzheimers-Disease-Epidemiology.aspx
  9. 9. Edward GA, Gamez N, Escobedo G, Calderon O, Gonzalez IM. Modifiable risk factors for Alzheimer’s disease. Frontiers in Aging Neuroscience. 2019;11:146
  10. 10. Chen JH, Lin KP, Chen YC. Risk factors for dementia. Journal of the Formosan Medical Association. 2009;108(10):754-764
  11. 11. National Institute of Aging. Alzheimer’s disease genetic fact sheet. Alzheimer’s disease education and referral centre. 2019. Publication No. 11-6424 [Accessed: 12 December 2020]
  12. 12. Wang B. Gene APOE4 causes Alzheimer’s disease in humans and a fix has been created. 2018. Available from: https://www.nextbigfuture.com/2018/04/gene-apoe4-causes-alzheimers-disease-in-humans-and-a-fix-has-been-created.html [Accessed: 10 May 2021]
  13. 13. Anonymous. Alzheimer’s association: Alzheimer’s disease facts and figures. Alzheimers Dement. 2015;11:332-384
  14. 14. Nicolas G, Acuña-Hidalgo R, Keogh MJ, Quenez O, Steehouwer M, Lelieveld S, et al. Somatic variants in autosomal dominant genes are a rare cause of sporadic Alzheimer’s disease. Alzheimers Dement. 2018;14(12):1632-1639
  15. 15. Menghani YR, Bhattad DM, Chandak KK, Taksande JB, Umekar MJ. A review: Pharmacological and herbal remedies in the management of neurodegenerative disorder (Alzheimer’s). International Journal of Pharmacognosy and Life Science. 2021;2(1):18-27
  16. 16. https://calgaryguide.ucalgary.ca/alzheimers-disease-pathogenesis-and-clinical-findings/ [Accessed: 21 June 2021]
  17. 17. Imbimbo BP, Lombard J, Pomara N. Pathophysiology of Alzheimer’s disease. Neuroimaging Clinics of North America. 2005;15(4):727-753
  18. 18. Sajjad R, Arif R, Shah AA, Manzoor I, Mustafa G. Pathogenesis of Alzheimer’s disease; Role of amyloid-beta and hyperphosphorylated Tau protein. Indian Journal of Pharmaceutical Sciences. 2018;80(4):581-591
  19. 19. Tiwari S, Atluri V, Kaushik A, Yndart A, Nair M. Alzheimer’s disease: Pathogenesis, diagnostics and therapeutics. International Journal of Nanomedicine. 2019;14:5541-5554
  20. 20. Thakur AK, Kamboj P, Goswami K. Pathophysiology and management of Alzheimer’s disease: An overview. Journal of Analytical & Pharmaceutical Research. 2018;9(2):226-235
  21. 21. Lipton SA. The molecular basis of memantine action in Alzheimer’s disease and other neurologic disorders: Low-affinity, uncompetitive antagonism. Current Alzheimer Research. 2005;2:155-165
  22. 22. Santos CY, Snyder PJ, Wu WC, Zhang M, Echeverria A, Alber J. Pathophysiologic relationship between Alzheimer’s disease, cerebrovascular disease, and cardiovascular risk: A review and synthesis. Alzheimer’s & Dementia: Diagnosis, Assessment & Disease Monitoring. 2017;7:69-87
  23. 23. Zhao Y, Zhao B. Oxidative stress and the pathogenesis of Alzheimer’s disease. Oxidative Medicine and Cellular Longevity. 2013;2013:1-10
  24. 24. Singh RP, Sharad S, Kapur S. Free radicals and oxidative stress in neurodegenerative diseases: Relevance of dietary antioxidants. Journal, Indian Academy of Clinical Medicine. 2004;5(3):218-225
  25. 25. Agostinho P, Cunha RA, Oliveria C. Neuroinflammation, oxidative stress and the pathogenesis of Alzheimer’s disease. Current Pharmaceutical Design. 2010;16(25):2766-2778
  26. 26. Ortiz JMP, Swerdlow RH. Mitochondrial dysfunction in Alzheimer’s disease: Role in pathogenesis and novel therapeutic opportunities. British Pharmacological Society. 2019;176(18):3489-3507
  27. 27. Konttinen H, Mauricio CC, Ohtonen S, Wojciechowski S, Shakirzyanova A, Caligola S, et al. PSEN1ΔE9, APPswe, and APOE4 confer disparate phenotypes in human iPSC-derived microglia. Stem Cell Reports. 2019;13(4):669-683
  28. 28. Azizi G, Navabi SS, Al-Shukaili A, Seyedzadeh MH, Yazdani R, Mirshafiey A. The role of inflammatory mediators in the pathogenesis of Alzheimer’s disease. Sultan Qaboos University Medical Journal. 2015;15(3):e305-e316
  29. 29. Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM, et al. Inflammation and Alzheimer’s disease. Neurobiology of Aging. 2000;21(3):383-421
  30. 30. Perez JMR, Ruiz JMM. A review: Inflammatory process in Alzheimer’s disease, role of cytokines. The Scientific World Journal. 2012;2012:1-15
  31. 31. Guo L, Zhong MB, Zhang L, Zhang B, Cai D. Sex differences in Alzheimer’s disease: Insights from the multiomics landscape. Biological Psychiatry. 2021;19(1):61-71
  32. 32. Briggs R, Kennelly SP, O’Neill D. Drug treatments in Alzheimer’s disease. Clinical Medicine. 2016;16(3):247-253
  33. 33. Lippincott, Williams, and Wilkins. Alzheimer disease drugs. In: Karch AM, editor. Nursing 2010 Drug Handbook. Philadelphia: Wolters Kluwer Health; 2010. pp. 546-551
  34. 34. Jeffrey C, Garam L, Travis M, Aaron R, Kate Z. Alzheimer’s disease drug development pipeline: 2017. Alzheimer’s & Dementia : Translational Research & Clinical Interventions. 2017;3(3):367-384
  35. 35. https://reference.medscape.com/drug/999990 [Accessed: 21 July 2021]
  36. 36. Adlimoghaddam A, Neuendorff M, Roy B, Albensi BC. A review of clinical treatment considerations of donepezil in severe Alzheimer’s disease. CNS Neuroscience & Therapeutics. 2018;24(10):876-888
  37. 37. Menting KW, Claassen JAHR. β-secretase inhibitor: A promising novel therapeutic drug in Alzheimer’s disease. Frontiers in Aging Neuroscience. 2014;6(165):1-9
  38. 38. May PC, Willis BA, Lowe SL, Dean RA, Monk SA, Cocke PJ, et al. The potent BACE1 inhibitor LY2886721 elicits robust central Aβ pharmacodynamics responses in mice, dogs, and humans. The Journal of Neuroscience. 2015;35(3):1199-1210
  39. 39. Vassar R, Kandalepas PC. The β-secretase enzyme BACE1 as a therapeutic target for Alzheimer’s disease. Alzheimer’s Research & Therapy. 2011;3(20):1-6
  40. 40. Yan R, Vassar R. Targeting the β secretase BACE1 for Alzheimer’s disease therapy. The Lancet Neurology. 2014;13(3):319-329
  41. 41. Imbimbo BP, Giardina GAM. 𝛾-secretase inhibitors and modulators for the treatment of Alzheimer’s disease: Disappointments and hopes. Current Topics in Medicinal Chemistry. 2011;11(12):1555-1570
  42. 42. Wolfe MS. 𝛾-secretase as a target for Alzheimer’s disease. Advances in Pharmacology. 2012;64(127):127-153
  43. 43. Doody RS, Raman R, Farlow M, Iwatsubo T, Vellas B, Joffe S, et al. A phase 3 trial of semagacestat for treatment of Alzheimer’s disease. The New England Journal of Medicine. 2013;369(4):341-350
  44. 44. Coric V, Dyck CHV, Salloway S, Andreasen N, Brody M, Richter RW, et al. Safety and tolerability of the 𝛾-secretase inhibitor avagacestat in a phase 2 study of mild to moderate Alzheimer disease. Archives of Neurology. 2012;69(11):1430-1440
  45. 45. Dockens R, Wang JS, Castaneda L, Sverdlov O, Huang SP, Slemmon R, et al. A placebo-controlled, multiple ascending dose study to evaluate the safety, pharmacokinetics and pharmacodynamics of avagacestat (BMS-708163) in healthy young and elderly subjects. Clinical Pharmacokinetics. 2012;51(10):681-693
  46. 46. Tong G, Castaneda L, Wang JS, Sverdlov O, Huang SP, Slemmon R, et al. Effects of single doses of avagacestat (BMS-708163) on cerebrospinal fluid Aβ levels in healthy young men. Clinical Drug Investigation. 2012;32(11):761-769
  47. 47. Jaturapatporn D, Isaac MGEKN, McCleery J, Tabet N. Aspirin, steroidal and non-steroidal anti-inflammatory drugs for the treatment of Alzheimer’s disease. Cochrane Database of Systematic Reviews. 2012;2:CD006378
  48. 48. Miguel-´Alvarez M, Santos-Lozano A, Sanchis-Gomar F, Fiuza-Luces C, Pareja-Galeano H, Garatachea N, et al. Non-steroidal anti-inflammatory drugs as a treatment for Alzheimer’s disease: A systematic review and meta-analysis of treatment effect. Drugs & Aging. 2015;32(2):139-147
  49. 49. Pasqualetti P, Bonomini C, Forno GD, Paulon L, Sinforiani E, Marra C, et al. A randomized controlled study on effects of ibuprofen on cognitive progression of Alzheimer’s disease. Aging Clinical and Experimental Research. 2009;21(2):102-110
  50. 50. Matlack KES, Tardiff DF, Narayan P, Hamamichi S, Caldwell KA, Lindquist S. Clioquinol promotes the degradation of metal-dependent amyloid-β (Aβ) oligomers to restore endocytosis and ameliorate Aβ toxicity. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(11):4013-4018
  51. 51. Robert A, Liu Y, Nguyen M, Meunier B. Regulation of copper and iron homeostasis by metal chelators: A possible chemotherapy for Alzheimer’s disease. Accounts of Chemical Research. 2015;48(5):1332-1339
  52. 52. Ryan TM, Roberts BR, McColl G, Hare DJ, Doble PA, Li QX, et al. Stabilization of nontoxic Aβ-oligomers: Insights into the mechanism of action of hydroxyquinolines in Alzheimer’s disease. The Journal of Neuroscience. 2015;35(7):2871-2884
  53. 53. Galasko D, Bell J, Mancuso JY, Kupiec JW, Sabbagh MN, Dyck CV, et al. Clinical trial of an inhibitor of RAGE-Aβ interactions in Alzheimer disease. Neurology. 2014;82(17):1536-1542
  54. 54. Dearie R, Sagare A, Zlokovic BV. The role of the cell surface LRP and soluble LRP in blood-brain barrier Aβ clearance in Alzheimer’s disease. Current Pharmaceutical Design. 2008;14(16):1601-1605
  55. 55. Gilman S, Koller M, Black RS, Jenkins L, Griffith SG, Fox NC, et al. Clinical effects of Aβ immunization (AN1792) in patients with AD in an interrupted trial. Neurology. 2005;64(9):1553-1562
  56. 56. Wiessner C, Wiederhold KH, Tissot AC, Frey P, Danner S, Jacobson LH, et al. The second generation active Aβ immunotherapy CAD106 reduces amyloid accumulation in APP transgenic mice while minimizing potential side effects. Journal of Neuroscience. 2011;31(25):9323-9331
  57. 57. Panza F, Solfrizzi V, Imbimbo BP, Logroscino G. Amyloid-directed monoclonal antibodies for the treatment of Alzheimer’s disease: The point of no return? Expert Opinion on Biological Therapy. 2014;14(10):1465-1476
  58. 58. Panza F, Solfrizzi V, Imbimbo BP, Tortelli R, Santamato A, Logroscino G. Amyloid-based immunotherapy for Alzheimer’s disease in the time of prevention trials: The way forward. Expert Review of Clinical Immunology. 2014;10(3):405-419
  59. 59. Salloway S, Sperling R, Fox NC, et al. Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer’s disease. The New England Journal of Medicine. 2014;370(4):322-333
  60. 60. Doody RS, Thomas RG, Farlow M, Iwatsubo T, Vellas B, Joffe S, et al. Phase 3 trials of solanezumab for mild-to-moderate Alzheimer’s disease. The New England Journal of Medicine. 2014;370(4):311-321
  61. 61. Tayeb HO, Murray ED, Price BH, Tarazi FI. Bapineuzumab and solanezumab for Alzheimer’s disease: Is the ‘amyloid cascade hypothesis’ still alive? Expert Opinion on Biological Therapy. 2013;13(7):1075-1084
  62. 62. Jindal H, Bhatt B, Malik JS. Alzheimer disease immunotherapeutics: Then and now. Human Vaccines & Immunotherapeutics. 2014;10(9):2741-2743
  63. 63. Cowan CM, Mudher A. Are tau aggregates toxic or protective in tauopathies? Frontiers in Neurology. 2013;4(114):1-13
  64. 64. West S, Bhugra P. Emerging drug targets for Aβ and tau in Alzheimer’s disease: A systematic review. British Journal of Clinical Pharmacology. 2015;80(2):221-234
  65. 65. Shefet-Carasso L, Benhar I. Antibody-targeted drugs and drug resistance-challenges and solutions. Drug Resistance Updates. 2015;18:36-46
  66. 66. Mehta DC, Short JL, Hilmer SN, Nicolazzo JA. Drug access to the central nervous system in Alzheimer’s disease: Preclinical and clinical insights. Pharmaceutical Research. 2015;32(3):819-839
  67. 67. Berk C, Paul G, Sabbagh M. Investigational drugs in Alzheimer’s disease: Current progress. Expert Opinion on Investigational Drugs. 2014;23(6):837-846
  68. 68. Gr¨uninger F. Invited review: Drug development for tauopathies. Neuropathology and Applied Neurobiology. 2015;41(1):81-96
  69. 69. Iqbal K, Gong CX, Liu F. Microtubule-associated protein tau as a therapeutic target in Alzheimer’s disease. Expert Opinion on Therapeutic Targets. 2014;18(3):307-318
  70. 70. Kimura T, Ishiguro K, Hisanaga SI. Physiological and pathological phosphorylation of tau by Cdk5. Frontiers in Molecular Neuroscience. 2014;7(65):1-10
  71. 71. Resnick L, Fennell M. Targeting JNK3 for the treatment of neurodegenerative disorders. Drug Discovery Today. 2004;9(21):932-939
  72. 72. Baddeley TC, McCaffrey J, Storey JMD, Cheung JKS, Melis V, Horsley D, et al. Complex disposition of methylthioninium redox forms determines efficacy in tau aggregation inhibitor therapy for Alzheimer’s disease. The Journal of Pharmacology and Experimental Therapeutics. 2015;352(1):110-118
  73. 73. Wischik CM, Staff RT, Wischik DJ, Bentham P, Murray AD, Storey JMD, et al. Tau aggregation inhibitor therapy: An exploratory phase 2 study in mild or moderate Alzheimer’s disease. Journal of Alzheimer’s Disease. 2015;44(2):705-720
  74. 74. Shemesh OA, Spira ME. Rescue of neurons from undergoing hallmark tau-induced Alzheimer’s disease cell pathologies by the antimitotic drug paclitaxel. Neurobiology of Disease. 2011;43(1):163-175
  75. 75. Wisniewski T, Goni F. Immunotherapeutic approaches for Alzheimer’s disease. Neuron. 2015;85(6):1162-1176
  76. 76. Kontsekova E, Zilka N, Kovacech B, Novak P, Novak M. First-in-man tau vaccine targeting structural determinants essential for pathological tau-tau interaction reduces tau oligomerisation and neurofibrillary degeneration in an Alzheimer’s disease model. Alzheimer’s Research & Therapy. 2014;6(4):44
  77. 77. Tata AM, Velluto L, D’angelo C, Reale M. Cholinergic system dysfunction and neurodegenerative diseases: Cause or effect? CNS & Neurological Disorders Drug Targets. 2014;13(7):1294-1303
  78. 78. Wallace TL, Bertrand D. Importance of the nicotinic acetylcholine receptor system in the prefrontal cortex. Biochemical Pharmacology. 2013;85(12):1713-1720
  79. 79. Lourenco MV, Ferreira ST, DeFelice FG. Neuronal stress signaling and eIF2𝛼 phosphorylation as molecular links between Alzheimer’s disease and diabetes. Progress in Neurobiology. 2015;129:37-57
  80. 80. Hokama M, Oka S, Leon J. Altered expression of diabetes-related genes in Alzheimer’s disease brains: The Hisayama study. Cerebral Cortex. 2014;24(9):2476-2488
  81. 81. Lourenco MV, Clarke JR, Frozza RL, Bomfim TR, Forny-Germano L, Batista AF, et al. TNF-𝛼 mediates PKR-dependent memory impairment and brain IRS-1 inhibition induced by Alzheimer’s β-amyloid oligomers in mice and monkeys. Cell Metabolism. 2013;18(6):831-843
  82. 82. Gold M, Alderton C, Zvartau-Hind M, Egginton S, Saunders AM, Irizarry M, et al. Rosiglitazone monotherapy in mild-to-moderate Alzheimer’s disease: Results from a randomized, double-blind, placebo-controlled phase III study. Dementia and Geriatric Cognitive Disorders. 2010;30(2):131-146
  83. 83. Blalock EM, Phelps JT, Pancani T, Searcy JL, Anderson KL, Gant JC, et al. Effects of longterm pioglitazone treatment on peripheral and central markers of aging. PLoS One. 2010;5(4):e10405
  84. 84. Qin W, Haroutunian V, Katsel P, Cardozo CP, Ho L, Buxbaum JD, et al. PGC-1𝛼 expression decreases in the Alzheimer disease brain as a function of dementia. Archives of Neurology. 2009;66(3):352-361
  85. 85. Katsouri L, Parr C, Bogdanovic N, Willem M, Sastre M. PPAR𝛾 co-activator-1𝛼 (PGC-1𝛼) reduces amyloid-β generation through a PPAR𝛾-dependent mechanism. Journal of Alzheimer’s Disease. 2011;25(1):151-162
  86. 86. Sato T, Hanyu H, Hirao K, Kanetaka H, Sakurai H, Iwamoto T. Efficacy of PPAR-𝛾 agonist pioglitazone in mild Alzheimer disease. Neurobiology of Aging. 2011;32(9):1626-1633
  87. 87. Claxton A, Baker LD, Hanson A, Trittschuh EH, Cholerton B, Morgan A, et al. Long-acting intranasal insulin detemir improves cognition for adults with mild cognitive impairment or early-stage Alzheimer’s disease dementia. Journal of Alzheimer’s Disease. 2015;44(3):897-906
  88. 88. Qiu WQ, Zhu H. Amylin and its analogs: A friend or foe for the treatment of Alzheimer’s disease? Frontiers in Aging Neuroscience. 2014;6:186
  89. 89. Valu M, Ducu C, Moga S, Negree D, Hritcu L, Boiangiu RS, et al. Effects of the hydroethanolic extract of Lycopodium selago L. on scopolamine-induced memory deficits in Zebrafish. Multidisciplinary Digital Publishing Institute. 2021;14(6):1-22
  90. 90. Datta S, Patil S. Evaluation of traditional herb extract Salvia officinalis in treatment of Alzheimer’s disease. Pharmacognosy Journal. 2020;12(1):131-143
  91. 91. Wang L, Jin GF, Yu HH, Lu XH, Zou ZH, Liang JQ, et al. Protective effects of tenuifolin isolated from Polygala tenuifolia Willd roots on neuronal apoptosis and learning and memory deficits in mice with Alzheimer’s disease. Food & Function. 2019;10(11):7453-7460
  92. 92. Kizhakke AP, Olakkaran S, Antony A, Tilagul S, Hunasanahally G. Convolvulus pluricaulis (Shankhapushpi) ameliorates human microtubule-associated protein tau (hMAPτ) induced neurotoxicity in Alzheimer’s disease Drosophila model. Journal of Chemistry Neuroanatomy. 2019;95:115-122
  93. 93. Medrano-Jiménez E, Carrillo IJF, Pedraza-Escalona M, Ramírez-Serrano CE, Álvarez-Arellano L, Cortés-Mendoza J, et al. Malva parviflora extract ameliorates the deleterious effects of a high fat diet on the cognitive deficit in a mouse model of Alzheimer’s disease by restoring microglial function via a PPAR-γ-dependent mechanism. Journal of Neuroinflammation. 2019;16(143):1-26
  94. 94. Pal G, Pradeep AK. Antioxidant, anti-Alzheimer and anti-Parkinson of Artemisia nilagirica leaves with flowering tops. UK Journal of Pharmaceutical and Biosciences. 2018;6(2):12-23
  95. 95. Akinyemi AJ, Faboya L, Awonegan AP, Olayide I. Antioxidant and anti-acetylcholinesterase activities of essential oils from garlic (Allium sativum) bulbs. International Journal of Plant Research. 2018;31(2):1-10
  96. 96. Parameshwari K, Kumar S, Priyadharshini Bai G, Prathima C, Neetha C. Nootropic activity of ethanolic extract of Alangium salvifolium leaves on Scopolamine mouse model of Alzheimer’s disease. National Journal of Physiology, Pharmacy and Pharmacology. 2018;8(12):1625-1630
  97. 97. Mahaman YAR, Huang F, Wu M, Wang Y, Wei Z, Bao J, et al. Moringa oleifera alleviates homocysteine-induced Alzheimer’s disease-like pathology and cognitive impairment. Journal of Alzheimer’s Disease. 2018;63(3):1141-1159
  98. 98. Manasa A, Karimulla SK, Gobinath M. Ameliorative effect of Cleome gynandra Linn against scopolamine induced amnesia in mice. International Journal of Research in Pharmacy and Science. 2017;8(4):642-649
  99. 99. Kaura S, Parle M. Evaluation of nootropic potential of green peas in mice. Journal of Applied Pharmaceutical Science. 2017;7(5):166-173
  100. 100. Phetcharat B, Wanida S, Supita T, Pennapa C. Ameliorative effect of Apium graveolens Linn on scopolamine-induced amnesia mice. International Conference on Herbal and Traditional Medicine. 2017;10(10):82-93
  101. 101. Jyothi CH, Shashikala G, Vidya HK, Shashikala GH. Evaluation of effect of alcoholic extract of Tinospora cordifolia on learning and memory in alprazolam induced amnesia in albino mice. International Journal of Basic & Clinical Pharmacology. 2016;5(5):2159-2163
  102. 102. Mahmoodzadeh T, Kashani MHK, Ramshini H, Moslem A, Zadeh MM. Effect of Camellia sinensis on spatial memory in a rat model of Alzheimer’s disease. Journal of Biomedical Science. 2016;1(1):e5340
  103. 103. Reddy N, Sultanpur CM, Saritha V. Evaluation of nootropic activity of Curcuma longa leaves in diazepam and scopolamine-induced amnesic mice and rats. International Journal of Basic & Clinical Pharmacology. 2015;4(4):714-719
  104. 104. Nemalapalli Y, Shaik A, Kadrivel D, Kumar D, Sundararajan P, Balagani PK. Effect of ethanolic seed extract of Bauhinia purpurea Linn on cognition in scopolamine induced Alzheimer’s disease rat’s model. Journal of Comprehensive Pharmacy. 2015;2(4):145-150
  105. 105. Kim HG, Park G, Lim S, Park H, Choi JG, UkJeong H, et al. Mori fructus improves cognitive and neuronal dysfunction induced by beta-amyloid toxicity through the GSK-3β pathway in vitro and in vivo. Journal of Ethnopharmacology. 2015;171:196-204
  106. 106. Oboh G, Ayodele J, Akinyemi OS, Oyeleye S. Anticholinesterase and antioxidative properties of aqueous extract of Cola acuminata seed in vitro. International Journal of Alzheimer’s Disease. 2014:498629
  107. 107. Ramachandran S, Sanjay S, Dhanaraju M. Antiamnesic effect of piracetam potentiated with Emblica officinalis and Curcuma longa in aluminium induced neurotoxicity of Alzheimer’s disease. International Journal of Advanced Research. 2013;1(7):185-196
  108. 108. Alikatte KL, Akondi BR, Yerragunta VG, Veerareddy PR, Palle S. Antiamnesic activity of Syzygium cumini against scopolamine induced spatial memory impairments in rats. Brain & Development. 2012;34(10):844-851
  109. 109. Faiyaz A, Narendra J, Sharath C. Acetylcholine and memory-enhancing activity of Ficus racemosa bark. Pharmacognosy Research. 2011;3(4):246-249
  110. 110. Muralidharan P, Kumar VR, Balamurugan G. Protective effect of Morinda citrifolia fruits on β-amyloid (25-35) induced cognitive dysfunction in mice: An experimental and biochemical study. Phytotherapy Research. 2010;24(2):252-258
  111. 111. https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=41706573-546f-6774-6872-5374726f6e67 [Accessed: 18 January 2022]
  112. 112. Alzheimer’s Association. Aducanumab approved for treatment of Alzheimer’s disease. Available from: https://www.alz.org/alzheimers-dementia/treatments/aducanumab [Accessed: 10 September 2021]
  113. 113. https://jamanetwork.com/journals/jamaneurology/fullarticle/2783263 [Accessed: 10 September 2021]
  114. 114. Huang LK, Chao SP, Hu CJ. Clinical trials of new drugs for Alzheimer disease. Journal of Biomedical Science. 2020;27(18):1-13
  115. 115. Cumming J, Lee G, Zhong K, Fonseca J, Taghva K. Alzheimer’s disease drug development pipeline: 2021. Alzheimer’s & Dementia. 2021;7:e12179

Written By

Afreen Hashmi, Vivek Srivastava, Syed Abul Kalam and Devesh Kumar Mishra

Submitted: 16 September 2021 Reviewed: 19 January 2022 Published: 15 March 2022