Next Article in Journal
The Change in Whole-Genome Methylation and Transcriptome Profile under Autophagy Defect and Nitrogen Starvation
Previous Article in Journal
Genomic and Transcriptomic Insights into the Evolution and Divergence of MIKC-Type MADS-Box Genes in Carica papaya
Previous Article in Special Issue
Neurodegenerative Diseases: Molecular Mechanisms and Therapies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advancements in the Application of Nanomedicine in Alzheimer’s Disease: A Therapeutic Perspective

Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(18), 14044; https://doi.org/10.3390/ijms241814044
Submission received: 22 August 2023 / Revised: 11 September 2023 / Accepted: 11 September 2023 / Published: 13 September 2023
(This article belongs to the Special Issue Neurodegenerative Diseases: Molecular Mechanisms and Therapies)

Abstract

:
Alzheimer’s disease (AD) is a progressive neurodegenerative disease that affects most people worldwide. AD is a complex central nervous system disorder. Several drugs have been designed to cure AD, but with low success rates. Because the blood–brain and blood–cerebrospinal fluid barriers are two barriers that protect the central nervous system, their presence has severely restricted the efficacy of many treatments that have been studied for AD diagnosis and/or therapy. The use of nanoparticles for the diagnosis and treatment of AD is the focus of an established and rapidly developing field of nanomedicine. Recent developments in nanomedicine have made it possible to effectively transport drugs to the brain. However, numerous obstacles remain to the successful use of nanomedicines in clinical settings for AD treatment. Furthermore, given the rapid advancement in nanomedicine therapeutics, better outcomes for patients with AD can be anticipated. This article provides an overview of recent developments in nanomedicine using different types of nanoparticles for the management and treatment of AD.

1. Introduction

Today, the world’s foremost cause of death and infirmity is central nervous system; (CNS) disorders, mostly involving neurodegenerative diseases (ND), which cause great pain to patients and their relatives. NDs, including Alzheimer’s disease (AD), Parkinson’s disease (PD), epilepsy, dementia, Huntington’s disease, amyotrophic lateral sclerosis, multiple sclerosis (MS), brain stroke, and injuries, are the most common CNS diseases [1,2]. Gustavsson et al. (2022) projected that 416 million people globally fall into the AD continuum, which is significantly higher than the frequently reported estimate of 50 million individuals experiencing dementia [3]. Neurofibrillary tangles (NFT) and amyloid beta (Aβ) plaques are signs of the disease. Although AD has well-known neural characteristics, its symptoms are momentarily alleviated by the available treatments [4]. Many medicines licensed for the treatment of cognitive deficits are based on the manipulation of neurotransmitters or enzymes. However, difficulties related to poor drug solubility, low bioavailability, and inability to overcome barriers along the drug delivery route impede the development of new therapeutic options [5]. Therefore, treatment technologies must overcome the obstacles posed by the blood–brain barrier (BBB). Only a few molecules can pass through this intricate and tightly controlled barrier, which examines the biochemical, physicochemical, and structural characteristics of neighboring molecules at the perimeter. Numerous nanotechnology-based platforms have been developed to improve therapeutic efficacy in the brain [6,7]. These aided medication delivery techniques utilize advanced design principles and have several advantages over conventional techniques [8]. For example, formulations are highly customizable to increase drug loading, targeting, and release efficacy, and nanoparticles (NPs) are often low-cost technologies that can be employed for non-invasive administration [9]. These nanoscale structures make it easier for drugs to cross the BBB, enhancing their pharmacokinetics, pharmacodynamics, and bioavailability. Polymeric NPs, dendrimers, and lipid-based NPs are examples of such nanocarriers [10]. This article aimed to gather relevant information concerning the barriers in drug design for AD and the clinical use of NPs in the development of a new formulation for AD treatment. We searched the PubMed, Google Scholar, and Scopus databases for review articles, research articles, studies on nanomedicine, and clinical trials relevant to the fields of nanomedicine-based treatments against AD. Moreover, this review provides a brief overview of AD, barriers to developing therapeutic agents, the role of NPs, and their clinical aspects.

2. Alzheimer’s Disease

AD is a severe, fatal, progressive brain illness that impairs cognitive abilities [11]. The two most popular theories for the pathogenesis of AD are the excess and aggregation of Aβ peptide in the form of plaques and microtubule-associated tau protein becoming abnormally hyperphosphorylated and forms NFT that accumulates in the brain of AD; however, synaptic disturbance for calcium influx, autophagy, microgliosis–astrogliosis, and neuron demyelination are considered major factors in AD pathophysiology (Figure 1). Recent research has shown that chronic oxidative stress (OS), mitochondrial dysfunction, hormone imbalance, inflammation, genetic components, or BBB dysfunction may also play crucial roles in the pathogenesis of AD [12,13]. Food and Drug Administration-approved therapies for AD are currently available and are related primarily to two biochemical pathways involving the buildup of Aβ-peptide and NFT of p-tau protein. However, they have poor effectiveness in treating AD, demonstrating the necessity for other therapeutic strategies. Identifying additional molecular targets through discovering novel biomarkers may help the development of new treatments for AD [14].
The emergence of intracellular NFT and the production of extracellular Aβ plaques in the brain are key indicators of AD onset [15,16]. The histopathological characteristics are the loss of hippocampal neurons, synaptic degeneration, and aneuploidy. Additionally, early pathophysiological changes in AD have been linked to neuroinflammation, OS, mitochondrial dysfunction, and an injured brain lymphatic system [17,18].
AD is complicated by several variables, including genetic, environmental, health, and lifestyle factors. Obesity, hypertension, high cholesterol, and diabetes are only a few conditions that increase the risk of developing AD [19,20,21].
Researchers have studied many therapeutic targets. Among the therapeutic targets, common strategies include Aβ-based, tau protein-based, NFT-based, mitochondrial, OS, anti-inflammatory, and blood–brain receptor targeting (Table 1).

3. Challenges of Drug Designing for AD Treatment

The brain is regarded as the best-protected body organ. Several protective barriers surround the brain, including the skull, meninges, cerebrospinal fluid, and BBB. These elements contribute to the brain’s defense against external and internal harm as well as helping it stay healthy. However, in a diseased state, these protective barriers make it more difficult for therapeutic drugs to reach the brain [58]. The major barriers, including the BBB, blood–cerebrospinal fluid barrier (BCFB), and multidrug resistance proteins (MDRPs), are briefly discussed.

3.1. The Blood–Brain Barrier

The BBB is the principal barrier that prevents nanomedicines from entering the brain, limiting their ability to treat and diagnose AD. Therefore, nanomedicines should be intelligently developed to increase their capacity for brain accumulation [59,60]. The BBB, which prevents many medications from entering the brain at adequate concentrations, is the main obstacle in treating neurological illnesses. Moreover, unintended adverse effects resulting from drug release from the carrier before it reaches the brain further complicate treatment efforts. The BBB comprises endothelial cells, basal membranes, pericytes, and astrocytes (Figure 2). Tight junctions provide unique characteristics to the BBB. Consequently, the BBB prevents the entry of medications that are effective in treating a variety of neurological illnesses.
Brain capillaries appear to have essentially no intercellular interstitial gaps. As a result, transit likely involves all cells. Consequently, lipid-soluble compounds can pass freely through all endothelial membranes and easily cross the BBB [61]. Various different strategies are mentioned in recent studies, including direct injection, nose-to-brain route, the opening of BBB, inhibition of efflux transporters, and use of nanocarrier [62]; however, among these strategies, nanocarrier seems to be the most appropriate and result-oriented.
Using cutting-edge therapeutic nano-delivery devices may solve the problem of crossing the BBB [63,64,65,66,67]. Because of their distinctive physicochemical features and capacity to penetrate the BBB, engineered NPs < 100 nm have multiple applications in resolving these biological and pharmacological problems [68]. The ability of NP to cross the BBB increases the likelihood of early diagnosis and successful treatment of neurological diseases [69]. However, six different access routes that could be used to cross the BBB are paracellular diffusion (hydrophilic molecule), transcellular diffusion, carrier-mediated transport, receptor-mediated transport, adsorptive-mediated transport, and cell-mediated transport [62]. Paracellular transport is a passive transport process that results in the transport of substances across an epithelium. Tight junctions are the major rate-limiting pathway in the paracellular transport of large molecules across the epithelium [70]. Large molecules such as polypeptides are generally excluded from paracellular diffusion due to their hydrophilicity and high molecular mass. Adsorption-mediated transcytosis has attracted much attention because of its potential for large-molecule drug delivery to the brain. Cationic albumin-mediated brain DDS is commonly used; however, coupling this component with NP DDS is proven to be successful in cross-BBB for CNS delivery. Polymeric NPs are commonly used for adsorption-mediated transcytosis [71]. Immunocytes, neutrophils, and lymphocytes have a high degree of mobility, and they can cross various barriers to reach the target site and release their medication cargo. To enhance drug delivery, these cells can be loaded with nanocarriers such as polymeric NPs. In receptor-mediated DDS, transferrin receptors, low-density lipoprotein receptors, and insulin receptors are commonly used. A complex is formed between the desired drug and a receptor-targeting agent. This component is then linked or incorporated into the NPs [72]. Transcellular diffusion of molecules is based on their solubility, their molecular mass, and charge [73]. One important process for drug transportation over the BBB carrier-mediated transcytosis is used for the entrance of nutrition and energy intake into the brain cells. The benefits of carrier-mediated transport have been grafted onto nano DDSs using a technique that involves functionalizing the surface of the tiny molecules with certain ligands [74]. Among these six pathways, except the cell-mediated pathway, other five pathways are commonly used in nanomedicine delivery that cross the BBB for AD management, as shown in Figure 3.

3.2. The Blood–Cerebrospinal Fluid Barrier

The BCFB, which is constructed by epithelial cells of the choroid plexus and systematically deals with drug compounds, is the next barrier to overcome after the BBB. The choroid plexus epithelium (CPE) is one of the most effective tissue types and is based on secretory systems that balance cellular transport mechanisms. This epithelial barrier divides the blood and CSF in a manner similar to that of the BBB. Cells near the CSF-facing surface had an adjacent CPE that was tightly preserved. The CPE comprises detoxifying enzymes and multi-specific efflux transport proteins that work together to block the entry of potentially deadly substances into the CNS. Several medication delivery strategies have been developed; most are suited to the BBB and target the microvascular endothelium [75,76]. For the treatment of CNS disorders, targeting the BCFB, which is sculpted by the choroid epithelium, is crucial, in addition to targeting the BBB [77].
In order to enhance the administration of CNS therapies and offer brain protection measures, the BCFB, a less well-studied brain barrier location than the BBB, may serve as a possible therapeutic target. Therefore, by conducting a preliminary evaluation of the anticipated physiochemical behavior of a drug against this barrier, the use of reliable and accurate in vitro models of the BCFB can reduce the time and effort spent on pointless or repetitive research efforts. The development of innovative CNS treatments and pharmacologically active candidates that take advantage of the choroid plexus epithelial cells’ inherent transporting ability will be beneficial for developing CNS drug delivery strategies through the BCFB. There are still questions concerning the CP’s suitability for this purpose despite the fact that it has not received much attention as a prospective route for medication delivery to the brain. By using the current in vitro BCFB models, preliminary mechanistic and transport experiments can be used to assess the efficiency of the design process and the rate of successful transport across this barrier. Protein receptors, solute carriers, and amino acid transporters are three key groups of possible transporters that could be targeted to facilitate medication delivery to the CNS across the BCFB [78].

3.3. Multidrug Resistance Proteins

The BBB is formed by endothelial cells within brain capillaries and this cerebral endothelial cells contain diverse metabolic enzymes, including cytochrome P450 enzymes, alkaline phosphatases, and glutathione transferases, as well as energy-dependent efflux transport proteins, such as P-glycoprotein (P-gp), breast cancer resistance protein (BCRP; ABCG2), and MDRPs, which are arranged asymmetrically and act as barrier [79,80,81]. These MDR transporters, P-gp and BCRP, belong to the ATP-binding cassette (ABC) superfamily and prevent exposure of the brain to a large range of molecules [82]; this makes therapy unsuccessful by preventing some drugs from precisely achieving their intended therapeutic goals while also reducing drug buildup in the brain [83]. Among these transporters, MDRP is a main barrier for drug distribution to the brain due to its concentrative efflux activity. Changes to the expression levels of MDRP possibly alter drug concentrations in brain and spinal cord tissues. An alternative approach could be used for drug delivery by inhibiting the expression and function of P-gp [84]. Therefore, one of the current key issues in pharmaceutical research is the development of tools that allow for the efficient and effective delivery of medications into the CNS by the MDRP system [85]. Simultaneously, there is a need to gain a better understanding of ABC transporter activity at the BBB and BCFB that could correctly predict the drug pharmacokinetics and pharmacodynamics in the CNS.
The major feature of AD, a multifactorial ND that primarily affects older people, is a progressive decline in cognition, emotion, language, and memory. Pharmaceutical medication formulations and cholinesterase inhibitors have been widely promoted as AD treatments because of their beneficial effects. To enhance patients’ cognitive results, these drugs were eventually found to be unsuccessful in addressing the underlying causes of AD pathogenesis and instead focused only on symptoms [86,87].
Therefore, the market’s current drug delivery technologies cannot offer sufficient cytoarchitectural recovery and interconnections, which are essential for real recovery in AD. The concepts and methods of nanotechnology mentioned in this review can also substantially eradicate AD. However, nanotechnology can overcome these limitations by developing novel carrier-based platforms focusing on the targeted selective release of pharmacological payloads with on-demand and controlled release kinetics and improved reach by altering or evading the BBB. Nanomaterials have been studied for administering anti-AD drugs in experimental models of AD. This review covers several synergistic strategies in depth, providing details on the symptoms, risk factors, treatment options, and function of nanocomposites in slowing the progression of AD [88].

4. Nanomedicine

Maintaining the bioavailability, pharmacodynamics, and pharmacokinetics of medications is crucial to exerting their full therapeutic effects. Therefore, the goal of integrating a drug into or onto a polymeric and/or lipid NP is to substantially increase the drug’s therapeutic effect. The use of NPs in drug delivery is advantageous because they increase a drug’s bioavailability by enhancing its aqueous solubility and lengthening its half-life, which, in turn, slows the pace of drug clearance and delivers the medication to its intended site of action [89,90,91]. Furthermore, a wide range of NPs is used to treat AD [92]. Figure 4 shows the use of various NPs in the development of nanomedicines for the management and treatment of AD.
Nanoliposome carriers are the most promising drug delivery methods because of their biocompatibility and high flexibility in delivering a variety of therapeutic compounds across the BBB and specifically targeting brain cells. Evidence demonstrates that PSLs can cross the BBB to enter the brain, where they interact with several types of neuronal cell receptors [93].

4.1. Metallic/Inorganic Nanoparticles

Metallic NPs have been extensively used in recent years for the diagnosis, imaging, and treatment of NDs. Metallic NPs commonly contain gold NPs (AuNP), silver (Ag) NPs, iron oxide (FeO) NPs, platinum (Pt) NPs, and ceria (Ce2O3) NPs. These NPs play a significant role as DDSs in brain-targeted therapies. Metallic NPs can be easily fabricated and functionalized by adding different functional groups, and their shapes and sizes can be modified [94]. The conjugation of metallic NPs with drug molecules, antibodies, DNA/RNA, peptides, and aptamers has been extensively studied in recent years [95]. Among the metallic NPs, AuNPs have been widely used in diagnosis and imaging, such as in DDS.
Because of their biocompatibility, fascinating optical features, surface functionalization, and non-immunological qualities, AuNPs have gained significant interest as novel platforms for catalysis, drug transport, and disease diagnosis and treatment. AuNPs have also been developed to treat various CNS illnesses owing to their various dimensions, forms, and surface features [96]. Numerous researchers have used AuNPs to develop therapeutic compounds for the treatment of AD. According to Shao et al. (2023), AuNPs have been used to modify Aβ fibrillation associated with AD in intracellular and extracellular areas [97]. An aptamer-conjugated polydopamine-coated AuNP formulation was prepared to treat AD and tested on PC12 cells. The formulation inhibits the aggregation of the Aβ peptides and prevents Aβ-induced cell membrane damage [98]. In 2015, Gao et al. designed an AuNPs-based formulation where AuNPs were conjugated with polyoxometalate as an Aβ inhibitor for AD treatment. The formulation effectively inhibits Aβ aggregation, dissociates Aβ fibrils, and reduces peroxidase activity and cytotoxicity. This success is attributed to the advantageous use of AuNPs as BBB-crossing carriers [99]. In a recent study, AD model rats were treated with AuNPs conjugated with okadaic acid (OA). AuNPs were observed to maintain p-tau levels in the cortex and hippocampus; however, OA increased p-tau levels. Rats treated simultaneously with OA and AuNP showed high interleukin (IL)-1β and IL-4 levels in the hippocampus and cortex. AuNPs exhibit antioxidant properties and reduce OS in the brain [100]. Sanati et al. studied the impact of AuNPs on Aβ-induced AD in experimental rats, finding that AuNPs may enhance learning and memory for spatial information, and the expression level of stromal interaction molecules was increased, leading to an improvement in neural survival [101]. Studies have shown that AuNPs are multifunctional therapeutic agents for the treatment of AD.
Selenium nanoparticles (SeNPs) have received considerable attention because of their exceptionally low toxicity and antioxidative qualities. Selenium is a micronutrient crucial for preserving human health and lowering OS in the brain. Because of their outstanding neuroprotective properties, they have been implicated in the prevention of AD [102]. Selenium- and sodium selenite-rich NPs have antioxidant qualities and are utilized to treat brain illnesses; they also enhance cognitive function in a size-dependent manner [103]. Yin et al. conducted in vitro studies to examine the interactions between B6-SA-SeNPs and amyloid peptides as well as to assess the effectiveness of B6-SA-SeNPs when delivered to brain capillary endothelial cells. Because of their anti-amyloid, antioxidant, and high brain transport efficiency, the findings indicated that B6-SA-SeNPs could be a useful treatment for AD and a novel nanomedicine for disease modification in AD [102].
Neurotoxicity, DNA damage by inorganic NPs, and biochemical properties of the brain and iono-regulatory processes are the major challenges of this nanocarrier in AD therapeutics.

4.2. Carbon-Based Nanoparticles

A large category of carbon-based nanomaterials, known as “carbon dots” (CDs), are broadly divided into carbon nanodots, polymer dots, carbon nitride dots, and graphene quantum dots (QDs), which are used as DDSs for the treatment of NDs [104]. Top–down and bottom–up strategies were used to synthesize CDs, and a range of reaction conditions and precursors were used. The successful penetration of the BBB by a variety of CDs and CD ligand conjugates indicates encouraging advancement in the use of CD-based DDS for the treatment of CNS disorders [105].
The development of CDs has resulted in numerous advancements in recent years, including the delivery of medications that can traverse the BBB [106,107,108]. CDs are biocompatible and harmless because they do not contain metals. Owing to their abundance of surface functional groups, CDs can be conjugated to a variety of pharmacological compounds as nanocarriers through either covalent or noncovalent interactions. Most importantly, the combination of substantial surface diversity and small particle size simplifies the transfer of drugs across the BBB via active pathways. Additionally, the photoluminescence of CDs makes it simple to follow their in vivo movements in real time. With the help of these promising NPs, additional medications can now be transported to the brain, rendering the fight against AD and other NDs a new weapon [109]. One published study demonstrated the significance of yellow-emissive CD (Y-CDs) in AD treatment. According to these results, Y-CD crosses the BBB, enters cells, and inhibits the overexpression of human amyloid precursor protein and Aβ [106]. Another study also revealed the potential of CDs as a multifunctional β-sheet breaker and provided a promising anti-Aβ aggregation strategy for AD treatments [110]. In 2022, Yan et al. developed multifunctional CD photosensitizer nanoassemblies to inhibit amyloid aggregates that could easily cross the BBB and prevent microbial growth after treatment [111]. These recent studies demonstrate the potential significance of CD in CNS and its promising role as an inhibiting agent for the Aβ-related pathology of AD. Chung et al. fabricated targeting CDs that were red light-responsive and capable of suppressing Aβ aggregation and neurotoxicity in the brain of a patient with AD [112].
QDs are NPs with electronic and optical characteristics such as fluorescence and strong light emission. They possess distinct qualities, such as photostability, high quantum yield, high emission, and size tunability. The use of theranostic NPs in sensing, imaging, and medication delivery is currently receiving considerable attention. Additionally, QDs hold significant promise for the identification and management of several CNS disorders, including PD, AD, and MS. In several areas of life sciences, QDs are semiconductor nanocrystals with unique optical features, including high photochemical stability, extended wavelength, and long fluorescent half-life [113]. Some varieties of conjugated QDs are suitable carriers and are valuable for traversing the BBB in a variety of applications, including accurate imaging and detection of brain illnesses. The primary factor for transferring across the BBB is the particle surface, not the NPs’ size or chemo-electric charge [114]. One study synthesized QDs from the extract of the flower of Clitoria ternatea and observed that the learning and memory capacity of treated rats improved in the radial arm and Morris water maze assays. These results also confirmed that QDs significantly inhibited AChE, increased the levels of glutathione and proteins, and decreased the levels of lipid peroxide and nitric oxide [115]. In a study by Wang et al., a nanoformulation of graphene oxide (GO)-loaded dauricine was developed for the treatment of AD and was evaluated in an AD experimental model both in vivo and in vitro. The findings demonstrated that GO loaded with dauricine significantly decreased OS by increasing superoxide dismutase levels, lowering reactive oxygen species, and decreasing malondialdehyde levels in vitro. It also significantly improved cognitive memory impairment and brain glial cell activation in mice with Ab1-42-induced AD. Thus, GO-loaded dauricine has the potential to be a useful drug for the rapid treatment of AD. This study showed that GO loaded with dauricine protected against Ab1-42-induced oxidative damage and apoptosis in both AD models [116].
A recent study investigated the role of graphene oxide as a chelator to dissolve the amyloid-β plaques in AD using the density functional theory study. They have determined the chelating ability of various metals and GO by comparing the binding energies between metal—chelator and metal–Aβ complexes and conclude that GO can be used in future AD drug delivery therapy to target toxic metal–Aβ interactions and reduce Aβ aggregation [117].
GO may block the mTOR signaling pathway and initiate autophagy in microglia and neurons. When microglia and neurons were co-cultured, GO triggered autophagy in both cell types, particularly in the microglia, which aided in the removal of Aβ and ultimately had a protective impact on the neurons. Additionally, GO was able to decrease the toxicity of Aβ to neurons by its clearance in addition to being non-cytotoxic to microglia and neurons. These findings demonstrate the possibility of GO as an AD treatment [118].
Carbon nanotubes (CNTs) have garnered attention for their unique properties and biomedical potential. The ability of high drug loading, along with the capability to readily cross the BBB and also facilitate the passage of drugs to the brain via the olfactory route with ease, opens up new possibilities for targeted drug delivery and neuroprotection. In recent research, β-cyclodextrin modified CNTs nanocomposite was prepared for delivery of donepezil hydrochloride drug for the treatment of AD and found the significant sensitivity and limit of detection [119].
Since their unique characteristics, such as their ability to easily pass through cell membranes, thermal properties, large surface areas, and ease of molecular modification, carbon NPs stand out as highly innovative materials that could be used in novel therapeutic regimens against CNS. However, their usage in humans is also constrained, mostly due to the beginning of toxic phenomena that impact the nerve cells and the onset of inflammatory and oxidative processes. The focus of the new research must now be on the hunt for novel, more useful target molecules while also using appropriate engineering to structure nanomaterials that are better suited for human use [120].

4.3. Lipid-Based Nanocarriers

The abovementioned constraints are addressed using nanotechnology-based methods that utilize liposomes, micelles, dendrimers, and solid lipid nanoparticles (SLNPs) as DDSs. This study focused on medication delivery to treat NDs using an SLNP formulation [121]. Notably, SLNP can pass physiological barriers to increase bioavailability without using high-dosage forms, direct the active compound toward the target site with a significant reduction in toxicity to the adjacent tissues, and protect drugs from chemical and enzymatic degradation. We believe that SLNP may be an effective method for medications to cross the BBB and reach damaged parts of the CNS in patients with NDs such as AD and PD [122]. One study showed that nicotinamide-loaded functionalized SLNPs improved cognition in AD animal models by reducing the tau hyperphosphorylation [123]. Rivastigmine tartrate-loaded SLNPs have been formulated for enhanced intranasal delivery to the brain in the Alzheimer’s therapeutics [124]. In a therapeutic study on AD treatment, RVG29-functionalized SLNPs were prepared for the delivery of quercetin to the brain, resulting in a 1.5-fold increase in drug permeability to the experimental cell line [125].
Small vehicles (≤100 nm) are frequently preferred because only specific sizes can pass through the BBB in AD. However, studies have demonstrated that liposomes between 100 and 140 nm offer several benefits, including a longer half-life in blood circulation and a lack of plasma proteins [126]. In a recent study, transferrin-functionalized liposomes conjugated with vitamin B12 were used as therapeutic agents for AD. The formulation targeted the BBB and neuronal cells and delayed the formation of Aβ fibrils, showing great potential for AD therapeutics [127]. Caffeic acid-loaded transferrin-functionalized liposome NPs were developed capable of preventing AD by blocking Aβ aggregation and fibril formation and disaggregating mature fibrils [128].
Numerous studies have demonstrated the effectiveness of dendrimers as DDSs for the treatment of various diseases [129,130]. Since positively charged NPs, in conjunction with mucus, have greater cellular absorption, as described in a previous study [131], superficially positively charged dendrimers are thought to facilitate enhanced drug transport to the brain. A recent study revealed that polyamidoamine (PAMAM) dendrimers are suitable for the delivery of donepezil, an AChE inhibitor used to treat AD, have good in vivo pharmacokinetics and can cross the BBB [132]. Tacrine and PAMAM dendrimers co-delivered as therapeutic molecules for AD treatment were observed to help reduce the side effects of the drug in an animal model [133]. In the ND therapeutic study, carbamazepine was co-administered with PAMAM dendrimers in zebrafish larvae, resulting in reduced side effects from the carbamazepine [134].
Nanoliposomes are a feasible and promising DDS for AD that has not yet been tested in clinical trials. They are highly biocompatible and can carry various therapeutic molecules across the BBB and brain cells. They can be tailored to extend blood circulation time and directed against individual or multiple pathological targets.
In comparison to equivalent free drug solutions/suspensions, nano-lipid drug delivery systems have been shown to have positive properties such as prolonged drug release, increased CNS bioavailability, and further improved therapeutic efficacy [135].

4.4. Polymeric Nanoparticles

Polymeric NPs are solid objects composed of organic colloidal NPs with polymetric, natural, or artificial materials. Numerous monomers can be used to create polymeric NPs, which can then be polymerized using various methods, and their characteristics can be tailored for diverse applications. Synthetic, natural, and hybrid polymeric nanoparticle systems are most frequently used for brain targeting [136]. Chitosan and PLGA are the most common polymeric NPs used in AD treatment. Mathew et al. conducted an in vitro study of PLGA-coated curcumin with Tet-1 in AD treatment, finding that PLGA-coated curcumin NPs have the potential for AD treatment because of their anti-amyloid and antioxidant properties [137]. Another study showed that PLGA NPs loaded with Aβ generation inhibitor S1 and curcumin remarkably decreased the level of Aβ, enhanced the activities of antioxidant enzymes, and attenuated memory deficits and neuropathology in AD mice [138]. A recent study found that a formulation of PEGylated biodegradable poly (lactic-co-glycolic) nanospheres loaded with dexibuprofen administered to APPswe/PS1dE9 mice reduced memory impairment and decreased brain inflammation more efficiently than the free drug [139].
The size, shape, and chemical and physical properties of the NPs included in this review were not uniform. Each nanoparticle has unique properties with many advantages and disadvantages. The advantages and limitations of these NPs as potential DDSs have been discussed in many articles; however, scientists and researchers are working on most nanomaterials to study their uniformity, drug loading, drug release capacity, and safety as DDS for treating AD and other neurological diseases. Several in vitro and in vivo studies have demonstrated the significance of different types of NPs for treating AD. A few of these are discussed in Table 2, along with their results and possible prospects or limitations/advantages.
Clinical failures might be brought on by polymeric nanoparticles’ potential toxicity and sluggish degradability. The capacity of the polymeric NPs to traverse the BBB is constrained without surface changes. Although these NPs have a good rate of disintegration, the chronic nature of neurodegenerative illnesses and the requirement for repeated doses of the therapeutic substance could result in polymer accumulation that could have unwanted side effects [78,175].

5. Discussion

Mental and CNS illnesses have a significant prevalence worldwide, including neuroinflammation, brain tumors, and NDs. Neuronal loss is the main characteristic of NDs. AD and PD are the most common NDs. Although several medications are now approved for managing NDs, most treat only the related symptoms [176]. Because of the presence of the BBB, developing therapeutic agents for various CNS-related disorders is highly challenging [177].
The BBB serves as the primary barrier to drug delivery in AD treatment. The physical and biochemical barriers of the BBB limit the effects of all hydrophilic drugs; efflux pumps inside the BBB frequently transfer lipophilic drugs back into the blood [178]. As a result, it is critical to understand the structure and activity of the BBB to suggest alternative methods for delivering AD medications via nano-DDS. Utilizing the endothelial cell-binding affinity of lipid-soluble NPs can increase the rate at which a drug is transported via endocytosis or lipophilic transcellular routes. Moreover, the adsorptive properties of NPs can be useful because they can adhere to blood capillaries in the BBB, boosting the likelihood that the target medicine will cross this barrier [179,180]. Altering NPs with specific receptors, carrier proteins, and receptor-mediated transcytosis can enhance medication uptake through the BBB. Despite the ability of NPs to pierce the BBB, only approximately 5% of medications reach the brain, leaving the remaining 95% at an ineffective location [181]. This phenomenon may cause systemic side effects because the conventional route of drug administration is ineffective for accurately delivering the agent to the brain. The transport of therapeutic substances to the CNS via the olfactory and trigeminal nerves of the nasal cavity is facilitated by intranasal administration.
Furthermore, intranasal administration is risk-free and non-invasive. This medicine can prevent hepatic first-pass metabolism and drug degradation, increasing drug bioavailability [182]. Nanotechnology is a cutting-edge method that may open the door to new ways to overcome blood–CNS barriers, particularly the BBB. The therapeutic effects of drug delivery systems depend on their capacity to bypass the immune system, pass through the BBB, and locate themselves in the target tissues [1]. NPs are widely used in the diagnosis and treatment of AD [183]; however, for successful results, several aspects must be considered, including the target-specific distribution and metabolism of NPs and the interactions between target molecules (Aβ/Tau/NFT) and NPs. Nanomaterials have good in vitro inhibitory efficiency; however, the in vivo inhibition efficiency needs to be increased [184]. Several metallic NPs provide a good platform for efficient inhibition of the aggregation of Aβ either directly or after conjugation with a drug molecule [185].
Traditional DDSs are ineffective because they cannot pass through the BBB. Although innovation in research ensures the creation of nanotheranostics, a major issue is its potential for use in human therapies. The BBB can be crossed by a number of NPs [186]. Researchers are working on the development of nanomedicine using different types of nanocarriers. Table 2 summarizes some of the in/ex vivo studies. Nanoliposome carriers are the most promising drug delivery methods because of their biocompatibility and high flexibility in delivering a variety of therapeutic compounds across the BBB and specifically targeting brain cells. Evidence demonstrates that PSLs can cross the BBB to enter the brain, where they interact with several types of neuronal cell receptors [93].
It is possible to achieve widespread drug delivery to the entire brain by opening the BCFB instead of the BBB because the BCFB is relatively leaky compared to the BBB; water-soluble substances that do not cross the BBB do so on the BCSF barrier and enter CSF at a rate inversely related to molecular weight [187]. Kung Y et al. (2022) effectively validated a strategy for noninvasively and selectively opening the BCFB to promote medication administration into CSF circulation by facilitating drug delivery in the CNS by opening the BCFB barrier with a single low-energy shockwave pulse [188].
A promising prospect for the L-type amino acid transporter 1 (LAT1)-mediated brain delivery of CNS medicines is provided by the relatively high expression of the LAT1, which is primarily expressed in the cerebral cortex, blood–brain barrier, and blood–retina barrier. The transporter has proven to be effective in delivering clinically used CNS medications and prodrugs like L-Dopa. The bulk of the time, a parent medication has been conjugated to an amino acid side chain using a biodegradable linker without substituting carboxyl and amino groups in order to effectively bind LAT1 [189].
An advanced method for combating MDR in cancer treatment is small interfering RNA (siRNAs) delivered by nanocarriers. This method increases the effectiveness of already available medications by inhibiting the no-flux and efflux-related protein pumps that are engaged in or overexpressed in the MDR [190,191]. This type of nanocarrier-based siRNA technology may be helpful for treating CNS conditions. Moreover, the downregulation of the pump involved in drug resistance, combining monotherapy with P-gp inhibitors, and bypassing of pumps related to drug efflux are common nanotechnology methods that could be used to overcome the MDR problem in AD treatment.
The rapid development of nanotechnology has made many promising nanomaterials available for biomedical applications. These materials are widely used in the treatment of ND and appear to compensate for some of the shortcomings of stem cell therapy, including the transportation of stem cells, genes, and drugs, control of stem cell differentiation, and real-time monitoring. Therefore, stem cell therapy combined with nanotherapeutic techniques is a promising therapeutic method for treating NDs [192]. Because nanomedicine has been a research area for approximately 20 years, discussing what the field should do in the next 20 years is appropriate for creating a framework based on science for ongoing, sustained progress [193].
Nanomedication particles have good stability, are less toxic, and cause fewer side effects when used to diagnose and treat AD in elderly patients. These particles also help patients live more comfortably and have favorable clinical outcomes [142].
Therefore, future research on NP-based brain delivery should concentrate on enhancing safety, precision targetability, and pharmacokinetic characteristics. Human clinical trials should be conducted to assess the efficacy and safety of certain NPs and to determine the most promising and affordable AD treatments. To ensure the safe use of NPs by patients and healthcare personnel, thorough handling and administration standards must be devised. In addition, for the future treatment of AD, the theranostic development of NPs is essential, promising substantial advancements in precision and customized therapy. The clinical outcomes of each patient with AD who received one of these tailored medicines greatly improved. They strongly emphasize evaluating the efficacy and safety of the treatment processes. Thus, NP-DDSs offer strong therapeutic promise for AD. Further translational research should be conducted to examine the interaction of NPs in a biological interface in light of the benefits of crossing the BBB and therapeutic modalities, including symptomatic alleviation, DMT therapy, and hallmark diagnostics. Future alternatives for treating and diagnosing AD are promising when NPs are combined with recently discovered medicines [194].
However, the number of patents for nanotechnology-based goods is increasing. Clinical trials are required to assess the therapeutic efficacy and potential toxicological effects on human health. Neurologists and patients will soon benefit from appropriate nanotechnology-based DDSs that may improve therapeutic outcomes at lower costs. Although no clinical studies have been conducted on using nanotechnology to treat AD, this field of medicine is projected to change, bringing new ways to diagnose AD and the ability to individualize a patient’s therapy profile [195].

6. Current Research and Future Directions of Nanomedicine

AD is one of the most devastating diseases of the CNS. The primary pharmaceutical obstacle to the effective treatment of AD is BBB and BCFB. BBB-targeted nanomedicine has recently been developed and is now given more consideration in treating AD. Numerous nanoformulations, as indicated in Table 2, have proven to be highly effective in laboratory and clinical investigations. However, clinical translation from bench to bedside is not particularly successful because of an inadequate understanding of the mechanisms of action, the fate of nanocarriers inside the body, systemic toxicity, biocompatibility, aggregation, and the quick clearance resulting from their nanometer size. Comprehensive and in-depth toxicological research should be focused on brain-targeting nanoformulations. The therapeutic potential of nanomedicine will depend heavily on the rational approach and ongoing design of nanomaterials based on in-depth and extensive knowledge of data gathered from biological processes. Polymeric NPs have shown potential toxicity, drug loading into lipid NPs for hydrophilic compounds is weak, and the in vivo stability of liposomes is subpar. Nano-emulsions exhibit poor stability during storage, which causes phase separation and a quick release effect. Dendrimers have toxicity problems as well. To optimize all of these concerns and handle them in the near future, additional in-depth research investigations are, therefore, necessary.
The major challenges for the development of nanomedicine for AD are shown in Figure 5; however, some critical limitations/challenges are as follows:
  • Biodegradable nature of nanocarrier;
  • Different kinds of functional groups;
  • Different research study has different protocols and show different biodistribution of nanomedicine at different times;
  • The ability of nanomedicine to control its morphological as well as chemical properties in the bloodstream/stability in blood;
  • Nanomaterial does not show aggregation and is non-toxic, target-specific along with being pharmacodynamic and pharmacokinetic;
  • In vivo condition;
  • Reproducibility, predictability, accessibility, and cost-effectiveness;
  • Should cross BBB and another barrier of CNS.
There are a few important things to consider regarding the nanomaterials utilized in the formation of nanomedicine: they are environmentally friendly; cross the BBB; non-toxic by nature; effective; and safe. In addition, nanomedicine delivery must be simple and non-intrusive.

7. Conclusions

The tremendous efforts currently needed to treat AD revolve around delivering the correct pharmaceuticals into the right neurons and obtaining enough of the right treatments for the brain parenchyma. However, many unanswered questions and gaps remain regarding the use of NPs for the treatment of neurodisorders, even though many studies have concentrated on the application of NPs in DDDs to overcome the obstacles faced by traditional medicine. Nanocomposites, including metal/inorganic NPs, CDs, QDs, and lipid NPs, have been used to treat AD and other CNS disorders. However, the nanomaterial itself may be cytotoxic and facilitate the passage of neurotoxic compounds across the BBB. Some administration methods may be more invasive or even cause a breach of the BBB, favoring the entry of infections or toxic substances into the brain. Therefore, studying the limitations of these NPs is necessary.
The most recent developments in this promising subject discussed in this review article can offer systematic knowledge for designing new initiatives and improving the use of NPs to treat AD.

Author Contributions

N.P. and M.S. proposed the concept and content; N.P. drafted the manuscript; N.P. and D.Y. collected documents and drew pictures; N.P., D.Y. and M.S. reviewed and revised this manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Research Foundation (NRF) of Korea (NRF-2021R1I1A3055750) and the National Institute of Biological Resources (NIBR202325101).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Babazadeh, A.; Vahed, F.M.; Jafari, S.M. Nanocarrier-mediated brain delivery of bioactives for treatment/prevention of neurodegenerative diseases. J. Control Release 2020, 321, 211–221. [Google Scholar] [CrossRef] [PubMed]
  2. Stephenson, J.; Nutma, E.; van der Valk, P.; Amor, S. Inflammation in cns neurodegenerative diseases. Immunology 2018, 154, 204–219. [Google Scholar] [CrossRef] [PubMed]
  3. Gustavsson, A.; Norton, N.; Fast, T.; Frölich, L.; Georges, J.; Holzapfel, D.; Kirabali, T.; Krolak-Salmon, P.; Rossini, P.M.; Ferretti, M.T.; et al. Global estimates on the number of persons across the alzheimer’s disease continuum. Alzheimers Dement. 2023, 19, 658–670. [Google Scholar] [CrossRef] [PubMed]
  4. DeTure, M.A.; Dickson, D.W. The neuropathological diagnosis of alzheimer’s disease. Mol. Neurodegener. 2019, 14, 32. [Google Scholar] [CrossRef] [PubMed]
  5. Bhalani, D.V.; Nutan, B.; Kumar, A.; Singh Chandel, A.K. Bioavailability enhancement techniques for poorly aqueous soluble drugs and therapeutics. Biomedicines 2022, 10, 2055. [Google Scholar] [CrossRef]
  6. Khan, M.I.; Zahra, Q.u.A.; Batool, F.; Kalsoom, F.; Gao, S.; Ali, R.; Wang, W.; Kazmi, A.; Lianliang, L.; Wang, G.; et al. Trends in nanotechnology to improve therapeutic efficacy across special structures. OpenNano 2022, 7, 100049. [Google Scholar] [CrossRef]
  7. Chauhan, P.S.; Yadav, D.; Koul, B.; Mohanta, Y.K.; Jin, J.O. Recent advances in nanotechnology: A novel therapeutic system for the treatment of alzheimer’s disease. Curr. Drug Metab. 2020, 21, 1144–1151. [Google Scholar] [CrossRef]
  8. Mitchell, M.J.; Billingsley, M.M.; Haley, R.M.; Wechsler, M.E.; Peppas, N.A.; Langer, R. Engineering precision nanoparticles for drug delivery. Nat. Rev. Drug Discov. 2021, 20, 101–124. [Google Scholar] [CrossRef]
  9. Chu, K.S.; Schorzman, A.N.; Finniss, M.C.; Bowerman, C.J.; Peng, L.; Luft, J.C.; Madden, A.J.; Wang, A.Z.; Zamboni, W.C.; DeSimone, J.M. Nanoparticle drug loading as a design parameter to improve docetaxel pharmacokinetics and efficacy. Biomaterials 2013, 34, 8424–8429. [Google Scholar] [CrossRef]
  10. Wechsler, M.E.; Stephenson, R.E.; Murphy, A.C.; Oldenkamp, H.F.; Singh, A.; Peppas, N.A. Engineered microscale hydrogels for drug delivery, cell therapy, and sequencing. Biomed. Microdevices 2019, 21, 31. [Google Scholar] [CrossRef]
  11. Wilson, B.; Geetha, K.M. Neurotherapeutic applications of nanomedicine for treating alzheimer’s disease. J. Control Release 2020, 325, 25–37. [Google Scholar] [CrossRef] [PubMed]
  12. Ettcheto, M.; Cano, A.; Busquets, O.; Manzine, P.R.; Sanchez-Lopez, E.; Castro-Torres, R.D.; Beas-Zarate, C.; Verdaguer, E.; García, M.L.; Olloquequi, J. A metabolic perspective of late onset alzheimer’s disease. Pharmacol. Res. 2019, 145, 104255. [Google Scholar] [CrossRef]
  13. Nation, D.A.; Sweeney, M.D.; Montagne, A.; Sagare, A.P.; D’Orazio, L.M.; Pachicano, M.; Sepehrband, F.; Nelson, A.R.; Buennagel, D.P.; Harrington, M.G. Blood–brain barrier breakdown is an early biomarker of human cognitive dysfunction. Nat. Med. 2019, 25, 270–276. [Google Scholar] [CrossRef] [PubMed]
  14. Cano, A.; Turowski, P.; Ettcheto, M.; Duskey, J.T.; Tosi, G.; Sánchez-López, E.; García, M.L.; Camins, A.; Souto, E.B.; Ruiz, A.; et al. Nanomedicine-based technologies and novel biomarkers for the diagnosis and treatment of alzheimer’s disease: From current to future challenges. J. Nanobiotechnol. 2021, 19, 122. [Google Scholar] [CrossRef] [PubMed]
  15. Murphy, M.P.; LeVine III, H. Alzheimer’s disease and the amyloid-β peptide. J. Alzheimers Dis. 2010, 19, 311–323. [Google Scholar] [CrossRef]
  16. Takahashi, R.H.; Nagao, T.; Gouras, G.K. Plaque formation and the intraneuronal accumulation of β-amyloid in alzheimer’s disease. Pathol. Int. 2017, 67, 185–193. [Google Scholar] [CrossRef]
  17. Khan, N.H.; Mir, M.; Ngowi, E.E.; Zafar, U.; Khakwani, M.; Khattak, S.; Zhai, Y.K.; Jiang, E.S.; Zheng, M.; Duan, S.F.; et al. Nanomedicine: A promising way to manage alzheimer’s disease. Front. Bioeng. Biotechnol. 2021, 9, 630055. [Google Scholar] [CrossRef]
  18. Slater, C.; Wang, Q. Alzheimer’s disease: An evolving understanding of noradrenergic involvement and the promising future of electroceutical therapies. Clin. Transl. Med. 2021, 11, e397. [Google Scholar] [CrossRef]
  19. Selman, A.; Burns, S.; Reddy, A.P.; Culberson, J.; Reddy, P.H. The role of obesity and diabetes in dementia. Int. J. Mol. Sci. 2022, 23, 9267. [Google Scholar] [CrossRef]
  20. Ezkurdia, A.; Ramírez, M.J.; Solas, M. Metabolic syndrome as a risk factor for alzheimer’s disease: A focus on insulin resistance. Int. J. Mol. Sci. 2023, 24, 4354. [Google Scholar] [CrossRef]
  21. Fan, Y.-C.; Hsu, J.-L.; Tung, H.-Y.; Chou, C.-C.; Bai, C.-H. Increased dementia risk predominantly in diabetes mellitus rather than in hypertension or hyperlipidemia: A population-based cohort study. Alzheimers Res. Ther. 2017, 9, 7. [Google Scholar] [CrossRef] [PubMed]
  22. Panza, F.; Lozupone, M.; Logroscino, G.; Imbimbo, B.P. A critical appraisal of amyloid-β-targeting therapies for alzheimer disease. Nat. Rev. Neurol. 2019, 15, 73–88. [Google Scholar] [CrossRef] [PubMed]
  23. Pinheiro, L.; Faustino, C. Therapeutic strategies targeting amyloid-β in alzheimer’s disease. Curr. Alzheimer Res. 2019, 16, 418–452. [Google Scholar] [CrossRef] [PubMed]
  24. Liu, S.; Park, S.; Allington, G.; Prelli, F.; Sun, Y.; Martá-Ariza, M.; Scholtzova, H.; Biswas, G.; Brown, B.; Verghese, P.B. Targeting apolipoprotein e/amyloid β binding by peptoid cpo_aβ17-21 p ameliorates alzheimer’s disease related pathology and cognitive decline. Sci. Rep. 2017, 7, 8009. [Google Scholar] [CrossRef]
  25. Xiao, X.; Jiao, B.; Liao, X.; Zhang, W.; Yuan, Z.; Guo, L.; Wang, X.; Zhou, L.; Liu, X.; Yan, X. Association of genes involved in the metabolic pathways of amyloid-β and tau proteins with sporadic late-onset alzheimer’s disease in the southern han chinese population. Front. Aging Neurosci. 2020, 12, 584801. [Google Scholar] [CrossRef]
  26. Gauthier, S.; Boxer, A.; Knopman, D.; Sims, J.; Doody, R.; Aisen, P.; Iwatsubo, T.; Bateman, R.; Vellas, B. Therapeutic targets for alzheimer’s disease: Amyloid vs. Non-amyloid. Where does consensus lie today? An ctad task force report. J. Prev. Alzheimers Dis. 2022, 9, 231–235. [Google Scholar] [CrossRef]
  27. Novak, P.; Kovacech, B.; Katina, S.; Schmidt, R.; Scheltens, P.; Kontsekova, E.; Ropele, S.; Fialova, L.; Kramberger, M.; Paulenka-Ivanovova, N. Adamant: A placebo-controlled randomized phase 2 study of aadvac1, an active immunotherapy against pathological tau in alzheimer’s disease. Nat. Aging 2021, 1, 521–534. [Google Scholar] [CrossRef]
  28. Coman, H.; Nemeş, B. New therapeutic targets in alzheimer’s disease. Int. J. Gerontol. 2017, 11, 2–6. [Google Scholar] [CrossRef]
  29. Congdon, E.E.; Sigurdsson, E.M. Tau-targeting therapies for alzheimer disease. Nat. Rev. Neurol. 2018, 14, 399–415. [Google Scholar] [CrossRef]
  30. Wang, J.; Chen, G.J. Mitochondria as a therapeutic target in alzheimer’s disease. Genes Dis. 2016, 3, 220–227. [Google Scholar] [CrossRef]
  31. Mary, A.; Eysert, F.; Checler, F.; Chami, M. Mitophagy in alzheimer’s disease: Molecular defects and therapeutic approaches. Mol. Psychiatry 2023, 28, 202–216. [Google Scholar] [CrossRef] [PubMed]
  32. Oliver, D.; Reddy, P.H. Dynamics of dynamin-related protein 1 in alzheimer’s disease and other neurodegenerative diseases. Cells 2019, 8, 961. [Google Scholar] [CrossRef]
  33. Cenini, G.; Voos, W. Mitochondria as potential targets in alzheimer disease therapy: An update. Front. Pharmacol. 2019, 10, 902. [Google Scholar] [CrossRef] [PubMed]
  34. Pritam, P.; Deka, R.; Bhardwaj, A.; Srivastava, R.; Kumar, D.; Jha, A.K.; Jha, N.K.; Villa, C.; Jha, S.K. Antioxidants in alzheimer’s disease: Current therapeutic significance and future prospects. Biology 2022, 11, 212. [Google Scholar] [CrossRef] [PubMed]
  35. Teixeira, J.P.; de Castro, A.A.; Soares, F.V.; da Cunha, E.F.F.; Ramalho, T.C. Future therapeutic perspectives into the alzheimer’s disease targeting the oxidative stress hypothesis. Molecules 2019, 24, 4410. [Google Scholar] [CrossRef]
  36. Cassidy, L.; Fernandez, F.; Johnson, J.B.; Naiker, M.; Owoola, A.G.; Broszczak, D.A. Oxidative stress in alzheimer’s disease: A review on emergent natural polyphenolic therapeutics. Complement. Ther. Med. 2020, 49, 102294. [Google Scholar] [CrossRef]
  37. Galasko, D.R.; Peskind, E.; Clark, C.M.; Quinn, J.F.; Ringman, J.M.; Jicha, G.A.; Cotman, C.; Cottrell, B.; Montine, T.J.; Thomas, R.G. Antioxidants for alzheimer disease: A randomized clinical trial with cerebrospinal fluid biomarker measures. Arch. Neurol. 2012, 69, 836–841. [Google Scholar] [CrossRef]
  38. Cenini, G.; Rüb, C.; Bruderek, M.; Voos, W. Amyloid β-peptides interfere with mitochondrial preprotein import competence by a coaggregation process. Mol. Biol. Cell 2016, 27, 3257–3272. [Google Scholar] [CrossRef]
  39. Si, Z.-Z.; Zou, C.-J.; Mei, X.; Li, X.-F.; Luo, H.; Shen, Y.; Hu, J.; Li, X.-X.; Wu, L.; Liu, Y. Targeting neuroinflammation in alzheimer’s disease: From mechanisms to clinical applications. Neural Regen. Res. 2023, 18, 708–715. [Google Scholar]
  40. Li, J.J.; Wang, B.; Kodali, M.C.; Chen, C.; Kim, E.; Patters, B.J.; Lan, L.; Kumar, S.; Wang, X.; Yue, J.; et al. In vivo evidence for the contribution of peripheral circulating inflammatory exosomes to neuroinflammation. J. Neuroinflammation 2018, 15, 8. [Google Scholar] [CrossRef]
  41. Bronzuoli, M.R.; Facchinetti, R.; Valenza, M.; Cassano, T.; Steardo, L.; Scuderi, C. Astrocyte function is affected by aging and not alzheimer’s disease: A preliminary investigation in hippocampi of 3xtg-ad mice. Front. Pharmacol. 2019, 10, 644. [Google Scholar] [CrossRef] [PubMed]
  42. Kuber, B.; Fadnavis, M.; Chatterjee, B. Role of angiotensin receptor blockers in the context of alzheimer’s disease. Fundam. Clin. Pharmacol. 2023, 37, 429–445. [Google Scholar] [CrossRef] [PubMed]
  43. Royea, J.; Hamel, E. Brain angiotensin ii and angiotensin iv receptors as potential alzheimer’s disease therapeutic targets. GeroScience 2020, 42, 1237–1256. [Google Scholar] [CrossRef] [PubMed]
  44. Gebre, A.K.; Altaye, B.M.; Atey, T.M.; Tuem, K.B.; Berhe, D.F. Targeting renin-angiotensin system against alzheimer’s disease. Front. Pharmacol. 2018, 9, 440. [Google Scholar] [CrossRef] [PubMed]
  45. Wharton, W.; Goldstein, F.C.; Zhao, L.; Steenland, K.; Levey, A.I.; Hajjar, I. Modulation of renin-angiotensin system may slow conversion from mild cognitive impairment to alzheimer’s disease. J. Am. Geriatr. Soc. 2015, 63, 1749–1756. [Google Scholar] [CrossRef]
  46. Mogi, M.; Iwanami, J.; Horiuchi, M. Roles of brain angiotensin ii in cognitive function and dementia. Int. J. Hypertens. 2012, 2012, 169649. [Google Scholar] [CrossRef]
  47. Soto, M.E.; Abellan van Kan, G.; Nourhashemi, F.; Gillette-Guyonnet, S.; Cesari, M.; Cantet, C.; Rolland, Y.; Vellas, B. Angiotensin-converting enzyme inhibitors and alzheimer’s disease progression in older adults: Results from the réseau sur la maladie d’alzheimer français cohort. J. Am. Geriatr. Soc. 2013, 61, 1482–1488. [Google Scholar] [CrossRef]
  48. Menting, K.W.; Claassen, J.A. Β-secretase inhibitor; a promising novel therapeutic drug in alzheimer’s disease. Front. Aging Neurosci. 2014, 6, 165. [Google Scholar] [CrossRef]
  49. De Strooper, B.; Vassar, R.; Golde, T. The secretases: Enzymes with therapeutic potential in alzheimer disease. Nat. Rev. Neurol. 2010, 6, 99–107. [Google Scholar] [CrossRef]
  50. MacLeod, R.; Hillert, E.-K.; Cameron, R.T.; Baillie, G.S. The role and therapeutic targeting of α-, β-and γ-secretase in alzheimer’s disease. Future Sci. OA 2015, 1. [Google Scholar] [CrossRef]
  51. Maia, M.A.; Sousa, E. Bace-1 and γ-secretase as therapeutic targets for alzheimer’s disease. Pharmaceuticals 2019, 12, 41. [Google Scholar] [CrossRef] [PubMed]
  52. Kurz, C.; Walker, L.; Rauchmann, B.S.; Perneczky, R. Dysfunction of the blood–brain barrier in alzheimer’s disease: Evidence from human studies. Neuropathol. Appl. Neurobiol. 2022, 48, e12782. [Google Scholar] [CrossRef] [PubMed]
  53. Pardridge, W.M. Blood-brain barrier and delivery of protein and gene therapeutics to brain. Front. Aging Neurosci. 2020, 11, 373. [Google Scholar] [CrossRef] [PubMed]
  54. Rofo, F.; Metzendorf, N.G.; Saubi, C.; Suominen, L.; Godec, A.; Sehlin, D.; Syvänen, S.; Hultqvist, G. Blood–brain barrier penetrating neprilysin degrades monomeric amyloid-beta in a mouse model of alzheimer’s disease. Alzheimers Res. Ther. 2022, 14, 180. [Google Scholar] [CrossRef]
  55. Mufson, E.J.; Counts, S.E.; Perez, S.E.; Ginsberg, S.D. Cholinergic system during the progression of alzheimer’s disease: Therapeutic implications. Expert Rev. Neurother. 2008, 8, 1703–1718. [Google Scholar] [CrossRef]
  56. H Ferreira-Vieira, T.; M Guimaraes, I.; R Silva, F.; M Ribeiro, F. Alzheimer’s disease: Targeting the cholinergic system. Curr. Neuropharmacol. 2016, 14, 101–115. [Google Scholar] [CrossRef]
  57. Francis, P.T.; Palmer, A.M.; Snape, M.; Wilcock, G.K. The cholinergic hypothesis of alzheimer’s disease: A review of progress. J. Neurol. Neurosurg. Psychiatry 1999, 66, 137–147. [Google Scholar] [CrossRef]
  58. Hanif, S.; Muhammad, P.; Chesworth, R.; Rehman, F.U.; Qian, R.-j.; Zheng, M.; Shi, B.-y. Nanomedicine-based immunotherapy for central nervous system disorders. Acta Pharmacol. Sin. 2020, 41, 936–953. [Google Scholar] [CrossRef]
  59. Zhang, L.; Sun, H.; Zhao, J.; Lee, J.; Low, L.E.; Gong, L.; Chen, Y.; Wang, N.; Zhu, C.; Lin, P. Dynamic nanoassemblies for imaging and therapy of neurological disorders. Adv. Drug Deliv. Rev. 2021, 175, 113832. [Google Scholar] [CrossRef]
  60. Tian, X.; Fan, T.; Zhao, W.; Abbas, G.; Han, B.; Zhang, K.; Li, N.; Liu, N.; Liang, W.; Huang, H.; et al. Recent advances in the development of nanomedicines for the treatment of ischemic stroke. Bioact. Mater. 2021, 6, 2854–2869. [Google Scholar] [CrossRef]
  61. Mittal, K.R.; Pharasi, N.; Sarna, B.; Singh, M.; Haider, S.; Singh, S.K.; Dua, K.; Jha, S.K.; Dey, A.; Ojha, S. Nanotechnology-based drug delivery for the treatment of cns disorders. Transl. Neurosci. 2022, 13, 527–546. [Google Scholar] [CrossRef] [PubMed]
  62. Sánchez-Dengra, B.; González-Álvarez, I.; Bermejo, M.; González-Álvarez, M. Access to the cns: Strategies to overcome the bbb. Int. J. Pharm. 2023, 636, 122759. [Google Scholar] [CrossRef] [PubMed]
  63. Jagaran, K.; Singh, M. Nanomedicine for neurodegenerative disorders: Focus on alzheimer’s and parkinson’s diseases. Int. J. Mol. Sci. 2021, 22, 9082. [Google Scholar] [CrossRef] [PubMed]
  64. Hwang, J.; Zhang, W.; Park, H.-B.; Yadav, D.; Jeon, Y.H.; Jin, J.-O. Escherichia coli adhesin protein-conjugated thermal responsive hybrid nanoparticles for photothermal and immunotherapy against cancer and its metastasis. J. Immunother. Cancer 2021, 9, e002666. [Google Scholar] [CrossRef] [PubMed]
  65. Yadav, D.; Kwak, M.; Chauhan, P.S.; Puranik, N.; Lee, P.C.; Jin, J.-O. Cancer Immunotherapy by Immune Checkpoint Blockade and Its Advanced Application Using Bio-Nanomaterials; Seminars in Cancer Biology; Elsevier: Amsterdam, The Netherlands, 2022. [Google Scholar]
  66. Koul, B.; Poonia, A.K.; Yadav, D.; Jin, J.O. Microbe-mediated biosynthesis of nanoparticles: Applications and future prospects. Biomolecules 2021, 11, 886. [Google Scholar] [CrossRef] [PubMed]
  67. Kim, S.-J.; Puranik, N.; Yadav, D.; Jin, J.-O.; Lee, P.C. Lipid nanocarrier-based drug delivery systems: Therapeutic advances in the treatment of lung cancer. Int. J. Nanomed. 2023, 2023, 2659–2676. [Google Scholar] [CrossRef]
  68. Mukherjee, S.; Madamsetty, V.S.; Bhattacharya, D.; Roy Chowdhury, S.; Paul, M.K.; Mukherjee, A. Recent advancements of nanomedicine in neurodegenerative disorders theranostics. Adv. Funct. Mater. 2020, 30, 2003054. [Google Scholar] [CrossRef]
  69. Ghalamfarsa, G.; Hojjat-Farsangi, M.; Mohammadnia-Afrouzi, M.; Anvari, E.; Farhadi, S.; Yousefi, M.; Jadidi-Niaragh, F. Application of nanomedicine for crossing the blood–brain barrier: Theranostic opportunities in multiple sclerosis. J. Immunotoxicol. 2016, 13, 603–619. [Google Scholar] [CrossRef]
  70. Maiti, S. Nanometric biopolymer devices for oral delivery of macromolecules with clinical significance. In Multifunctional Systems for Combined Delivery, Biosensing and Diagnostics; Elsevier: Amsterdam, The Netherlands, 2017; pp. 109–138. [Google Scholar]
  71. Zhu, X.; Jin, K.; Huang, Y.; Pang, Z. Brain drug delivery by adsorption-mediated transcytosis. In Brain Targeted Drug Delivery System; Elsevier: Amsterdam, The Netherlands, 2019; pp. 159–183. [Google Scholar]
  72. Pulgar, V.M. Transcytosis to cross the blood brain barrier, new advancements and challenges. Front. Neurosci. 2019, 12, 1019. [Google Scholar] [CrossRef]
  73. Khan, N.U.; Miao, T.; Ju, X.; Guo, Q.; Han, L. Carrier-mediated transportation through bbb. In Brain Targeted Drug Delivery System; Elsevier: Amsterdam, The Netherlands, 2019; pp. 129–158. [Google Scholar]
  74. Hervé, F.; Ghinea, N.; Scherrmann, J.-M. Cns delivery via adsorptive transcytosis. AAPS J. 2008, 10, 455–472. [Google Scholar] [CrossRef]
  75. Hersh, D.S.; Wadajkar, A.S.; Roberts, N.; Perez, J.G.; Connolly, N.P.; Frenkel, V.; Winkles, J.A.; Woodworth, G.F.; Kim, A.J. Evolving drug delivery strategies to overcome the blood brain barrier. Curr. Pharm. Des. 2016, 22, 1177–1193. [Google Scholar] [CrossRef] [PubMed]
  76. Pawar, B.; Vasdev, N.; Gupta, T.; Mhatre, M.; More, A.; Anup, N.; Tekade, R.K. Current update on transcellular brain drug delivery. Pharmaceutics 2022, 14, 2719. [Google Scholar] [CrossRef] [PubMed]
  77. Guo, Z.-H.; Khattak, S.; Rauf, M.A.; Ansari, M.A.; Alomary, M.N.; Razak, S.; Yang, C.-Y.; Wu, D.-D.; Ji, X.-Y. Role of nanomedicine-based therapeutics in the treatment of cns disorders. Molecules 2023, 28, 1283. [Google Scholar] [CrossRef] [PubMed]
  78. Dabbagh, F.; Schroten, H.; Schwerk, C. In vitro models of the blood-cerebrospinal fluid barrier and their applications in the development and research of (neuro)pharmaceuticals. Pharmaceutics 2022, 14, 1729. [Google Scholar] [CrossRef] [PubMed]
  79. Meyer, J.; Mischeck, U.; Veyhl, M.; Henzel, K.; Galla, H.J. Blood-brain barrier characteristic enzymatic properties in cultured brain capillary endothelial cells. Brain Res. 1990, 514, 305–309. [Google Scholar] [CrossRef]
  80. Macdonald, J.A.; Murugesan, N.; Pachter, J.S. Endothelial cell heterogeneity of blood-brain barrier gene expression along the cerebral microvasculature. J. Neurosci. Res. 2010, 88, 1457–1474. [Google Scholar] [CrossRef]
  81. Vašková, J.; Kočan, L.; Vaško, L.; Perjési, P. Glutathione-related enzymes and proteins: A review. Molecules 2023, 28, 1447. [Google Scholar] [CrossRef]
  82. Lye, P.; Bloise, E.; Matthews, S.G. Effects of bacterial and viral pathogen-associated molecular patterns (pamps) on multidrug resistance (mdr) transporters in brain endothelial cells of the developing human blood–brain barrier. Fluids Barriers CNS 2023, 20, 8. [Google Scholar] [CrossRef]
  83. Yalamarty, S.S.K.; Filipczak, N.; Li, X.; Subhan, M.A.; Parveen, F.; Ataide, J.A.; Rajmalani, B.A.; Torchilin, V.P. Mechanisms of resistance and current treatment options for glioblastoma multiforme (gbm). Cancers 2023, 15, 2116. [Google Scholar] [CrossRef]
  84. Aryal, M.; Fischer, K.; Gentile, C.; Gitto, S.; Zhang, Y.Z.; McDannold, N. Effects on p-glycoprotein expression after blood-brain barrier disruption using focused ultrasound and microbubbles. PLoS ONE 2017, 12, e0166061. [Google Scholar] [CrossRef]
  85. Asil, S.M.; Ahlawat, J.; Barroso, G.G.; Narayan, M. Nanomaterial based drug delivery systems for the treatment of neurodegenerative diseases. Biomater. Sci. 2020, 8, 4109–4128. [Google Scholar] [CrossRef]
  86. Thoe, E.S.; Fauzi, A.; Tang, Y.Q.; Chamyuang, S.; Chia, A.Y.Y. A review on advances of treatment modalities for alzheimer’s disease. Life Sci. 2021, 276, 119129. [Google Scholar] [CrossRef] [PubMed]
  87. Moss, D.E.; Perez, R.G.; Kobayashi, H. Cholinesterase inhibitor therapy in alzheimer’s disease: The limits and tolerability of irreversible cns-selective acetylcholinesterase inhibition in primates. J. Alzheimers Dis. 2017, 55, 1285–1294. [Google Scholar] [CrossRef] [PubMed]
  88. Pathak, M.; Singhal, R. Therapeutic and diagnostic applications of nanocomposites in the treatment alzheimer’s disease studies. Biointerface Res. Appl. Chem. 2022, 12, 940–960. [Google Scholar]
  89. Yadav, D.; Wairagu, P.M.; Kwak, M.; Jin, J.O. Nanoparticle-based inhalation therapy for pulmonary diseases. Curr. Drug Metab. 2022, 23, 882–896. [Google Scholar] [CrossRef] [PubMed]
  90. Joseph, T.M.; Kar Mahapatra, D.; Esmaeili, A.; Piszczyk, Ł.; Hasanin, M.S.; Kattali, M.; Haponiuk, J.; Thomas, S. Nanoparticles: Taking a unique position in medicine. Nanomaterials 2023, 13, 574. [Google Scholar] [CrossRef]
  91. Tenchov, R.; Bird, R.; Curtze, A.E.; Zhou, Q. Lipid nanoparticles─from liposomes to mrna vaccine delivery, a landscape of research diversity and advancement. ACS Nano 2021, 15, 16982–17015. [Google Scholar] [CrossRef]
  92. Feng, L.; Wang, H.; Xue, X. Recent progress of nanomedicine in the treatment of central nervous system diseases. Adv. Ther. 2020, 3, 1900159. [Google Scholar] [CrossRef]
  93. Saffari, P.M.; Alijanpour, S.; Takzaree, N.; Sahebgharani, M.; Etemad-Moghadam, S.; Noorbakhsh, F.; Partoazar, A. Metformin loaded phosphatidylserine nanoliposomes improve memory deficit and reduce neuroinflammation in streptozotocin-induced alzheimer’s disease model. Life Sci. 2020, 255, 117861. [Google Scholar] [CrossRef]
  94. El-Seedi, H.R.; El-Shabasy, R.M.; Khalifa, S.A.; Saeed, A.; Shah, A.; Shah, R.; Iftikhar, F.J.; Abdel-Daim, M.M.; Omri, A.; Hajrahand, N.H. Metal nanoparticles fabricated by green chemistry using natural extracts: Biosynthesis, mechanisms, and applications. RSC Adv. 2019, 9, 24539–24559. [Google Scholar] [CrossRef]
  95. Zhang, W.; Taheri-Ledari, R.; Ganjali, F.; Afruzi, F.H.; Hajizadeh, Z.; Saeidirad, M.; Qazi, F.S.; Kashtiaray, A.; Sehat, S.S.; Hamblin, M.R.; et al. Nanoscale bioconjugates: A review of the structural attributes of drug-loaded nanocarrier conjugates for selective cancer therapy. Heliyon 2022, 8, e09577. [Google Scholar] [CrossRef] [PubMed]
  96. Yeh, Y.C.; Creran, B.; Rotello, V.M. Gold nanoparticles: Preparation, properties, and applications in bionanotechnology. Nanoscale 2012, 4, 1871–1880. [Google Scholar] [CrossRef] [PubMed]
  97. Shao, X.; Yan, C.; Wang, C.; Wang, C.; Cao, Y.; Zhou, Y.; Guan, P.; Hu, X.; Zhu, W.; Ding, S. Advanced nanomaterials for modulating alzheimer’s related amyloid aggregation. Nanoscale Adv. 2023, 5, 46–80. [Google Scholar] [CrossRef] [PubMed]
  98. Qin, J.; Guan, Y.; Li, Z.; Guo, X.; Zhang, M.; Wang, D.; Tang, J. Aptamer conjugated polydopamine-coated gold nanoparticles as a dual-action nanoplatform targeting β-amyloid peptide for alzheimer’s disease therapy. J. Mater. Chem. B Mater. Biol. Med. 2022, 10, 8525–8534. [Google Scholar] [CrossRef] [PubMed]
  99. Gao, N.; Sun, H.; Dong, K.; Ren, J.; Qu, X. Gold-nanoparticle-based multifunctional amyloid-β inhibitor against alzheimer’s disease. Chemistry 2015, 21, 829–835. [Google Scholar] [CrossRef]
  100. Dos Santos Tramontin, N.; da Silva, S.; Arruda, R.; Ugioni, K.S.; Canteiro, P.B.; de Bem Silveira, G.; Mendes, C.; Silveira, P.C.L.; Muller, A.P. Gold nanoparticles treatment reverses brain damage in alzheimer’s disease model. Mol. Neurobiol. 2020, 57, 926–936. [Google Scholar] [CrossRef]
  101. Sanati, M.; Aminyavari, S.; Khodagholi, F.; Hajipour, M.J.; Sadeghi, P.; Noruzi, M.; Moshtagh, A.; Behmadi, H.; Sharifzadeh, M. Pegylated superparamagnetic iron oxide nanoparticles (spions) ameliorate learning and memory deficit in a rat model of alzheimer’s disease: Potential participation of stims. NeuroToxicology 2021, 85, 145–159. [Google Scholar] [CrossRef]
  102. Yin, T.; Yang, L.; Liu, Y.; Zhou, X.; Sun, J.; Liu, J. Sialic acid (sa)-modified selenium nanoparticles coated with a high blood–brain barrier permeability peptide-b6 peptide for potential use in alzheimer’s disease. Acta Biomater. 2015, 25, 172–183. [Google Scholar] [CrossRef]
  103. Nazıroğlu, M.; Muhamad, S.; Pecze, L. Nanoparticles as potential clinical therapeutic agents in alzheimer’s disease: Focus on selenium nanoparticles. Expert Rev. Clin. Pharmacol. 2017, 10, 773–782. [Google Scholar] [CrossRef]
  104. Liu, J.; Li, R.; Yang, B. Carbon dots: A new type of carbon-based nanomaterial with wide applications. ACS Cent. Sci. 2020, 6, 2179–2195. [Google Scholar] [CrossRef]
  105. Zhang, W.; Sigdel, G.; Mintz, K.J.; Seven, E.S.; Zhou, Y.; Wang, C.; Leblanc, R.M. Carbon dots: A future blood–brain barrier penetrating nanomedicine and drug nanocarrier. Int. J. Nanomedicine 2021, 16, 5003–5016. [Google Scholar] [CrossRef] [PubMed]
  106. Zhou, Y.; Liyanage, P.Y.; Devadoss, D.; Guevara, L.R.R.; Cheng, L.; Graham, R.M.; Chand, H.S.; Al-Youbi, A.O.; Bashammakh, A.S.; El-Shahawi, M.S. Nontoxic amphiphilic carbon dots as promising drug nanocarriers across the blood–brain barrier and inhibitors of β-amyloid. Nanoscale 2019, 11, 22387–22397. [Google Scholar] [CrossRef] [PubMed]
  107. Seven, E.S.; Sharma, S.K.; Meziane, D.; Zhou, Y.; Mintz, K.J.; Pandey, R.R.; Chusuei, C.C.; Leblanc, R.M. Close-packed langmuir monolayers of saccharide-based carbon dots at the air–subphase interface. Langmuir 2019, 35, 6708–6718. [Google Scholar] [CrossRef]
  108. Li, J.; Tian, M.; Cui, L.; Dwyer, J.; Fullwood, N.J.; Shen, H.; Martin, F.L. Low-dose carbon-based nanoparticle-induced effects in a549 lung cells determined by biospectroscopy are associated with increases in genomic methylation. Sci. Rep. 2016, 6, 20207. [Google Scholar] [CrossRef]
  109. Zhang, W.J.; Li, D.N.; Lian, T.H.; Guo, P.; Zhang, Y.N.; Li, J.H.; Guan, H.Y.; He, M.Y.; Zhang, W.J.; Zhang, W.J.; et al. Clinical features and potential mechanisms relating neuropathological biomarkers and blood-brain barrier in patients with alzheimer’s disease and hearing loss. Front. Aging Neurosci. 2022, 14, 911028. [Google Scholar] [CrossRef]
  110. Chung, Y.J.; Lee, B.I.; Park, C.B. Multifunctional carbon dots as a therapeutic nanoagent for modulating cu (ii)-mediated β-amyloid aggregation. Nanoscale 2019, 11, 6297–6306. [Google Scholar] [CrossRef]
  111. Yan, C.; Wang, C.; Shao, X.; Teng, Y.; Chen, P.; Hu, X.; Guan, P.; Wu, H. Multifunctional carbon-dot-photosensitizer nanoassemblies for inhibiting amyloid aggregates, suppressing microbial infection, and overcoming the blood–brain barrier. ACS Appl. Mater. Interfaces 2022, 14, 47432–47444. [Google Scholar] [CrossRef]
  112. Chung, Y.J.; Lee, C.H.; Lim, J.; Jang, J.; Kang, H.; Park, C.B. Photomodulating carbon dots for spatiotemporal suppression of alzheimer’s β-amyloid aggregation. ACS Nano 2020, 14, 16973–16983. [Google Scholar] [CrossRef]
  113. Gil, H.M.; Price, T.W.; Chelani, K.; Bouillard, J.-S.G.; Calaminus, S.D.J.; Stasiuk, G.J. Nir-quantum dots in biomedical imaging and their future. iScience 2021, 24, 102189. [Google Scholar] [CrossRef]
  114. Hasannejadasl, B.; Janbaz, F.P.; Choupani, E.; Fadaie, M.; Hamidinejad, M.A.; Ahmadvand, D. Quantum dots application in neurodegenerative diseases. Thrita 2020, 9. [Google Scholar] [CrossRef]
  115. Tak, K.; Sharma, R.; Dave, V.; Jain, S.; Sharma, S. Clitoria ternatea mediated synthesis of graphene quantum dots for the treatment of alzheimer’s disease. ACS Chem. Neurosci. 2020, 11, 3741–3748. [Google Scholar] [CrossRef] [PubMed]
  116. Wang, K.; Wang, L.; Chen, L.; Peng, C.; Luo, B.; Mo, J.; Chen, W. Intranasal administration of dauricine loaded on graphene oxide: Multi-target therapy for alzheimer’s disease. Drug Deliv. 2021, 28, 580–593. [Google Scholar] [CrossRef]
  117. Liu, C.; Luo, X. Potential molecular and graphene oxide chelators to dissolve amyloid-β plaques in alzheimer’s disease: A density functional theory study. J. Mater. Chem. B Mater. Biol. Med. 2021, 9, 2736–2746. [Google Scholar] [CrossRef] [PubMed]
  118. Li, X.; Li, K.; Chu, F.; Huang, J.; Yang, Z. Graphene oxide enhances β-amyloid clearance by inducing autophagy of microglia and neurons. Chem. Biol. Interact. 2020, 325, 109126. [Google Scholar] [CrossRef] [PubMed]
  119. Qin, Q.; Wang, X.; Shi, J.; Wu, D. Preparation of electrochemical sensor based on β-cyclodextrin/carbon nanotube nanocomposite for donepezil hydrochloride as drug for treatment of alzheimer’s disease. Int. J. Electrochem. Sci. 2022, 17, 220119. [Google Scholar] [CrossRef]
  120. Caffo, M.; Curcio, A.; Rajiv, K.; Caruso, G.; Venza, M.; Germanò, A. Potential role of carbon nanomaterials in the treatment of malignant brain gliomas. Cancers 2023, 15, 2575. [Google Scholar] [CrossRef]
  121. Cacciatore, I.; Ciulla, M.; Fornasari, E.; Marinelli, L.; Di Stefano, A. Solid lipid nanoparticles as a drug delivery system for the treatment of neurodegenerative diseases. Expert Opin. Drug Deliv. 2016, 13, 1121–1131. [Google Scholar] [CrossRef]
  122. Cacciatore, I.; Baldassarre, L.; Fornasari, E.; Mollica, A.; Pinnen, F. Recent advances in the treatment of neurodegenerative diseases based on gsh delivery systems. Oxidative Med. Cell. Longev. 2012, 2012, 240146. [Google Scholar] [CrossRef]
  123. Vakilinezhad, M.A.; Amini, A.; Akbari Javar, H.; Baha’addini Beigi Zarandi, B.F.; Montaseri, H.; Dinarvand, R. Nicotinamide loaded functionalized solid lipid nanoparticles improves cognition in alzheimer’s disease animal model by reducing tau hyperphosphorylation. DARU J. Pharm. Sci. 2018, 26, 165–177. [Google Scholar] [CrossRef]
  124. Arora, D.; Bhatt, S.; Kumar, M.; Verma, R.; Taneja, Y.; Kaushal, N.; Tiwari, A.; Tiwari, V.; Alexiou, A.; Albogami, S. Qbd-based rivastigmine tartrate loaded solid lipid nanoparticles for enhanced intranasal delivery to the brain for alzheimer’s therapeutics. Front. Aging Neurosci. 2022, 14, 960246. [Google Scholar] [CrossRef]
  125. Pinheiro, R.; Granja, A.; Loureiro, J.A.; Pereira, M.; Pinheiro, M.; Neves, A.; Reis, S. Rvg29-functionalized lipid nanoparticles for quercetin brain delivery and alzheimer’s disease. Pharm. Res. 2020, 37, 139. [Google Scholar] [CrossRef] [PubMed]
  126. Ross, C.; Taylor, M.; Fullwood, N.; Allsop, D. Liposome delivery systems for the treatment of alzheimer’s disease. Int. J. Nanomed. 2018, 13, 8507–8522. [Google Scholar] [CrossRef] [PubMed]
  127. Andrade, S.; Ramalho, M.J.; Loureiro, J.A.; Pereira, M.C. Transferrin-functionalized liposomes loaded with vitamin vb12 for alzheimer’s disease therapy. Int. J. Pharm. 2022, 626, 122167. [Google Scholar] [CrossRef]
  128. Andrade, S.; Pereira, M.C.; Loureiro, J.A. Caffeic acid loaded into engineered lipid nanoparticles for alzheimer’s disease therapy. Colloids Surf. B Biointerfaces 2023, 225, 113270. [Google Scholar] [CrossRef] [PubMed]
  129. Chauhan, A.S. Dendrimers for drug delivery. Molecules 2018, 23, 938. [Google Scholar] [CrossRef]
  130. Fazil, M.; Md, S.; Haque, S.; Kumar, M.; Baboota, S.; Sahni, J.K.; Ali, J. Development and evaluation of rivastigmine loaded chitosan nanoparticles for brain targeting. Eur. J. Pharm. Sci. 2012, 47, 6–15. [Google Scholar] [CrossRef]
  131. Sharma, A.K.; Gupta, L.; Sahu, H.; Qayum, A.; Singh, S.K.; Nakhate, K.T.; Gupta, U. Chitosan engineered pamam dendrimers as nanoconstructs for the enhanced anti-cancer potential and improved in vivo brain pharmacokinetics of temozolomide. Pharm. Res. 2018, 35, 9. [Google Scholar] [CrossRef]
  132. Singh, A.K.; Mishra, S.K.; Mishra, G.; Maurya, A.; Awasthi, R.; Yadav, M.K.; Atri, N.; Pandey, P.K.; Singh, S.K. Inorganic clay nanocomposite system for improved cholinesterase inhibition and brain pharmacokinetics of donepezil. Drug Dev. Ind. Pharm. 2020, 46, 8–19. [Google Scholar] [CrossRef]
  133. Igartúa, D.E.; Martinez, C.S.; Del, V.A.S.; Prieto, M.J. Combined therapy for alzheimer’s disease: Tacrine and pamam dendrimers co-administration reduces the side effects of the drug without modifying its activity. AAPS PharmSciTech 2020, 21, 110. [Google Scholar] [CrossRef]
  134. Igartúa, D.E.; Martinez, C.S.; Temprana, C.F.; Alonso, S.D.V.; Prieto, M.J. Pamam dendrimers as a carbamazepine delivery system for neurodegenerative diseases: A biophysical and nanotoxicological characterization. Int. J. Pharm. 2018, 544, 191–202. [Google Scholar] [CrossRef]
  135. Witika, B.A.; Poka, M.S.; Demana, P.H.; Matafwali, S.K.; Melamane, S.; Malungelo Khamanga, S.M.; Makoni, P.A. Lipid-based nanocarriers for neurological disorders: A review of the state-of-the-art and therapeutic success to date. Pharmaceutics 2022, 14, 836. [Google Scholar] [CrossRef] [PubMed]
  136. Zhang, W.; Mehta, A.; Tong, Z.; Esser, L.; Voelcker, N.H. Development of polymeric nanoparticles for blood-brain barrier transfer-strategies and challenges. Adv. Sci. 2021, 8, 2003937. [Google Scholar] [CrossRef] [PubMed]
  137. Mathew, A.; Fukuda, T.; Nagaoka, Y.; Hasumura, T.; Morimoto, H.; Yoshida, Y.; Maekawa, T.; Venugopal, K.; Kumar, D.S. Curcumin loaded-plga nanoparticles conjugated with tet-1 peptide for potential use in alzheimer’s disease. PLoS ONE 2012, 7, e32616. [Google Scholar] [CrossRef] [PubMed]
  138. Huang, N.; Lu, S.; Liu, X.-G.; Zhu, J.; Wang, Y.-J.; Liu, R.-T. Plga nanoparticles modified with a bbb-penetrating peptide co-delivering aβ generation inhibitor and curcumin attenuate memory deficits and neuropathology in alzheimer’s disease mice. Oncotarget 2017, 8, 81001. [Google Scholar] [CrossRef] [PubMed]
  139. Sánchez-López, E.; Ettcheto, M.; Egea, M.A.; Espina, M.; Calpena, A.C.; Folch, J.; Camins, A.; García, M.L. New potential strategies for alzheimer’s disease prevention: Pegylated biodegradable dexibuprofen nanospheres administration to appswe/ps1de9. Nanomed. Nanotechnol. Biol. Med. 2017, 13, 1171–1182. [Google Scholar] [CrossRef]
  140. Liu, X.G.; Zhang, L.; Lu, S.; Liu, D.Q.; Huang, Y.R.; Zhu, J.; Zhou, W.W.; Yu, X.L.; Liu, R.T. Superparamagnetic iron oxide nanoparticles conjugated with aβ oligomer-specific scfv antibody and class a scavenger receptor activator show therapeutic potentials for alzheimer’s disease. J. Nanobiotechnology 2020, 18, 160. [Google Scholar] [CrossRef]
  141. Zhang, W.; Zhang, Y.; Liu, X.; Zhang, Y.; Liu, Y.; Wang, W.; Su, R.; Sun, Y.; Huang, Y.; Song, D. Ratiometric fluorescence and colorimetric dual-mode sensing platform based on carbon dots for detecting copper (ii) ions and d-penicillamine. Anal. Bioanal. Chem. 2022, 414, 1651–1662. [Google Scholar] [CrossRef]
  142. Li, C.; Xiang, Y.; Wang, Y.; Li, P. Study on nano drug particles in the diagnosis and treatment of alzheimer’s disease in the elderly. Bioinorg. Chem. Appl. 2022, 2022, 3335581. [Google Scholar] [CrossRef]
  143. Han, Y.; Chu, X.; Cui, L.; Fu, S.; Gao, C.; Li, Y.; Sun, B. Neuronal mitochondria-targeted therapy for alzheimer’s disease by systemic delivery of resveratrol using dual-modified novel biomimetic nanosystems. Drug Deliv. 2020, 27, 502–518. [Google Scholar] [CrossRef]
  144. Agwa, M.M.; Abdelmonsif, D.A.; Khattab, S.N.; Sabra, S. Self-assembled lactoferrin-conjugated linoleic acid micelles as an orally active targeted nanoplatform for alzheimer’s disease. Int. J. Biol. Macromol. 2020, 162, 246–261. [Google Scholar] [CrossRef]
  145. Nanaki, S.G.; Spyrou, K.; Bekiari, C.; Veneti, P.; Baroud, T.N.; Karouta, N.; Grivas, I.; Papadopoulos, G.C.; Gournis, D.; Bikiaris, D.N. Hierarchical porous carbon-plla and plga hybrid nanoparticles for intranasal delivery of galantamine for alzheimer’s disease therapy. Pharmaceutics 2020, 12, 227. [Google Scholar] [CrossRef] [PubMed]
  146. Sintov, A.C. Amylolipid nanovesicles: A self-assembled lipid-modified starch hybrid system constructed for direct nose-to-brain delivery of curcumin. Int. J. Pharm. 2020, 588, 119725. [Google Scholar] [CrossRef] [PubMed]
  147. Hou, K.; Zhao, J.; Wang, H.; Li, B.; Li, K.; Shi, X.; Wan, K.; Ai, J.; Lv, J.; Wang, D. Chiral gold nanoparticles enantioselectively rescue memory deficits in a mouse model of alzheimer’s disease. Nat. Commun. 2020, 11, 4790. [Google Scholar] [CrossRef] [PubMed]
  148. Gothwal, A.; Kumar, H.; Nakhate, K.T.; Ajazuddin; Dutta, A.; Borah, A.; Gupta, U. Lactoferrin coupled lower generation pamam dendrimers for brain targeted delivery of memantine in aluminum-chloride-induced alzheimer’s disease in mice. Bioconjug. Chem. 2019, 30, 2573–2583. [Google Scholar] [CrossRef]
  149. Dara, T.; Vatanara, A.; Meybodi, M.N.; Vakilinezhad, M.A.; Malvajerd, S.S.; Vakhshiteh, F.; Shamsian, A.; Sharifzadeh, M.; Kaghazian, H.; Mosaddegh, M.H. Erythropoietin-loaded solid lipid nanoparticles: Preparation, optimization, and in vivo evaluation. Colloids Surf. B Biointerfaces 2019, 178, 307–316. [Google Scholar] [CrossRef]
  150. Silva-Abreu, M.; Espinoza, L.C.; Halbaut, L.; Espina, M.; García, M.L.; Calpena, A.C. Comparative study of ex vivo transmucosal permeation of pioglitazone nanoparticles for the treatment of alzheimer’s disease. Polymers 2018, 10, 316. [Google Scholar] [CrossRef]
  151. Meng, Q.; Wang, A.; Hua, H.; Jiang, Y.; Wang, Y.; Mu, H.; Wu, Z.; Sun, K. Intranasal delivery of huperzine a to the brain using lactoferrin-conjugated n-trimethylated chitosan surface-modified plga nanoparticles for treatment of alzheimer’s disease. Int. J. Nanomed. 2018, 13, 705–718. [Google Scholar] [CrossRef]
  152. Muller, A.P.; Ferreira, G.K.; Pires, A.J.; de Bem Silveira, G.; de Souza, D.L.; Brandolfi, J.A.; de Souza, C.T.; Paula, M.M.S.; Silveira, P.C.L. Gold nanoparticles prevent cognitive deficits, oxidative stress and inflammation in a rat model of sporadic dementia of alzheimer’s type. Mater. Sci. Eng. C Mater. Biol. Appl. 2017, 77, 476–483. [Google Scholar] [CrossRef]
  153. Sun, D.; Li, N.; Zhang, W.; Zhao, Z.; Mou, Z.; Huang, D.; Liu, J.; Wang, W. Design of plga-functionalized quercetin nanoparticles for potential use in alzheimer’s disease. Colloids Surf. B Biointerfaces 2016, 148, 116–129. [Google Scholar] [CrossRef]
  154. Misra, S.; Chopra, K.; Sinha, V.R.; Medhi, B. Galantamine-loaded solid-lipid nanoparticles for enhanced brain delivery: Preparation, characterization, in vitro and in vivo evaluations. Drug Deliv. 2016, 23, 1434–1443. [Google Scholar] [CrossRef]
  155. Meng, F.; Asghar, S.; Xu, Y.; Wang, J.; Jin, X.; Wang, Z.; Wang, J.; Ping, Q.; Zhou, J.; Xiao, Y. Design and evaluation of lipoprotein resembling curcumin-encapsulated protein-free nanostructured lipid carrier for brain targeting. Int. J. Pharm. 2016, 506, 46–56. [Google Scholar] [CrossRef]
  156. Shah, B.; Khunt, D.; Bhatt, H.; Misra, M.; Padh, H. Application of quality by design approach for intranasal delivery of rivastigmine loaded solid lipid nanoparticles: Effect on formulation and characterization parameters. Eur. J. Pharm. Sci. 2015, 78, 54–66. [Google Scholar] [CrossRef]
  157. Yin, H.; Si, J.; Xu, H.; Dong, J.; Zheng, D.; Lu, X.; Li, X. Resveratrol-loaded nanoparticles reduce oxidative stress induced by radiation or amyloid-in transgenic caenorhabditis elegans. J. Biomed. Nanotechnol. 2014, 10, 1536–1544. [Google Scholar] [CrossRef]
  158. Liu, Z.; Gao, X.; Kang, T.; Jiang, M.; Miao, D.; Gu, G.; Hu, Q.; Song, Q.; Yao, L.; Tu, Y.; et al. B6 peptide-modified peg-pla nanoparticles for enhanced brain delivery of neuroprotective peptide. Bioconjug. Chem. 2013, 24, 997–1007. [Google Scholar] [CrossRef]
  159. Yang, Z.Z.; Zhang, Y.Q.; Wang, Z.Z.; Wu, K.; Lou, J.N.; Qi, X.R. Enhanced brain distribution and pharmacodynamics of rivastigmine by liposomes following intranasal administration. Int. J. Pharm. 2013, 452, 344–354. [Google Scholar] [CrossRef] [PubMed]
  160. Zhang, C.; Chen, J.; Feng, C.; Shao, X.; Liu, Q.; Zhang, Q.; Pang, Z.; Jiang, X. Intranasal nanoparticles of basic fibroblast growth factor for brain delivery to treat alzheimer’s disease. Int. J. Pharm. 2014, 461, 192–202. [Google Scholar] [CrossRef] [PubMed]
  161. Luppi, B.; Bigucci, F.; Corace, G.; Delucca, A.; Cerchiara, T.; Sorrenti, M.; Catenacci, L.; Di Pietra, A.M.; Zecchi, V. Albumin nanoparticles carrying cyclodextrins for nasal delivery of the anti-alzheimer drug tacrine. Eur. J. Pharm. Sci. 2011, 44, 559–565. [Google Scholar] [CrossRef] [PubMed]
  162. Canovi, M.; Markoutsa, E.; Lazar, A.N.; Pampalakis, G.; Clemente, C.; Re, F.; Sesana, S.; Masserini, M.; Salmona, M.; Duyckaerts, C. The binding affinity of anti-aβ1-42 mab-decorated nanoliposomes to aβ1-42 peptides in vitro and to amyloid deposits in post-mortem tissue. Biomaterials 2011, 32, 5489–5497. [Google Scholar] [CrossRef] [PubMed]
  163. Wilson, B.; Samanta, M.K.; Santhi, K.; Kumar, K.P.; Paramakrishnan, N.; Suresh, B. Poly(n-butylcyanoacrylate) nanoparticles coated with polysorbate 80 for the targeted delivery of rivastigmine into the brain to treat alzheimer’s disease. Brain Res. 2008, 1200, 159–168. [Google Scholar] [CrossRef] [PubMed]
  164. Kong, L.; Li, X.-t.; Ni, Y.-n.; Xiao, H.-h.; Yao, Y.-j.; Wang, Y.-y.; Ju, R.-j.; Li, H.-y.; Liu, J.-j.; Fu, M. Transferrin-modified osthole pegylated liposomes travel the blood-brain barrier and mitigate alzheimer’s disease-related pathology in app/ps-1 mice. Int. J. Nanomed. 2020, 15, 2841–2858. [Google Scholar] [CrossRef]
  165. Rotman, M.; Welling, M.M.; Bunschoten, A.; de Backer, M.E.; Rip, J.; Nabuurs, R.J.; Gaillard, P.J.; van Buchem, M.A.; van der Maarel, S.M.; van der Weerd, L. Enhanced glutathione pegylated liposomal brain delivery of an anti-amyloid single domain antibody fragment in a mouse model for alzheimer’s disease. J. Control. Release 2015, 203, 40–50. [Google Scholar] [CrossRef] [PubMed]
  166. Arora, S.; Layek, B.; Singh, J. Design and validation of liposomal apoe2 gene delivery system to evade blood–brain barrier for effective treatment of alzheimer’s disease. Mol. Pharm. 2020, 18, 714–725. [Google Scholar] [CrossRef] [PubMed]
  167. Ruff, J.; Hassan, N.; Morales-Zavala, F.; Steitz, J.; Araya, E.; Kogan, M.J.; Simon, U. Clpffd–peg functionalized nir-absorbing hollow gold nanospheres and gold nanorods inhibit β-amyloid aggregation. J. Mater. Chem. B Mater. Biol. Med. 2018, 6, 2432–2443. [Google Scholar] [CrossRef]
  168. Li, M.; Xu, C.; Wu, L.; Ren, J.; Wang, E.; Qu, X. Self-assembled peptide-polyoxometalate hybrid nanospheres: Two in one enhances targeted inhibition of amyloid β-peptide aggregation associated with alzheimer’s disease. Small 2013, 9, 3455–3461. [Google Scholar] [CrossRef] [PubMed]
  169. Lu, Y.; Guo, Z.; Zhang, Y.; Li, C.; Zhang, Y.; Guo, Q.; Chen, Q.; Chen, X.; He, X.; Liu, L.; et al. Microenvironment remodeling micelles for alzheimer’s disease therapy by early modulation of activated microglia. Adv. Sci. 2019, 6, 1801586. [Google Scholar] [CrossRef]
  170. Gothwal, A.; Lamptey, R.N.L.; Singh, J. Multifunctionalized cationic chitosan polymeric micelles polyplexed with pvgf for noninvasive delivery to the mouse brain through the intranasal route for developing therapeutics for alzheimer’s disease. Mol. Pharm. 2023, 20, 3009–3019. [Google Scholar] [CrossRef]
  171. Parikh, A.; Kathawala, K.; Li, J.; Chen, C.; Shan, Z.; Cao, X.; Zhou, X.-F.; Garg, S. Curcumin-loaded self-nanomicellizing solid dispersion system: Part ii: In vivo safety and efficacy assessment against behavior deficit in alzheimer disease. Drug Deliv. Transl. Res. 2018, 8, 1406–1420. [Google Scholar] [CrossRef]
  172. Elnaggar, Y.S.; Etman, S.M.; Abdelmonsif, D.A.; Abdallah, O.Y. Intranasal piperine-loaded chitosan nanoparticles as brain-targeted therapy in alzheimer’s disease: Optimization, biological efficacy, and potential toxicity. J. Pharm. Sci. 2015, 104, 3544–3556. [Google Scholar] [CrossRef]
  173. Jiang, Z.; Dong, X.; Sun, Y. Charge effects of self-assembled chitosan-hyaluronic acid nanoparticles on inhibiting amyloid β-protein aggregation. Carbohydr. Res. 2018, 461, 11–18. [Google Scholar] [CrossRef]
  174. Kuang, Y.; Zhang, J.; Xiong, M.; Zeng, W.; Lin, X.; Yi, X.; Luo, Y.; Yang, M.; Li, F.; Huang, Q. A novel nanosystem realizing curcumin delivery based on fe3o4@ carbon dots nanocomposite for alzheimer’s disease therapy. Front. Bioeng. Biotechnol. 2020, 8, 614906. [Google Scholar] [CrossRef]
  175. De la Torre, C.; Ceña, V. The delivery challenge in neurodegenerative disorders: The nanoparticles role in alzheimer’s disease therapeutics and diagnostics. Pharmaceutics 2018, 10, 190. [Google Scholar] [CrossRef]
  176. Lamptey, R.N.L.; Chaulagain, B.; Trivedi, R.; Gothwal, A.; Layek, B.; Singh, J. A review of the common neurodegenerative disorders: Current therapeutic approaches and the potential role of nanotherapeutics. Int. J. Mol. Sci. 2022, 23, 1851. [Google Scholar] [CrossRef] [PubMed]
  177. Kimura, S.; Harashima, H. Current status and challenges associated with cns-targeted gene delivery across the bbb. Pharmaceutics 2020, 12, 1216. [Google Scholar] [CrossRef] [PubMed]
  178. Ren, J.; Jiang, F.; Wang, M.; Hu, H.; Zhang, B.; Chen, L.; Dai, F. Increased cross-linking micelle retention in the brain of alzheimer’s disease mice by elevated asparagine endopeptidase protease responsive aggregation. Biomater. Sci. 2020, 8, 6533–6544. [Google Scholar] [CrossRef] [PubMed]
  179. Ding, S.; Khan, A.I.; Cai, X.; Song, Y.; Lyu, Z.; Du, D.; Dutta, P.; Lin, Y. Overcoming blood-brain barrier transport: Advances in nanoparticle-based drug delivery strategies. Mater. Today 2020, 37, 112–125. [Google Scholar] [CrossRef] [PubMed]
  180. Hersh, A.M.; Alomari, S.; Tyler, B.M. Crossing the blood-brain barrier: Advances in nanoparticle technology for drug delivery in neuro-oncology. Int. J. Mol. Sci. 2022, 23, 4153. [Google Scholar] [CrossRef]
  181. Saucier-Sawyer, J.K.; Deng, Y.; Seo, Y.E.; Cheng, C.J.; Zhang, J.; Quijano, E.; Saltzman, W.M. Systemic delivery of blood-brain barrier-targeted polymeric nanoparticles enhances delivery to brain tissue. J. Drug Target. 2015, 23, 736–749. [Google Scholar] [CrossRef]
  182. Ling, T.S.; Chandrasegaran, S.; Xuan, L.Z.; Suan, T.L.; Elaine, E.; Nathan, D.V.; Chai, Y.H.; Gunasekaran, B.; Salvamani, S. The potential benefits of nanotechnology in treating alzheimer’s disease. Biomed. Res. Int. 2021, 2021, 5550938. [Google Scholar] [CrossRef]
  183. Bashir, W.; Shahzadi, S. Nanoparticles–a novel theranostic approach to treat alzheimer’s disease. J. Appl. Biotechnol. Bioeng. 2022, 9, 216–220. [Google Scholar] [CrossRef]
  184. Harish, V.; Tewari, D.; Gaur, M.; Yadav, A.B.; Swaroop, S.; Bechelany, M.; Barhoum, A. Review on nanoparticles and nanostructured materials: Bioimaging, biosensing, drug delivery, tissue engineering, antimicrobial, and agro-food applications. Nanomaterials 2022, 12, 457. [Google Scholar] [CrossRef]
  185. Chakraborty, A.; Mohapatra, S.S.; Barik, S.; Roy, I.; Gupta, B.; Biswas, A. Impact of nanoparticles on amyloid beta-induced alzheimer’s disease, tuberculosis, leprosy and cancer: A systematic review. Biosci. Rep. 2023, 43, BSR20220324. [Google Scholar] [CrossRef] [PubMed]
  186. Kulavi, S.; Kaur, R.; Iyer, K.; Bandyopadhyay, J.; Sengupta, T. A smart & precise approach with nanoparticles-based therapeutic intervention in neurodegenerative diseases. Nanomed. J. 2023, 10, 96–106. [Google Scholar]
  187. Pardridge, W.M. Csf, blood-brain barrier, and brain drug delivery. Expert Opin. Drug Deliv. 2016, 13, 963–975. [Google Scholar] [CrossRef] [PubMed]
  188. Kung, Y.; Chen, K.-Y.; Liao, W.-H.; Hsu, Y.-H.; Wu, C.-H.; Hsiao, M.-Y.; Huang, A.P.H.; Chen, W.-S. Facilitating drug delivery in the central nervous system by opening the blood-cerebrospinal fluid barrier with a single low energy shockwave pulse. Fluids Barriers CNS 2022, 19, 3. [Google Scholar] [CrossRef] [PubMed]
  189. Puris, E.; Gynther, M.; Auriola, S.; Huttunen, K.M. L-type amino acid transporter 1 as a target for drug delivery. Pharm. Res. 2020, 37, 88. [Google Scholar] [CrossRef] [PubMed]
  190. Mir, S.A.; Hamid, L.; Bader, G.N.; Shoaib, A.; Rahamathulla, M.; Alshahrani, M.Y.; Alam, P.; Shakeel, F. Role of nanotechnology in overcoming the multidrug resistance in cancer therapy: A review. Molecules 2022, 27, 6608. [Google Scholar] [CrossRef]
  191. Zhang, C.; Zhou, X.; Zhang, H.; Han, X.; Li, B.; Yang, R.; Zhou, X. Recent progress of novel nanotechnology challenging the multidrug resistance of cancer. Front. Pharmacol. 2022, 13, 776895. [Google Scholar] [CrossRef]
  192. Wei, M.; Yang, Z.; Li, S.; Le, W. Nanotherapeutic and stem cell therapeutic strategies in neurodegenerative diseases: A promising therapeutic approach. Int. J. Nanomedicine 2023, 18, 611–626. [Google Scholar] [CrossRef]
  193. Richardson, J.J.; Caruso, F. Nanomedicine toward 2040; ACS Publications: Washington, DC, USA, 2020; Volume 20, pp. 1481–1482. [Google Scholar]
  194. Poudel, P.; Park, S. Recent advances in the treatment of alzheimer’s disease using nanoparticle-based drug delivery systems. Pharmaceutics 2022, 14, 835. [Google Scholar] [CrossRef]
  195. Fonseca-Santos, B.; Gremião, M.P.D.; Chorilli, M. Nanotechnology-based drug delivery systems for the treatment of alzheimer’s disease. Int. J. Nanomedicine 2015, 10, 4981–5003. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions, and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of the MDPI and/or editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions, or products referred to in the content.
Figure 1. Pathophysiological changes in the CNS in AD. (A) Formation of Aβ plaques. (B) Formation of NFT. (C) Synaptic disruption. (D) Demyelination. (E) Autophagy. (F) Astrogliosis–Microgliosis. (created with BioRender.com accessed on 10 August 2023).
Figure 1. Pathophysiological changes in the CNS in AD. (A) Formation of Aβ plaques. (B) Formation of NFT. (C) Synaptic disruption. (D) Demyelination. (E) Autophagy. (F) Astrogliosis–Microgliosis. (created with BioRender.com accessed on 10 August 2023).
Ijms 24 14044 g001
Figure 2. A schematic representation of the blood–brain barrier (created with BioRender.com accessed on 10 August 2023).
Figure 2. A schematic representation of the blood–brain barrier (created with BioRender.com accessed on 10 August 2023).
Ijms 24 14044 g002
Figure 3. A graphical illustration of possible pathways of nanomaterial-based DDSs that effectively cross the blood–brain barrier for treating Alzheimer’s disease (created with BioRender.com accessed on 10 August 2023).
Figure 3. A graphical illustration of possible pathways of nanomaterial-based DDSs that effectively cross the blood–brain barrier for treating Alzheimer’s disease (created with BioRender.com accessed on 10 August 2023).
Ijms 24 14044 g003
Figure 4. A general overview of nanoparticles used as drug delivery carriers in the management and treatment of AD (created with BioRender.com accessed on 10 August 2023).
Figure 4. A general overview of nanoparticles used as drug delivery carriers in the management and treatment of AD (created with BioRender.com accessed on 10 August 2023).
Ijms 24 14044 g004
Figure 5. Challenges of nanomedicine for Alzheimer’s therapeutics.
Figure 5. Challenges of nanomedicine for Alzheimer’s therapeutics.
Ijms 24 14044 g005
Table 1. Therapeutic targets for management and treatment of AD (Individual image was adopted from BioRender.com; 10 August 2023).
Table 1. Therapeutic targets for management and treatment of AD (Individual image was adopted from BioRender.com; 10 August 2023).
Target and Its StructureRole in ADTherapeutic StrategiesCommon DrugsReferences
Amyloid beta protein
Ijms 24 14044 i001
Increases in Aβ concentration, forms oligomers and leads to neurotoxicityAβ-protein-targeted (Aβ oligomers treatment) therapeutic strategies for treating ADAducanumab, CPO-Aβ17–21 peptide, Huperzine A [22,23,24,25]
Tau protein
Ijms 24 14044 i002
The misfolded Tau is self-assembled, forming a Tau oligomer, and then, Tau inclusions lead to ADReduction in toxic Tau gain of function may be an effective therapeutic strategy for ADGosuranemab, lonafarnib tilavonemab, semorinemab, zagotenemab, [26,27]
Neurofibrillary tangles
Ijms 24 14044 i003
Excessive Tau phosphorylation and aggregation are the key processes in the formation of NFT that cause neuron death and lead to ADPrevent phosphatase activity; promote microtubule stability; reduce Tau aggregate and formation of NFTMemantine, AADvac1, ACI-35; Epithilone D[28,29]
Mitochondria
Ijms 24 14044 i004
Mitochondrial dysfunction cause AD, which involves OS-induced respiratory chain dysfunction, loss of mitochondrial biogenesis, and mitochondrial DNA mutationsTherapeutics based on antioxidants, anti-apoptotic agents, and molecules that enhance glucose metabolism and mitochondrial bioenergeticsKaempferol (flavonoid) and rhapontigenin (stilbenoid), P110 and mdivi-1, selenium, SkQ1, MitoApo, astaxanthin[30,31,32,33]
Oxidative stress
Ijms 24 14044 i005
Neuronal cell abnormalities; apoptosis of neurons, cognitive dysfunction that leads to dementiaAntioxidant drug therapy has been investigated as a potential AD treatmentVitamin C, Vitamin E, -lipoic acid, CoQ10, Curcumin; coenzyme Q and glutathione; melatonin, α-lipoic acid (LA), N-Acetyl-cysteine (NAC)[34,35,36,37,38]
Neuroinflammation
Ijms 24 14044 i006
Neuroinflammation leading to memory loss and cognitive declineNeuroprotective therapeutics using nonsteroidal anti-inflammatory drugsPterostilbene, Sulforaphane, Artemisinin[39,40,41]
Angiotensin receptors
Ijms 24 14044 i007
Their role in blood pressure regulation and hypertension leads to neural injury, neuroinflammation, and cognitive functionTherapeutic agents that inhibit RAS, Angiotensin-converting enzymes, ARBs, and block Angiotensin Receptor Ramipril (ACE inhibitor), ARBs, ACE-Is, RAS-Ms, telmisartan, candesartan, valsartan[42,43,44,45,46,47]
Secretase receptors
Ijms 24 14044 i008
Caused AD by changes in Aβ stability and aggregationTherapeutic agents that cleaved APP α-secretase have potential to treat ADMK-8931 (inhibitor of β-site APP cleaving enzyme 1), PRX-03,140 (α-Secretase inhibitors), Begacestat (γ-Secretase inhibitors), CTS-2166 (β-Secretase inhibitors)[48,49,50,51]
Blood–brain barrier
Ijms 24 14044 i009
BBB characterizes a link between ND, vascular damage, and neuro-inflammationDrugs that can penetrate the BBB ApoE-modifying agents; Neprilysin[52,53,54]
Cholinergic insufficiency
Ijms 24 14044 i010
Reduced choline acetyltransferase (ChAT) and AChE activity in the cortex in AD Cholinesterase inhibitors Physostigmine, Aricept (donepezil), Exelon (rivastigmine), and Metrifonate[55,56,57]
Table 2. Summary of Nanocarriers Used for AD treatment (only in/ex vivo study mentioned here).
Table 2. Summary of Nanocarriers Used for AD treatment (only in/ex vivo study mentioned here).
Nanocarrier MaterialModifications/
Functionalized by
Therapeutic AgentExperimental ModelRoute of AdministrationExperimental Doses and PeriodsResultLimitations
and Possibilities
Reference
Solid Lipid Nanoparticles Polysorbate 80Rivastigmine tartrateSheep mucosaIntranasal0.178 mg/mL
6 months
Formulation shows safe intranasal delivery without any toxicityAn in vivo study is needed to see the safety and effectiveness of formulation[124]
Superparamagnetic Iron Oxide
NPs
-AβOs Antibody and Class A scavenger receptor activatorMiceIntravenously1 mg/day
28 days
Formulation targeting the AβOs improved the uptake of AβOs by microgliaThe distribution and the half-life of formulation will be studies in brains for long time benefits[140]
Carbon Dots-MemantineWild-type zebrafishIntravascularly-Cross the BBB and inhibes the tau aggregationThis study shows the significant therapeutic potential in AD treatment[141]
Nano drug particles-DrugAD patientsOral-
45 days
Have high stability and less
toxicity
A large level study is needed to conclude the role of nanodrug in AD treatment[142]
Graphene oxide-DauricineMiceIntranasal1 µg/µL
21 days
The formulation protects against oxidative damage and apoptosis Graphene oxide-dauricine crosses the BBB, enters the brain and shows potential therpeutic agent in AD treatment[116]
Nanostructured lipid carriers RBC membrane vesiclesGlycoprotein of Rabies virus and triphenylphosphine cationAPP/PS1 miceIntravenously2 mg/kg/every 2 days
30 days
Conquered the ROS-induced mitochondrial dysfunction Mitochondria-targeted nanosystems showing a capable therapeutic candidate for AD treatment[143]
Graphene Quantum Dots-Flower Clitoria ternatea extractRat-3 mg/kg
7 days
Improved cognitive behavior and memory capacity
in experimental model
QDs significantly crossed the BBB and significantly reduced the Alzheimer-like symptoms in rats; shows potential as therapeutic DDS[115]
Linoleic acid micelles-lactoferrinWistar ratsOral500 mg/Kg
60 days (AlCl3 induction) and +30 days (treatment regimen)
Enhanced cognitive capabilities, reduced brain OS, apoptosis, inflammation, and AchE activityMultiple functions of formulation show a greater potential in AD treatment and can be further evaluated at large level [144]
PLLA/PLGA
Hybrid NPs
Hierarchical porous
carbon (HPC)
GalantamineWistar ratsIntranasal3 mg/kg
24–48 h (clinical evaluation)
Successful delivery to the hippocampus area Further evaluation in rodent models of AD is needed[145]
Amylo Lipid NanovesiclesLipid-modified starch hybridCurcuminRatIntranasal160 μg/kg
24 h
Curcumin targeting specifically to the brain Result shows potential of mylo lipid nanovesicles in drug delivery to brain[146]
Chiral AuNPsL- and D-glutathioneNAKM miceIntravenous25 mg/kg
6 days
Formulation crossed the BBB and inhibited Aβ42 aggregation The formulation shows a promising therapeutic strategy for AD by significantly rescueing the spatial learning and memory impairments in AD model[147]
PAMAM DendrimersLactoferrin coupledMemantineMiceIntravenousmg/kg
8 weeks
The formulation enhances the memory improvementPAMAM dendrimers shows a significant impact on the memory aspects in AD-induced mice and could be further evaluated in other AD model[148]
SLNPs-ErythropoietinWistar ratsInjected in CA1
Region of hippocampus
1250 and 2500 U/kg
28 days
Formulation reduced the OS, ADP/ATP ratio and Aβ
plaque deposition
The result of in vivo study is encouraging for further evaluations[149]
Polymeric PLGA NPsPEG linkedPioglitazone (PGZ)Buccal and nasal mucosaOral110 µg/mL
6 h
Cross BBB and drug delivered to the brainIn vivo study is required to see the actual result [150]
SLNPsPolysorbate 80,
phosphatidylserine or phosphatidic acid
NicotinamideRatIntravenous
and
Intraperitoneal
200 mg/kg
4 days
Improving cognition, protecting the neuronal cells and
suppress Tau hyperphosphorylation
Not sufficient in improving the AD at early stages[123]
Polylactide-coglycoside NPsLactoferrin-conjugated N-trimethylated
chitosan
Huperzine AKM miceIntranasal1 µg/mL
12 h
Have a prolonged release time of the drug and target specific deliveryEvaluation of therapeutic efficacy for formulation in animal AD models is needed[151]
AuNPs-StreptozotocinWistar male ratsIntraperitoneal2.5 mg/kg body weight
21 days
Prevent mitochondrially
ATP production, neuroinflammation, and OS
AuNPs treatment prevented the pathological events of brain during AD pathogenesis possibly use for treatment of AD[152]
PLGA-NPsPVAQuercetinAD control miceIntravenous20 mg/kg body
Weight
30 days
Significantly improved
the spatial memory
The formulation have high therapeutic index and reduced the side effects[153]
SLNPs-Galantamine hydrobromideWistar ratsIntraperitoneal5 mg/kg
26 days
Significant restored memory capability in cognitive deficit RatsHigh bioavailability of drug in brain shows potential therapeutic role of formulation[154]
Nanostructured lipid carrier (NLC)Lactoferrin modifiedCurcuminRatsIntraperitoneal10 mg/kg body
Weight
24 h
Penetrate BBB and release the drug in
the brain
The in vitro and in vivo results shows the safe and biocompatible nanoformulation and could be used for brain targeted DDS[155]
SLNPs-RivastigmineGoat nasal
mucosa
Incubated with formulation10 mg/mL
9 h
Showed intact nasal mucosa with RHT SLN
indicating the safety of RHT SLN
In vivo study is needed to observe the actual biodistribution and release profile of drug [156]
Polyethyleneglycol-poly-caprolactone (mPEG-PCL)-ResveratrolTransgenic Caenorhabditis elegans (C. elegans)
(CL4176 strains)
Through culture media0–100 µg/mL
50 days
The formulation shows protection against OS in C. elegans induced by both γ-ray radiation and amyloid- peptide, confirming the
successful development of antioxidant NPs.
Further study in large animals is suggested for confirmation of potential DDS[157]
Poly(lactic acid) NPsPEG-coatedB6 peptide (CGHKAKGPRK)MiceIntravenous1 mg/kg
30 days
Showed excellent
amelioration in learning impairments, cholinergic disruption
In vivo imaging results show a good biodistribution profile of formulation with significant level of accumulation in the brain[158]
LiposomesModified by adding Cell-penetrating
peptide
RivastigmineMale ratsIntranasal1 g/kg body
Weight
7 days
Enhance the pharmacological properties of drug and BBB penetration Liposomes improve the drug delivery to brain by penetrating BBB and decrease the hepatic first pass metabolism and gastrointestinal adversative effects[159]
Polyethylene glycol-polylactide-polyglycolide NPsLectins modifiedFibroblast growth factorRatIntranasal20 and 40 µg/kg/d 17 daysSignificantly
improved spatial learning and memory in experimental rats
The formulation has a promising DDS for peptide and protein drugs for CNS and play the therapeutic role in AD[160]
Albumin NPsCyclodextrinsTacrine hydrochlorideSheep nasal mucosa/Ex vivoNasal delivery2 mg/mL
24 h
Modified NPs enhanced the drug loading and permeation In vivo study is
required
[161]
LiposomesBiotin-coatedAβ-mAbPost-mortem AD tissueIncubation10 pMoles/mg
Lipid
30 days
Aβ deposits in post-mortem AD brainPresent study shows the diagnostic potential only[162]
Poly(n-butylcyanoacrylate) NPsCoated with polysorbate 80RivastigmineWistar rats_273.0 and 408.2 ng/mL
24 h
Enhanced drug delivery 3.82-fold comparatively without nano-formulationsIn vitro release studies were performed; however, in vivo drug release studies were not performed[163]
Nanoliposomes -MetforminRatsi.c.v injection50 mg/kg
21 days
Improve memory deficit and reduce neuroinflammation in streptozotocin-induced AD modelNanoliposome conjugated metformin enhanced the learning and memory
impairment in patients suffering from AD and also suppressed the neuroinflammation
[93]
PEGylated LiposomesTransferrin OstholeAPP/PS-1 MiceIntravenous10 mg/kg
48 h
The formulation exert a protective effect in animal by targeting the Osthole to brain tissue and accumulating Osthole in the brain for long time of periodThe in vivo study along with pharmacodynamic and pharmacokinetic confirmed that liposome-based delivery of osthole has long-term circulation, controlled release, and target-specific release and effects[164]
PEGylated LiposomesGlutathioneAnti-amyloid single domain antibody fragmentAPPswe/PS1dE9 miceIntravenous5 μg/body weight
24 h
Liposome-based delivery of antibody fragments cross the BBB and reach into the brainPharmacokinetics and biodistribution studies of formulation confirmed the potential of liposome as DDS[165]
LiposomesCPPApoE2 GeneC57BL/6 miceIntravenous1 μg pApoE2/g body weight
5 days
Modified liposomes crossed the BBB and delivered the target gene to brain Liposome-based gene delivery is safe and have potential in AD treatment [166]
Gold nanospheres and gold nanorodsPolyethylene glycolCLPFFD peptideIn vitro/SH-SY5Y cells-20 μM to 1.4 nM
2 h
Conjugates inhibits Aβ-fibrillation An in vivo/ex vivo study is needed [167]
Polyoxometalate Nanospheres-Aβ peptideIn vitro/PC12 cells-10 μM
48 h
Inhibits the aggregation of amyloid β-peptide in AD model An in vivo/ex vivo study is needed[168]
Polymeric micellePEGCurcuminAPPswe/PSEN1dE9 model miceIntravenous5 mg/kg
38 weeks
Decreased Aβ plaque accumulation and enhanced cognitive behavior in miceFormulation shows targeted multiple target strategies that are more effective than single target strategies[169]
Linoleic acid micelles-LactoferrinWistar ratsOrallyAlCl3—100 mg/kg—30 days
LF-CLA micelles-500 mg/kg
30 days
Formulation enhanced cognitive capabilities, reduced brain oxidative stress, inflammationAdditional animal studies are essential to reconnoiter the potential effect of formulated micelles in AD prevention[144]
Polymeric MicellesCationic ChitosanpVGFC57BL6/J miceIntranasal1 mg/kg body weight
7 days
Formulation effectively deliver pVGF gene to the brain Formulation shows the potential of a nonviral gene delivery system for brain-targeted gene[170]
Nanomicellizing solid dispersion-CurcuminAPPSwe/PS1deE9Orally47 mg/kg
17 months
Formulation shows potential therapeutic
candidate
This study of the effect on other organ demonstrates that the formulation is safe and have a
promising potential as a therapeutic candidate for AD
[171]
Chitosan Nanoparticles_PiperineWistar ratsIntranasal0.250 mg/kg/day
22 days
Targated delivery and showing via anti-apoptosis and anti-inflammatory effectsThe formulation shows a tremendous efficiency as therpeutic agent, and could be further studied at large level[172]
Chitosan-Hyaluronic acidSH-SY5Y cells-25 µM
48 h
Inhibited Aβ aggregationAn in vivo/ex vivo study is needed to understand the impact of negative and positive surface charges of nano-inhibitors[173]
Selenium (Se) nanoparticles Sialic acidPeptide-B6 peptidePC12 cells.-10 μM
4 h
High permeability and cross the BBB An in vivo/ex vivo study is needed[102]
PLGA Nanoparticles CurcuminTet-1 PeptideGI-1 glioma cells-0.75 μg/mL, 1.5 μg/mL and 3 μg/mL
72 h
The formulation destroy amyloid aggregates and exhibit anti-oxidative properties An in vivo/ex vivo study is needed[137]
Poly(lactic-co-glycolic) PegylatedDexibuprofenAPPswe/PS1dE9Oral39 μg/mL–5290 μg/mL
30 min
It reduces the memory impairment and decrease brain inflammationThe accumulation of drug can be
observed on the liver and will be studied for safety purpose
[139]
Fe3O4@Carbon DotsFe3O4CurcuminPC12 cells-0.4 mg/mL
24–28 h
Inhibits the extracellular Aβ fibrillationAn in vivo/ex vivo study is needed[174]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Puranik, N.; Yadav, D.; Song, M. Advancements in the Application of Nanomedicine in Alzheimer’s Disease: A Therapeutic Perspective. Int. J. Mol. Sci. 2023, 24, 14044. https://doi.org/10.3390/ijms241814044

AMA Style

Puranik N, Yadav D, Song M. Advancements in the Application of Nanomedicine in Alzheimer’s Disease: A Therapeutic Perspective. International Journal of Molecular Sciences. 2023; 24(18):14044. https://doi.org/10.3390/ijms241814044

Chicago/Turabian Style

Puranik, Nidhi, Dhananjay Yadav, and Minseok Song. 2023. "Advancements in the Application of Nanomedicine in Alzheimer’s Disease: A Therapeutic Perspective" International Journal of Molecular Sciences 24, no. 18: 14044. https://doi.org/10.3390/ijms241814044

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop