Next Article in Journal
Regulatory Effect of Ficus carica Latex on Cell Cycle Progression in Human Papillomavirus-Positive Cervical Cancer Cell Lines: Insights from Gene Expression Analysis
Next Article in Special Issue
Combination Treatment with Liposomal Doxorubicin and Inductive Moderate Hyperthermia for Sarcoma Saos-2 Cells
Previous Article in Journal
The Impact of Sex on the Response to Proton Pump Inhibitor Treatment
Previous Article in Special Issue
Molecular Resonance Imaging of the CAIX Expression in Mouse Mammary Adenocarcinoma Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Single and Multitarget Systems for Drug Delivery and Detection: Up-to-Date Strategies for Brain Disorders

by
Clara Grosso
1,*,
Aurora Silva
1,2,
Cristina Delerue-Matos
1 and
Maria Fátima Barroso
1
1
REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida 431, 4249-015 Porto, Portugal
2
Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, Ourense Campus, Universidad de Vigo, E-32004 Ourense, Spain
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2023, 16(12), 1721; https://doi.org/10.3390/ph16121721
Submission received: 31 October 2023 / Revised: 1 December 2023 / Accepted: 7 December 2023 / Published: 12 December 2023
(This article belongs to the Special Issue Molecular Systems for the Delivery of Drugs and Contrast Agents)

Abstract

:
This review summarizes the recent findings on the development of different types of single and multitarget nanoparticles for disease detection and drug delivery to the brain, focusing on promising active principles encapsulated and nanoparticle surface modification and functionalization. Functionalized nanoparticles have emerged as promising tools for the diagnosis and treatment of brain disorders, offering a novel approach to addressing complex neurological challenges. They can act as drug delivery vehicles, transporting one or multiple therapeutic agents across the blood–brain barrier and precisely releasing them at the site of action. In diagnostics, functionalized nanoparticles can serve as highly sensitive contrast agents for imaging techniques such as magnetic resonance imaging and computed tomography scans. By attaching targeting ligands to the nanoparticles, they can selectively accumulate in the affected areas of the brain, enhancing the accuracy of disease detection. This enables early diagnosis and monitoring of conditions like Alzheimer’s or Parkinson’s diseases. While the field is still evolving, functionalized nanoparticles represent a promising path for advancing our ability to diagnose and treat brain disorders with greater precision, reduced invasiveness, and improved therapeutic outcomes.

Graphical Abstract

1. Introduction

The prevalence of neurodegenerative and neuropsychiatric disorders is increasing dramatically. According to the World Health Organization (WHO), in 2023, more than 55 million people have dementia worldwide, with Alzheimer’s disease (AD) being the most common form (60 to 70% of cases) [1], while the data from 2019 indicate that the global prevalence of Parkinson’s disease (PD) was 8.5 million individuals [2]. The pathogenesis of AD is a complex process that remains unclear. The aggregation of beta-amyloid (Aβ) to form senile plaques and oligomers of Aβ, aggregation of tau to form neurofibrillary tangles (NFTs), change in acetylcholine levels, oxidative stress, and neuroinflammation are the main hallmarks in AD. However, AD is still managed with symptomatic treatments consisting of acetylcholinesterase inhibitors (AChEIs) (donepezil, rivastigmine, and galantamine) and/or N-methyl-D-aspartate (NMDA) receptor antagonists (memantine). Although being a topic of debate, Aducanumab, an Aβ-directed monoclonal antibody, was granted accelerated approval by the US Food and Drug Administration (FDA) in 2021 as the first disease-modifying therapy for AD. Other mechanisms and pathways of interest are being explored to develop new therapies, including focusing in other neurotransmitters, metabolism, the neuroimmune system, the microbiome, and the activity of a variety of nutraceuticals [3]. PD is characterized by the presence of Lewy bodies (LBs) and Lewy neurites (LNs), which are intraneuronal inclusion bodies observed in postmortem histopathological examinations of the brains of PD patients. The prodromal phase of PD is marked by a variety of non-motor symptoms that can precede the emergence of classical motor symptoms by several years. These early symptoms encompass issues such as constipation, gastric motility problems, sleep disruptions, depression, anxiety disorder, and cognitive impairments. As the disease progresses to stages 3 and 4, it affects key brain regions like the substantia nigra, thalamus, and amygdala, and the characteristic motor symptoms become clinically apparent. At stages 5 and 6, the pathology extends to sensory and motor areas in the neocortex, culminating in the most pronounced manifestation of the disease. In advanced stages of PD, patients may experience more severe motor symptoms, including instability, an increased risk of falls, difficulty swallowing (dysphagia), and non-motor symptoms like dementia and psychosis [3]. Currently, dopaminergic drugs are used to control the progression of the disease [4].
Exploring novel neuroprotective drugs with different mechanisms of action from those already used, combining drugs aiming at a multitarget approach, or finding new delivery routes to deliver the currently available drugs more efficiently to the brain are three possible ways to meet the requirements of the patients. In spite of new or combined mechanisms of action, multitarget therapy can offer multiple advantages, being more efficient than the conventional single-target treatment approaches. This can be achieved either using “cocktail drug–multiple targets” or “one compound–multiple targets” approaches [5]. For instance, combining α-tocopherol (an antioxidant) with donepezil (a cholinesterase inhibitor) showed promising results in an animal model of AD [6], as well as the co-delivery of BACE1 antisense sh-RNA and D-peptide, which induced both a reduction in Aβ plaque deposition and the inhibition of p-tau-related fibril formation in double transgenic (Tg) mice ((APPswe, PSEN1dE9)85Dbo/Mmjax), respectively [7]. Moreover, it has been shown that neuronal survival and repair processes were promoted by the combinatory effect of curcumin and docosahexaenoic acid [8]. Glinz et al. conducted a systematic review and meta-analysis of nine randomized controlled trials (2604 patients) comparing the clinical efficacy and safety of a combination therapy of AChEIsand memantine to monotherapy with either substance in patients with moderate to severe AD. The authors concluded that combined therapy had a significantly greater effect on cognition than AChEI monotherapy [9]. The benefits of combined therapy for AD were also highlighted by Kabir et al. [10]. Hybrid compounds have been designed for AD treatment by incorporating multiple known pharmacophores. For instance, compounds with dual-acting effects such as AChEI and butyrylcholinesterase inhibitor (BuChEI); AChEI and β-secretase (BACE 1) inhibitor; AChEI and glycogen synthase kinase 3 beta (GSK-3β) inhibitor; AChEI and NMDA antagonist; AChEI and serotonin receptor (5-HT4R) partial agonist; AChEI and serotonin transporter (SERT) inhibitor; ChEI and antioxidant; and AChEI and MAO-B inhibitor, among others, have been developed [5,11]. NLX-112, a non-dopaminergic drug (a highly selective 5-HT1A receptor agonist), is a dual-effect drug, acting both on dyskinesia and the movement symptoms of PD [12].
Concerning neuropsychiatric disorders, depression and anxiety are the most common neuropsychiatric disorders, affecting approximately 280 [13] and 301 million people [14], respectively. Progress in discovering and developing new and improved psychiatric drugs has been slow and disappointing. The vast majority of currently prescribed drugs are arguably no more effective than the first generation of psychiatric drugs introduced in the 1950s and 1960s [15]. The etiology of depression (major depressive disorder) and anxiety are still not clear, but it is generally believed that they are multifactorial diseases caused by the interaction of social, psychological, and biological aspects. The symptoms include avoiding contact with friends; neglecting hobbies and interests; having difficulties in home, work, or family life; continuous low mood or sadness; feeling hopeless and helpless; having low self-esteem; feeling irritable and intolerant of others; having no motivation or interest in things; finding it hard to make decisions; moving or speaking more slowly than usual; changes in appetite or weight; unexplained aches and pains; and disturbed sleep. Despite several monoaminergic drugs being available on the market, 1/3 of depressive patients are unresponsive to this kind of treatment, Spravato® (S-ketamine, an NMDA receptor antagonist) being their only hope [16,17]. However, concerns such as the transient adverse effects and potential misuse liability of Spravato® (Janssen: Pharmaceuticals, Inc., Titusville, NJ, USA) have limited its widespread use [18]. Henssler et al. [19] performed a meta-analysis of 39 trials comprising 6751 patients and found that combined antidepressant therapy using a reuptake inhibitor with an antagonist of presynaptic α2-autoreceptors (mianserin, mirtazapine, trazodone) was associated with significantly superior treatment outcomes compared with antidepressant monotherapy.
Symptoms of anxiety include feeling restless; being easily fatigued or irritable; having difficulty in concentrating; having unexplained pains (e.g., headaches, muscle aches, stomachaches); having difficulty in controlling feelings of worry; and having sleep problems. The first-line treatment for anxiety is based on selective serotonin reuptake inhibitors (SSRIs), serotonin–norepinephrine reuptake inhibitors (SNRIs), and benzodiazepines that target the gamma-amino butyric acid (GABA) receptor. However, the drugs currently in clinical development for the treatment of anxiety disorder include other mechanisms of action, namely NMDA receptor antagonism, negative allosteric modulation of metabotropic glutamate receptors 1–5, sodium channel inhibition, vasopressin receptor 1A antagonism/agonism, antagonism of orexin receptors 1,2, and agonism of neuropeptide Y receptors 1,2,5 [20,21].
Other important aspect deals with the delivery route of drugs to the brain. Developing an efficient therapeutic system for brain disorders faces a significant challenge due to the presence of the blood–brain barrier (BBB), a barrier that separates the blood and the brain interstitial fluid and obstructs the passage of drugs, peptides, and large molecules into the brain [22]. Several strategies have been developed to increase the passage of the drugs through the BBB. Thus, this review paper provides a comprehensive analysis of the latest developments in nanoparticle-based drug delivery and diagnostics for brain disorders, encompassing neurodegenerative (AD and PD) and neuropsychiatric conditions (depression and anxiety). An array of nanoparticles, including lipid nanoparticles (liposomes, exosomes, solid lipid nanoparticles, and nanoemulsions), polymeric nanoparticles (PLGA, chitosan, alginate-based polymeric nanoparticles, and cyclodextrins), as well as magnetic nanoparticles, have emerged as promising tools in the quest to enhance the therapeutic and diagnostic outcomes for these challenging diseases. Strategies to functionalize these nanoparticles will also be discussed in the next sections (Figure 1).

2. Materials and Methods

This review underscores the significance of nanoparticle-based drug delivery and diagnostic platforms in addressing the complexities of brain disorders. We offer a detailed and up-to-date survey of these developments published between 2010 and 2023, with a focus on their design, applications, and potential for improving the management of brain disorders. The keywords selected for the database search included: “liposome”, “exosome”, ”solid lipid nanoparticle”, “nanoemulsion”, “polymeric nanoparticles”, “PLGA”, “Chitosan”, “cyclodextrins”, “nanoparticle functionalization”, “covalent bonding”, “non-covalent bonding”, “magnetic nanoparticles”, “depression”, “major depressive disorder”, “anxiety”, Alzheimer’s disease”, “Parkinson’s disease”, “blood–brain barrier” “cytotoxicity”, and a combination of these terms. Thus, 93 studies were selected based on the following criteria: (1) the abstract and entire text available; (2) documents written in the English language; (3) contained information regarding nanoparticle composition, physicochemical characteristics, and pharmacological effects.

3. Blood–Brain Barrier and Delivery Routes

The BBB’s intricate structure comprises brain capillary endothelial cells, pericytes, perivascular mast cells, basement membranes, astrocytes, and neuronal cells, collectively responsible for regulating the exchange of molecules between the bloodstream and the brain. Of particular importance are the brain capillary endothelial cells, which play a crucial role in safeguarding the brain via the formation of tight junctions. These tight junctions serve as a primary defense mechanism by forming a high-resistance paracellular barrier limiting the crossing of molecules, ions, harmful toxins, and pathogens into the brain [22]. Transcellular transport, on the other hand, is regulated by specialized transporters, pumps, and receptors. Efflux transport systems, metabolizing enzymes, and multiple membrane transporters and receptors exist in the brain capillary endothelial cells, reinforcing the barrier properties of the BBB. They are responsible for removing undesirable substances from the brain into the systemic circulation [23,24]. Examples of these systems include multidrug resistance transporters, monocarboxylate transporters, and organic anion transporters [23]. Figure 2 exemplifies the five main transport mechanisms to transport drugs or nanocarriers across the BBB.
Passive transport occurs via transmembrane transport (transcellular) or tight junctions (paracellular). Only small lipophilic molecules, with a molecular weight < 500 Da and less than eight hydrogen bonds, or small gas molecules (such as CO2 or O2) can escape the P-glycoprotein (P-gp)-type multidrug resistance efflux pumps and freely diffuse through the BBB via transmembrane diffusion. However, the transport of most nutrients requires specialized transporters (carriers), receptors, or vesicular processes (such as adsorptive transcytosis). Carrier-mediated transport includes the use of large neutral amino acid transporters (LAT1) for the transport of amino acids, nucleosides, and certain drugs, and of the glucose transporter (GLUT1) for glucose transport, while several receptors are involved in receptor-mediated transport, such as transferrin receptor (TfR), insulin receptor (INSR), insulin-like growth factor-1 receptor (IGF1R), and leptin receptor (LEPR), among others. Positively charged molecules (e.g., polymers, cationic lipids, albumin, and nanoparticles) undergo adsorptive endocytosis, due to interactions with the negatively charged cell membrane. In paracellular transport, small water-soluble molecules pass through small intercellular pores in tight junctions, possibly involving claudins [22,23,25,26].
Different strategies have been suggested to deliver drugs to the brain: (a) drugs can be delivered to the brain via local delivery (injection via a catheter). Biodegradable polymer implants can also be used for sustained release of drugs. These procedures require surgery, are highly invasive, and are mostly used to treat glioblastomas or other brain tumors; (b) drugs can be delivered via the intranasal route. After reaching the nasal cavity, a drug loaded inside nanocarriers can be transported along the olfactory bulb (olfactory pathway) and the trigeminal nerve (trigeminal pathway) directly to the central nervous system (CNS), circumventing the BBB. Although an extremely attractive option, the intranasal route has drawbacks such as a high variability of the delivered dose depending on the state of the nasal mucosa; (c) the most popular delivery route remains the systemic pathway, in which drugs have to cross the BBB; (d) finally, an innovative way to solve the permeation problem with the BBB is to load drugs inside nanoparticles [27]. Within this context, the surface functionalization of nanoparticles is commonly employed as a strategy to enhance the drug’s ability to traverse the BBB, as well as to reduce toxicity and to increase nanoparticles stability in biological fluids [28]. Different methods for nanoparticle surface modification have been employed, namely chemical ligand exchange (which involves replacing existing surface ligands on nanoparticles with new ones) [29,30,31], covalent bonding (based on the attachment of ligands directly to the nanoparticle surface by sharing electrons) [32], and adsorption or non-covalent bonding (involving adherence of molecules onto the nanoparticle surface via weak forces, for instance, van der Waals, electrostatic or hydrogen bonding) [33,34]. Non-covalent bonding has the advantage of being relatively simple; however, covalent bonding is more robust and can combat detachment irrespective of the process used [28,35].
Among the most common ligands used to functionalize nanoparticle’s surface are antibodies (e.g., the single chain fragment of variable region 46.1 (scFv46.1) [36]; the OX26 monoclonal antibody to the rat transferrin receptor; and monoclonal antibodies to the insulin receptor [37]) RNA aptamers [38], transferrin (targeting the transferrin receptor at the BBB), lactoferrin (targeting the lactoferrin receptor at the BBB), glucose (targeting glucose transporters at the BBB), apolipoprotein E (ApoE) (targeting the low-density lipoprotein receptor at the BBB), glutathione (targeting the Na+-dependent GSH transporter at the BBB), and angiopep-2 (which has good transcytosis ability across the BBB) [22].
Nanoparticles can also be modified with polyethylene glycol (PEG), which acts as a shield to protect nanoparticles from plasma protein binding and prevents opsonization activity and phagocytosis [39]. This is extremely important since uncoated nanoparticles are recognized as foreign agentsbeing readily cleared from systemic circulation by the cells of the mononuclear phagocyte system (MPS), barring accumulation in target cells and tissues. The MPS is composed of dendritic cells, blood monocytes, granulocytes, and tissue-resident macrophages in the liver, spleen, and lymph nodes that are responsible for clearing, processing, and degrading exogenous agents in the bloodstream [40].
Dual- and multitarget nanoparticles represent advanced nanoplatforms that go beyond the conventional approach by incorporating not only the core scaffold but also two or more functionalizing ligands. This innovative design facilitates the precise delivery of therapeutic agents to their intended destinations [41]. For instance, Arora et al. [42] demonstrated an efficient brain-targeted delivery of ApoE2-encoding plasmid DNA (pApoE2) using liposomes. The liposomes were functionalized with a glucose transporter-1 targeting ligand mannose (MAN) and a cell-penetrating peptide, rabies virus glycoprotein peptide (RVG29), or penetratin (Pen). Dual (RVGMAN and PenMAN)-functionalized liposomes were developed, taking advantage of the high density of glucose transporter-1 in the luminal side of the BBB and that cell-penetrating peptides have the ability to cross cell membrane bilayers, enhancing the transport of different cargo across the BBB [42,43]. Indeed, experiments with an in vitro BBB model (coculture of primary astrocytes and bEnd.3 cells) showed that the dual-modified liposomes promoted ∼2 times more protein expression than other formulations (monofunctionalized and unmodified liposomes) in neurons cultured below the in vitro BBB model. Neves et al. [44] encapsulated curcumin in solid lipid nanoparticles and observed that functionalization with transferrin produced a 1.5-fold increase in the permeability across hCMEC/D3 cells compared with the non-functionalized nanoparticles. Similarly, resveratrol-loaded solid lipid nanoparticles functionalized with ApoE also crossed the endothelial monolayer more efficiently (1.8-fold higher) than the non-functionalized nanoparticles [45]. ApoE-modified solid lipid nanoparticles were found to be internalized by clathrin-mediated endocytosis [46]. Thiazolidinedione-loaded PLGA nanoparticles decorated with a monoclonal anti-transferrin receptor antibody (8D3 mAb) were synthetized by Monge et al. [47]. The authors also produced three other nanoparticles—bare PLGA nanoparticles, 8D3-PLGA nanoparticles, and PLGA–thiazolidinedione—and incubated them with human serum to simulate bloodstream conditions. LC-MS/MS analysis revealed different protein corona compositions among functionalized and non-functionalized nanoparticles, with the 8D3-PLGA-thiazolidinedione and 8D3-PLGA nanoparticles being the only types of nanoparticles containing afamin and with ApoE and ApoA-I adsorbed into their protein corona. Afamin may play an important role in regulating vitamin E uptake, favoring its transport across the BBB. Likewise, apolipoproteins are involved in promoting the transport of drugs across the BBB [47].

4. Types of Nanoparticles

4.1. Lipid-Based Nanoparticles

Liposomes are the most explored nanocarriers used in drug delivery due to their biocompatibility, stability, ease to synthesize, high drug loading efficiency, and high bioavailability. Due to their combined hydrophobic and hydrophilic characteristics, both polar and apolar drugs can be encapsulated in the aqueous interior core or in the lipophilic membrane, respectively. They are spherical phospholipid-based vesicles (with diameters between 50 and 500 nm) composed of one or more lipid bilayers, being classified as small unilamellar vesicles (SUV), large unilamellar vesicles (LUV), multilamellar vesicle (MLV), and multivesicular vesicles (MVV). To enhance the stability of the liposomal structure and prevent the leakage of its inner cargo, cholesterol or sphingomyelin can be introduced into the lipid mixture. These additives effectively regulate the membrane permeability, alter fluidity, and enhance the robustness of bilayer membranes [48,49]. The phospholipids frequently used in liposome composition include natural lipids such as phosphatidylcholine, sphingomyelin, and lecithin or synthetic lipids like 1,2-dipalmitoyl-sn-glycero-3-phosphocholine and ethyl-phosphatidylcholine [50].
Several liposome formulations have been developed to incorporate neuroprotective drugs, aiming at treating neurodegenerative and neuropsychiatric disorders (Table 1). Moreover, liposomes are also used for diagnostic purposes. Congo red can bind with amyloid plaques, and liposomes modified with Congo red have been patented for AD diagnosis [49].
Exosomes are a class of extracellular biovesicles of endocytic origin, which are constitutively or inductively released by virtually all cell types and are accessible in almost all body fluids, such as saliva, urine, plasma, cerebrospinal fluid, amniotic fluid, and semen [51]. These nanostructures originate from budding processes occurring upon the fusion of multivesicular bodies and the plasma membrane and are involved in many biological functions, especially communication and transfer of biological information between cells [51]. They are involved in synaptic plasticity, neuroinflammation, neurotoxicity, neurotransmission, neuronal stress, neurodevelopment, neurogenesis, and neurodegeneration in the brain [52].
Table 1. Examples of drug loaded into liposomes for neurodegenerative and neuropsychiatric disorder treatment.
Table 1. Examples of drug loaded into liposomes for neurodegenerative and neuropsychiatric disorder treatment.
Target/DiseaseDrug(s)Nanoparticle Type CompositionFunctionalizationDiameter of the Optimized Formulation(s) (nm)ObservationsRef.
ADAnti-amyloid single-domain antibody fragment (VHH-pa2H)Liposome1,2-dimyristoyl-sn-glycero-3-phosphocholine or egg-yolk phosphatidylcholineGlutathione-DSPE-PEG2000 (PEG)108, 110Liposomes were injected into APPswe/PS1dE9 double transgenic mice, a mouse model of AD. GSH-PEG liposomes increased VHH-pa2H uptake in the brain. [53]
ADApoE2-encoding plasmid DNA/chitosanLiposome Dioleoyl-3-trimethylammonium-propane chloride (DOTAP), dioleoyl-sn-glycero-3phosphoethanolamine (DOPE)DSPE-PEG2000, Glucose transporter-1 (Glut-1) targeting ligand mannose (MAN)+ cell penetrating peptides (CPP) (rabies virus glycoprotein peptide (RVG) or penetratin (Pen))168, 172 Significantly higher expression of ApoE2 in bEnd.3 cells, primary neurons, and astrocytes compared to monofunctionalized and unmodified liposomes. Dual-modified liposomes (RVG-MAN and Pen-MAN) also showed ∼2 times higher protein expression than single-targeted formulation (MAN or Pen) in neurons cultured below an in vitro BBB model.[42]
ADRivastigmineLiposomeCholesterol and dipalmitoylphosphatidyl choline (DPPC) methylcellulose dimethyl-β-cyclodextrin or sodium taurocholateAbsorption enhancers
(dimethyl-β-cyclodextrin or sodium taurocholate)
3.4 *, 4.8 * Rivastigmine liposomes and solutions were also administered to mice orally and intraperitoneally. The highest AChE inhibition was observed for rivastigmine-sodium-taurocholate liposomes.[54]
ADRivastigmineLiposomeEgg phosphatidylcholine, cholesterolDSPE-PEG2000-CPP179Intranasal administration of rivastigmine liposomes to rats demonstrated the capacity to improve rivastigmine distribution and adequate retention in CNS regions (hippocampus and cortex) and affected acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) activities. The effects were more pronounced that those induced by free rivastigmine and uncoated liposomes.[55]
ADRivastigmineLiposome1,2-diacyl-sn-glycero-3-phosphocholine, dihexadecyl phosphate, cholesterol-68, 529Liposomes resulted in faster memory regain and the amelioration of metabolic disturbances in AlCl3-treated rats than free rivastigmine solution.[56]
ADα-tocopherol + donepezil hydrochlorideLiposomeL-α-phosphatidylcholine and cholesterolTetradecyltriphenylphosphonium bromide105–115Intranasal administration to APP/PS1 mice resulted in enhanced learning abilities and a reduction in the formation rate of amyloid beta (Aβ) plaques in the entorhinal cortex and hippocampus of the brain.[6]
ADGalantamine hydrobromideLiposomeSoya phosphatidylcholine and cholesterol-112Intranasal administration of the liposomes could readily transport galantamine into rat brain tissues where it could inhibit cholinesterase.[57]
ADQuercetinLiposomeEgg L-α-phosphatidylcholine, cholesterol--Effect of nasal administration of quercetin liposomes on neurodegeneration in an animal model of AD. Quercetin liposomes attenuated the degeneration of neurons and cholinergic neurons in the hippocampus, promoted the elevation of superoxide dismutase, catalase, and glutathione peroxidase activities and induced the reduction in malondialdehyde in the hippocampus.[58]
PDLevodopaLiposomeHydrogenated soybean phosphatidylcholine, cholesterolDSPE-PEG2000-Chlorotoxin107After intraperitoneal injection to mice, liposomes loaded with levodopa significantly increased the distribution of dopamine (DA) and dihydroxyphenyl acetic acid in the substantia nigra and striata. In a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice model, levodopa-loaded chlorotoxin liposomes significantly attenuated the serious behavioral disorders and diminished the MPTP-induced loss of tyrosine hydroxylase-positive dopaminergic neurons.[59]
PDGlial-cell-derived neurotrophic factor (GDNF)LiposomeDioleoylphosphatidylcholine (DOPC), cholesterol, and stearylamine-149Rats were nasally administered with GDNF solution or with cationic liposomes containing GDNF before the injection of 6-hydroxydopamine (6-OHDA). Both intranasal GDNF treatments induced a neurotrophic effect in the substantia nigra since the number of tyrosine hydroxylase (TH)-positive neurons was significantly higher than in controls given intranasal PBS liposomes.[60]
PDDopamineLiposomeCholesterol, 1,2-dioleyl-sn-glycero-3-phosphocholine (DOPC), L-α-phosphatidic acidAPP-derived peptide100 Intraperitoneal injection of the APP-targeted liposomes loaded with dopamine resulted in a significant increase in striatal DA in amphetamine-treated mice.[61]
PDResveratrolLiposomeSoybean lecithin
and cholesterol
-146–585Resveratrol liposomes could significantly enhance the activity of mitochondrial electron transfer chain complex I in the substantia nigra cells of 6-hydroxydopamine-treated rats, promote the expression of complex I subcomponent MT-ND1-37kD, improve mitochondrial membrane potential, inhibit the release of mitochondrial cytochrome C and apoptotic inducible factor, enhance the expression of mitochondrial functional protein PINK1, increase the phosphorylated TRAP1 level, and elevate the phosphorylated TRAP1/TRAP1 levels.[62]
PDPolyphenol-rich grape pomace extractsLiposomeBrain lipids, 1,2-distearoylsn-glycero-3-phosphoethanolamine-N-(PEG)5000anti-transferrin receptor antibody133The antioxidant nanoplatform was successfully tested in a rotenone-induced in vitro PD model, where it completely regulated the reactive oxygen species (ROS) levels, prevented the aggregation of α-synuclein fibrils, and restored cell viability.[63]
AnxietyNimodipineLiposomeSoybean phosphatidylcholine
and cholesterol
-107The results suggest that the administration of nimodipine liposomes has no sedative or muscle relaxant effect in animals but displayed anxiolytic-like activity in bright-field test. The results obtained in the open arms test suggest that the new formulation acts on benzodiazepine receptors.[64]
AnxietyEugenolLiposomeLecithin, cholesterol-91The mRNA expression of glyoxylase-1 (GLO-1) and GLO-1 protein expression were measured in 42 BALB/c mice submitted to stress using a conventional restraint model. The mRNA and protein expressions were found to be increased in animals given anxiety as compared to the normal control. Eugenol and its liposome-based nanocarriers counteracted this behavior, with liposomal eugenol behaving better than the compound alone.[65]
AnxietyEugenolLiposomeLecithin, cholesterol-91Stress was induced in 42 BALB/c mice using a conventional restraint model. The mRNA expression of neurokinin 1 receptor (NK1R) and the NK1R protein expression were increased in animals given anxiety as compared to the normal control. Both parameters decreased in animals treated with eugenol and its liposome-based nanocarriers, the results being better for nanocarriers.[66]
Anxiety/DepressionQuercetinLiposomeEgg L-α phosphatidylcholine, cholesterol --Wistar rats were intranasally administered quercetin liposomes. These nanocarriers possessed anxiolytic and anti-depression like activity and a cognitive-enhancing effect, which were assessed using the elevated plus maze test, forced swimming test, and Morris water maze test[67]
DepressionPlasmid-harboring brain-derived neurotrophic factor (BDNF)Liposome1,2-Dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), and 1,2-dioleoyl-3-trimethylammonium-propane chloride (DOTAP), cholesterolDSPE-PEG2000, transferrin, and arginine125Physicochemical characterization of the nanoparticles produced.[68]
DepressionNimodipineLiposomeSoybean phosphatidylcholine
and cholesterol
-107Tail suspension test, forced swimming test, and MAO-B activity assay suggested that nimodipine-liposomes displayed more antidepressant activity than imipramine and paroxetine but with a lower effect than that observed in the group receiving liposomes + reserpine.[69]
DepressionSertraline hydrochlorideLiposomeHydrogenated soya phosphatidylcholine-L-α-phosphatidylcholine (HSPC), distearoyl phosphatidyl glycerol sodium (DSPG) and cholesterol-152The use of liposomes increased drug transport to the brain compared with administration with free sertraline [70]
DepressionPiperineLiposomeEgg L-α-phosphatidylcholine, cholesterolPolyethylene glycol 1000100Piperine-encapsulated liposomes displayed antidepressant-like activity in Wistar rats submitted to forced swimming, Morris water maze, and spontaneous motor behavior tests. [71]
* μm. Alzheimer’s disease (AD), amyloid beta (Aβ), amyloid precursor protein (APP), mouse model of Alzheimer’s disease (APP/PS1), acetylcholinesterase (AChE), beta-secretase (BACE1), isogenic mice (BALB), blood–brain barrier (BBB), bovine serum albumin (BSA), brain-derived neurotrophic factor (BDNF), butyrylcholinesterase (BuChE), cell-penetrating peptide (CPP), dopamine (DA), dioleoyl-3-trimethylammonium-propane chloride (DOTAP), dioleoyl phosphatidylcholine or 1,2-dioleyl-sn-glycero-3-phosphocholine (DOPC), L-α-phosphatidic acid (DOPC), central nervous system (CNS), dioleoyl-3-trimethylammonium-propane chloride (DOTAP), dioleoyl-sn-glycero-3phosphoethanolamine (DOPE), dipalmitoyl phosphatidyl choline (DPPC), 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)-2000] (ammonium salt) (DSPE-PEG2000), epigallocatechin-3-O-gallate (EGCG), glial cell derived neurotrophic factor (GDNF), glutathione PEGylated liposomes (GSH-PEG), glyoxylase-1(GLO-1), 6-hydroxydopamine (6-OHDA), glucose transporter-1 targeting ligand mannose (MAN), monoamine oxidase (MAO), 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), mitochondrially encoded NADH dehydrogenase (MT-ND1), neurofibrillary tangles (NFTs), neurokinin 1 receptor (NK1R), Parkinson’s disease (PD), penetratin (Pen), poly ethylene glycol (PEG), PTEN-induced kinase-1 (PINK1), poly(lactic-co-glycolic acid) (PLGA), rabies virus glycoprotein peptide (RVG), reactive oxygen species (ROS), necrosis factor receptor-associated protein (TRAP), anti-amyloid single-domain antibody fragment (VHH-pa2H).
Exosomes are characterized by a single lipid bilayer membrane, containing phospholipids such as phosphatidylcholine, phosphatidylserine, phosphatidylethanolamine, phosphatidylinositol, phosphatidic acid, as well as cholesterol, ceramides, sphingomyelin, and glycine [51]. They also carry cell-specific cargo of proteins, lipids, and genetic materials (RNA and DNA), and can be selectively taken up by neighboring or remote cells, reprogramming the recipient cells [51,72,73].
Exosomes from various sources have revealed a wide array of constituent molecules, comprising approximately 4400 proteins, 194 lipids, 1639 mRNAs, and 764 miRNAs. These findings underscore the intricate nature of exosomes and hint at their potential for diverse functional roles [51,72]. Typically, exosomes are highly enriched in proteins with various functions, such as tetraspanins, which take part in cell penetration, invasion, and fusion events; heat shock proteins, as part of the stress response, which are involved in antigen binding and presentation; multivesicular body formation proteins that are involved in exosome release; and proteins responsible for membrane transport and fusion; metabolic enzymes; growth factors; and cytokines [51,72] Spherical and with diameters in the range of 40 to 150 nm, exosomes have garnered significant attention in targeted drug delivery research, primarily owing to their persistent presence within the circulatory system and their remarkable ability to shield their cargo from degradation by various proteases and nucleases. Compared to liposomes, exosomes have higher penetration, prolonged blood circulation, biocompatibility, and enhanced biodistribution [73].
Besides being a promising therapeutic vehicle, exosomes can also act as valuable biomarkers in diagnosing brain diseases. Within the CNS, exosomes play pivotal roles in various physiological processes, including intercellular communication, the maintenance of myelination, synaptic plasticity, antigen presentation, and providing trophic support to neurons. Nonetheless, in pathological circumstances, especially in conditions marked by abnormal protein accumulation, exosomes are increasingly involved in the removal of accumulated, undesirable biomolecules. Specifically, the disposal of unwanted cellular components via exosomes becomes a crucial mechanism when other cellular clearance systems, such as the proteasome and autophagy–lysosome system, gradually fail to effectively eliminate the aggregated proteins. In such situations, exosomes originating from the CNS have been observed to participate in cell-to-cell spreading of amyloidogenic proteins, namely Aβ, tau, and α-synuclein [74]. Aβ42, pT181-tau, pS396-tau, total-tau, and α-synuclein are some of the potential fluid biomarkers for the diagnosis of neurodegenerative diseases [74]. Moreover, there are distinct profiles of exosomal miRNAs even before the onset of irreversible neurological damage, indicating that exosomal miRNAs within tissues and biological fluids could serve as better biomarkers than aggregated proteins. Even more importantly, exosomes are highly enriched with miRNAs, compared to their parental cells and cell-free blood, making them of choice par excellence for biomarker profiling [75].
Solid lipid nanoparticles (SLN) are lipid-based biocompatible nanocarrier systems composed by lipids or modified lipids (e.g., triglycerides, fatty acids, or waxes) with a size diameter in the range of 10–1000 nm. These nanoparticles have a solid hydrophobic lipid core, in which both hydrophilic and lipophilic drugs can be dispersed. Compared to liposomes and polymeric nanoparticles, SLNs safeguard better the encapsulated drug from biochemical degradation due to the solid lipid nature of their core instead of the water-soluble core of other types of nanoparticles [76].
Examples of drug-loaded exosomes and SLNs containing bioactive compounds for brain disorder treatments are depicted in Table 2 and Table 3, respectively.
Nanoemulsions are colloidal biphasic dispersions systems consisting of two immiscible liquids: one is the dispersed phase and the other is a continuous phase that is stabilized using appropriate emulsifying agents (e.g., surfactant and co-surfactant). They can be of water-in-oil (w/o) or oil-in-water (o/w) types and their droplet or globular dimension typically varies from 200 nm to 100 µm. Nanoemulsions offer a wide range of applications since they are well absorbed through the mucosal layer and are hence suitable for site-specific drug delivery via different routes, namely the oral, parental, nasal, and ocular routes [94,95]. The higher encapsulation of lipophilic drugs in o/w-type nanoemulsions offers greater solubility, better absorption, and higher bioavailability with minimum enzymatic degradation, being the preferable type of nanoemulsion to permeate the BBB [95].
Nanoemulsions normally contain 5–20% oil/lipid droplets in the case of o/w emulsions. However, this percentage can be higher (up to 70%) [96]. Concerning the lipidic phase, several oils are employed, namely cottonseed oil, coconut oil, sesame oil, safflower oil, rice bran oil, and soybean oil. The use of fatty acids such as linolenic acid, omega-6 fatty acids, oleic acid, and linolenic acids alone or in combination has shown benefits for the development of nanoemulsions to be delivered via the intranasal route [95,96]. Surfactants like phosphatidylcholine, bilesalt, chitosan, starch, Tweens (Tween 60, 80), sodium dodecyl sulphate, cetyl pyridinium chloride, labrasol, Span (Span 60, 80), poloxamers, casein, PEG-containing block polymers, and certain proteins and lipids are commonly used to reduce the interfacial tension between both phases of the biphasic system and also to increase their stability. In addition, chiral alcohols, Tween 80, PEG 600, glycerol, butan-1-ol, transcutol-P, and sorbitol are included in the formulations as co-surfactants [95,96]. Preservatives, antioxidants, and chemoprotectants are also included in nanoemulsion composition. Among them, benzoic acid, sorbic acid, propionic acid, and dehydro-acetic acid have antifungal properties, and ascorbic acid, citric acid, tocopherols, butylated hydroxytoluene (BHT), and butylated hydroxyanisole (BHA), among others, are antioxidant agents [96]. Table 4 resumes the application of drugs loaded into nanoemulsions for neurodegenerative and neuropsychiatric disorder treatment.

4.2. Polymeric Nanoparticles

Polymeric nanoparticles, ranging in size from 1 to 1000 nm, serve as versatile carriers for active compounds, which can either be encapsulated within or adsorbed onto their polymeric structure. Within this category, two distinct nanocarriers emerge, namely nanocapsules and nanospheres. Nanocapsules are composed of an oily core in which the drug is usually dissolved, enveloped by a polymeric shell responsible for regulating the controlled release of the drug from the core. Nanospheres are characterized by a continuous polymeric network that can either retain the drug within its matrix or adsorb it onto the nanoparticle’s surface [109].
Natural and synthetic polymers are usually used to prepare polymeric nanoparticles. Natural polymers include chitosan/chitosan derivatives, alginate, gelatin, and albumin, while polylactic acid (PLA), poly(lactic-co-glycolic acid) (PLGA), poly-ε-caprolactone (PLC), and poly(amido amine) are among the synthetic ones [110].
Another class of polymeric nanoparticles are cyclodextrins. Cyclodextrins (CDs) are a family of cyclic oligosaccharides produced from starch or starch-based materials by the bacterial enzyme cyclodextrin glycosyltransferase (CGT). CGTs from Bacillus species are the most extensively studied enzymes CDs, consisting of six (αCD), seven (βCD), eight (γCD), or even more glucopyranose monomers connected via α-1,4-glycosidic bonds. Cyclodextrins exhibit a unique structural geometry characterized by a truncated cone shape, featuring a hydrophilic outer surface and a more lipophilic interior cavity. Due to this configuration, cyclodextrins possess the remarkable property of being water-soluble while also being capable of accommodating hydrophobic molecules simultaneously. The inclusion of lipophilic substances into CDs enhances their stability and bioavailability while also increasing their aqueous solubility [111] The OH groups of CDs are also involved in the binding processes via electrostatic forces and Van der Waals and hydrogen bonding interactions [112]. There are several cyclodextrin-based drugs for brain disorders already on the market, for instance, Geodon® IM, Pfizer Inc., New York, NY, USA (Captisol®-enabled ziprasidone mesylate for schizophrenia and bipolar disorder), Abilify®, Bristol-Myers Squibb, Princeton, NY, USA (Captisol®-enabled aripiprazole for bipolar I disorder), and Zulresso®, Sage Therapeutics, Inc., Cambridge, MA, USA (Captisol®-enabled brexanolone for moderate to severe postpartum depression) [113].
Multiple ligands can be affixed to the surface of polymeric nanoparticles to aid in their transport through the BBB. There are four primary categories of ligands employed for the functionalization of polymeric nanoparticles. These include ligands that adsorb proteins from the bloodstream, enabling them to interact directly with receptors or transporters found on the endothelial layer of the BBB (e.g., Tween 80); ligands that directly bind to BBB receptors or transporters (e.g., antibodies designed for receptors like the transferrin receptor, insulin receptor, or glucose transporter); ligands that enhance the charge and hydrophobicity of the nanoparticles, such as amphiphilic peptides; and ligands that extend the circulation time of nanoparticles in the bloodstream, such as PEG [114].
Table 5 presents some examples of neuroprotective drugs encapsulated in polymeric nanoparticles.

4.3. Metallic Nanoparticles

Metallic nanoparticles for biomedical purposes include gold nanoparticles, magnetic nanoparticles, and silver nanoparticles. Materials such as pure metals (Fe, Co, and Ni), alloys (FeCo, Alnico, and Permalloy), and ferrites (Mn0.6Zn0.4Fe2O4 and CoFe2O4) with high saturation magnetizations are usually selected to produce magnetic nanoparticles for biomedical applications, namely, magnetic hyperthermia (for the focal treatment of tumors), contrast agents for magnetic resonance imaging (MRI), and targeted drug delivery systems. Because of the oxidative characteristics and high toxicity levels of pure metals, they are not suitable for in vivo use. Iron oxides (e.g., Fe3O4, γ-Fe2O3) are highly stable and biocompatible [138,139].
With exceptional spatial resolution and tissue contrast, MRI offers detailed anatomical images of soft tissues, establishing itself as a pivotal diagnostic modality for imaging the brain, cartilage, heart, and blood vessels, and tumor detection. In contrast to other imaging platforms like computerized axial tomography (CAT), positron emission tomography (PET), and single-photon emission computed tomography (SPECT), MRI techniques eliminate the need for radioactive agents and ionizing radiation. Despite its ability to generate detailed soft tissue images, MRI’s intrinsic low sensitivity poses challenges in distinguishing normal tissues from lesions. The introduction of contrast agents addresses this limitation by amplifying the contrast effect in regions of interest via the acceleration of magnetic relaxation [140]. Although more expensive than analogic MRI systems, modern digital MRI systems play a crucial role in acquiring, processing, and displaying the images produced during the scanning process. Moreover, digitalization has had a significant impact on healthcare in recent years, mainly in telemedicine. It can be principally useful for people in rural or underserved areas who may not have easy access to healthcare facilities [141].
Several authors have produced iron oxide nanoparticles (IONPs) for MRI purposes [142,143,144,145,146,147]. For instance, Tang et al. [147] have developed sulfated dextran-coated iron oxide nanoparticles for the imaging of activated microglia during brain inflammation. However, uncoated IONPs usually have some degree of toxicity, inducing, at high concentrations, increased generation of reactive oxygen species, reduction in cellular proliferation, or even induction of cell death [145]. As shown by Yu et al. [148], uncoated superparamagnetic iron oxide particles (SIONPs, 0.5 mg/mL) displayed up to a 6-fold increase in cytotoxicity compared to dextran-coated or PEG-coated iron oxide nanoparticles. Yang et al. [144] evaluated the potential reproductive toxicity of IONPs in ICR mice. The results showed that the nanoparticles could cause reversible damage to the reproductive system of male mice without affecting the main organs.
Various ligands specific to Aβ plaques have been found and utilized to functionalize or be encapsulated in nanoparticles to achieve early detection and treatment for AD. For instance, Cheng et al. [149] functionalized SIONPs loading curcumin, a natural compound that binds to Aβ plaques, and successfully visualized them in transgenic mouse brains using MRI. Near Aβ plaques, there is a deposition and accumulation of ferritin protein. Fe3O4-dextran coated magnetic nanoparticles were conjugated with anti-ferritin antibody to be developed as contrast agents to detect Aβ plaques. In both in vitro and in vivo tests (intravenous injection), the nanoparticles were able to recognize and bind specifically to the ferritin protein accumulated in the subiculum area of the AD transgenic mice [150].
Kim and Chang [151] investigated the therapeutic potential of human adipose-derived stem cells (hASCs) using magnetic nanoparticles in a 6-OHDA-induced PD mouse model. MRI showed hASC distribution in the substantia nigra of hASC-injected PD mice and in behavioral evaluations (apomorphine-induced rotation and Rota rod performance), hASCs significantly induced recovery in 6-OHDA-treated PD mice when compared with a saline-treated control group.
Magnetic, silver, and gold nanoparticles have also been tested for neurodegenerative and neuropsychiatric disorder treatment. For instance, Sivaji and Kannan developed gold nanoparticles conjugated with donepezil and functionalized them with polysorbate 80 and polyethylene glycol [152]. This nanocarrier was able to cross the zebrafish BBB and showed an increase in AChE inhibition activity of 30–38% in the zebrafish brain. Silver nanoparticles containing an extract from Mucuna pruriens seeds (rich in L-DOPA) significantly lowered catalepsy symptoms in mice, showing its potential for PD treatment [153]. Kim et al. [154] compared the neuroprotective effect of anthocyanins alone and anthocyanin-loaded PEG/gold nanoparticles in Aβ1-42-injected mouse and in in vitro models of AD. Both anthocyanins alone or conjugated reduced the Aβ1-42-induced neuroinflammatory and neuro-apoptotic markers via inhibiting the p-JNK/NF-κB/p-GSK3β pathway in both in vivo and in vitro AD models, although the conjugated form was more efficient.
Khadrawy et al. [155] evaluated the antidepressant effect of curcumin-coated iron oxide nanoparticles in a rat model of depression. Wistar male rats were intraperitoneally injected with reserpine to produce depressive symptoms. In depressed rats, the FST showed an increased immobilization time and a reduced swimming time, which were counterbalanced by curcumin-coated iron oxide nanoparticles. While depressed rats showed significantly decreased levels of serotonin, noradrenaline, DA, and glutathione and significantly increased levels of MDA, nitric oxide, glutathione S-transferases, monoamine oxidase A, AChE, Na+, K+, and ATPase activities in the cortex and hippocampus, treatment with nanoparticles for two weeks restored the levels of all these biomarkers to the control levels.

5. Comparison between Different Types of Nanoparticles for Brain Delivery

This review summarized the latest advances in the encapsulation of drugs to be delivered to the brain. Different types of nanocarriers were considered, including lipid-based, polymeric, and metallic nanoparticles. The advantages and disadvantages of each type of nanoparticle are discussed below.
Concerning lipid-based nanocarriers, solid lipid nanoparticles offer numerous advantages, such as facile manufacturing, enhanced pharmaceutical stability, increased drug content, efficient drug release, and prolonged stability. Solid lipid nanoparticles are effective in encapsulating poorly water-soluble compounds owing to their elevated lipid content. In contrast, although liposomes are the most widely used lipid-based encapsulation nanocarrier, they face challenges in achieving high drug loading for hydrophobic drugs due to their limited bilayer space [156]. Nanoemulsions, on the other hand, boast easy preparation without the necessity for organic solvents or cosolvents. Primarily derived from edible oils, they entail a low risk of toxicity [156]. van der Koog et al. [157] recently reviewed the advantages and limitations of exosomes and liposomes. According to the authors, both vesicle types have their own advantages. While liposomes offer extensive control over contents, exosomes have the advantage of inherent biocompatibility and a complex biological composition that will be hard to fully recapitulate in liposomes. Moreover, exosomes are of natural origin, while liposomes are of synthetic origin.
Akel et al. [158] compared the efficiency of PLGA nanoparticles and chitosan-modified solid lipid nanoparticles for the intranasal administration of insulin. Chitosan-coated solid lipid nanoparticles were the best nanocarriers since they increased the mucoadhesion, nasal diffusion, and drug release rate. Nonetheless, compared with solid lipid nanoparticles and liposomes, polymeric nanoparticles have some limitations, such as possible self-aggregation, which, when it occurs, can impact brain delivery [159].
Ross et al. [160] compared the stability of Au, Ag, Fe2O3, TiO2, and ZnO nanoparticles of 5, 20, and 50 nm in sterile-filtered water; cell culture media DMEM supplemented with 10% fetal bovine serum, 1% l-glutamine, 1% non-essential amino acids, and 1% penicillin-streptomycin; and human serum, to understand what would occur after intravenous delivery. The authors concluded that the gold and silver nanoparticles were the most stable, and, concerning the particle size, the 20 nm nanoparticles appeared to be the least stable size across materials.

6. Emerging Trends in Nanomedicine: Challenges and Future Directions

Managing and treating neurodegenerative and neuropsychiatric disorders present three main challenges. These challenges encompass: (1) the absence of advanced diagnostic tools for early disease detection, and in this sense, nanoparticles could be used for the construction of high-performance biosensors for detecting biomarkers at the early stages of diagnosis, either as amplifier labels for signal augmentation or as a practical option for biorecognition element immobilization. Moreover, nanoparticle-mediated clinical imaging technologies can provide observation of localized protein accumulation, playing a complementary role in CNS disease diagnosis; (2) the limitation of drugs in effectively crossing the BBB, necessitating the development of encapsulation strategies, as reviewed in the preceding sections; and (3) the deficiency in personalized disease management for timely decision-making in therapy [161,162]. The intersection between personalized medicine and nanomedicine occurs in both the diagnostic and therapeutic areas. At the diagnostic level, besides being used for exploring the status of specific drug targets, nanotechnology is also very useful for pharmacogenetic approaches and for in vitro and in vivo testing. In the therapeutic area, nanomedicine can tailor drugs based on individualized patient conditions [163].
Challenges related to safety, efficacy, and regulatory considerations pose significant hurdles in advancing personalized nanomedicine for CNS diseases toward clinical applications. More research needs to be conducted to gather more information about the pharmacokinetic profile of nanoparticles, tissue distribution, cytotoxicity, and side effects in order for regulatory agencies, like the FDA and EMA, to approve new nanoparticle-based therapies or diagnostic tools. Although some liposomes and metallic nanoparticles have gained regulatory approval for clinical use for cancer, for pain management, or as antifungal treatment [164], Sinerem® (GUERBET, Paris, France), an ultrasmall super paramagnetic iron oxide (USPIO) contrast agent for MRI, was withdrawn from the market in 2008 by the EMA due to concerns raised in clinical trials [164,165]. As far as what concerns CNS disorders, only a few clinical trials have yet been conducted. One single-center open-label pilot study (NCT03815916) was developed between 2019 and 2021 and tested the efficacy of gold nanocrystals (CNM-Au8) in PD, and a phase-II trial (NCT03806478) will evaluate (between June 2023 and December of 2024) the efficacy of the intranasal delivery of APH-1105 for the treatment of mild to moderate AD in adults [166].
Another important aspect is related to nanoparticle production. While it is easy to obtain stable nanoparticles at a small scale, difficulties with the scaling up and stability of large-scale manufacture have been widely experienced. Indeed, increasing the installation size from the laboratory scale to the industrial scale presents many difficulties, such as the maintenance of high reproducibility, homogeneity, and control over properties [167].
Altogether, nanotechnology emerges as an essential avenue for implementing a personalized medicine approach; however, it confronts challenges, including mainly safety concerns and scalability issues.

7. Conclusions

Significant research efforts have been dedicated to uncovering innovative strategies for delivering neuroprotective drugs to the brain. Circumventing the BBB to achieve high drug concentrations within the brain is highly challenging, and nanotechnology is one of the most efficient approaches to this challenge. In this review, we compiled recent studies on the development of nanoparticles for brain drug delivery and diagnostics, including lipidic, polymeric, and metallic nanoparticles. Examples of nanoparticle-loaded natural and synthetic drugs for neurodegenerative and neuropsychiatric treatment were presented alongside magnetic nanoparticles for brain imaging. Nanoparticle functionalization strategies to promote active transport through the BBB were also discussed, including with antibodies, RNA aptamers, transferrin, lactoferrin, glucose, ApoE, glutathione, and angiopep-2. In spite of the extensive progress made in the field of nanotechnology to boost diagnostic and therapeutic strategies for neurological and neuropsychiatric disorders, the integration of nanomedicine into clinical studies remains an ongoing challenge.

Author Contributions

Conceptualization, C.G. and M.F.B.; methodology, C.G., A.S. and M.F.B.; validation, C.G., A.S., C.D.-M. and M.F.B.; investigation, C.G., A.S. and M.F.B.; resources, C.G., C.D.-M. and M.F.B.; writing—original draft preparation, C.G., A.S. and M.F.B.; writing—review and editing, C.G., A.S., C.D.-M. and M.F.B.; visualization, C.G., A.S., C.D.-M. and M.F.B.; funding acquisition, C.G., C.D.-M. and M.F.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Ibero-American Program on Science and Technology (CYTED—GENOPSYSEN, P223RT0141), and was also financially supported by Portuguese national funds via the projects UIDB/50006/2020, UIDP/50006/2020, and LA/P/0008/2020 from the Fundação para a Ciência e a Tecnologia (FCT) and Ministério da Ciência, Tecnologia e Ensino Superior (MCTES). C.G. (CEECIND/03436/2020) and M.F.B. (2020.03107.CEECIND) are grateful for their investigator contacts.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data are contained within the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. WHO Dementia—Key Facts. Available online: https://www.who.int/news-room/fact-sheets/detail/dementia (accessed on 15 October 2023).
  2. WHO Parkinson Disease—Key Facts. Available online: https://www.who.int/news-room/fact-sheets/detail/parkinson-disease (accessed on 15 October 2023).
  3. Bloomingdale, P.; Karelina, T.; Ramakrishnan, V.; Bakshi, S.; Véronneau-Veilleux, F.; Moye, M.; Sekiguchi, K.; Meno-Tetang, G.; Mohan, A.; Maithreye, R.; et al. Hallmarks of neurodegenerative disease: A systems pharmacology perspective. CPT Pharmacomet. Syst. Pharmacol. 2022, 11, 1399–1429. [Google Scholar] [CrossRef] [PubMed]
  4. Mizuno, Y. Definition and Classification of Parkinsonian Drugs. In NeuroPsychopharmacotherapy; Springer International Publishing: Cham, Switzerland, 2022; pp. 2823–2852. [Google Scholar]
  5. Cheong, S.L.; Tiew, J.K.; Fong, Y.H.; Leong, H.W.; Chan, Y.M.; Chan, Z.L.; Kong, E.W.J. Current Pharmacotherapy and Multi-Target Approaches for Alzheimer’s Disease. Pharmaceuticals 2022, 15, 1560. [Google Scholar] [CrossRef] [PubMed]
  6. Vasileva, L.; Gaynanova, G.; Valeeva, F.; Belyaev, G.; Zueva, I.; Bushmeleva, K.; Sibgatullina, G.; Samigullin, D.; Vyshtakalyuk, A.; Petrov, K.; et al. Mitochondria-Targeted Delivery Strategy of Dual-Loaded Liposomes for Alzheimer’s Disease Therapy. Int. J. Mol. Sci. 2023, 24, 10494. [Google Scholar] [CrossRef] [PubMed]
  7. Liu, Y.; An, S.; Li, J.; Kuang, Y.; He, X.; Guo, Y.; Ma, H.; Zhang, Y.; Ji, B.; Jiang, C. Brain-targeted co-delivery of therapeutic gene and peptide by multifunctional nanoparticles in Alzheimer’s disease mice. Biomaterials 2016, 80, 33–45. [Google Scholar] [CrossRef]
  8. Guerzoni, L.P.B.; Nicolas, V.; Angelova, A. In Vitro Modulation of TrkB Receptor Signaling upon Sequential Delivery of Curcumin-DHA Loaded Carriers towards Promoting Neuronal Survival. Pharm. Res. 2017, 34, 492–505. [Google Scholar] [CrossRef]
  9. Glinz, D.; Gloy, V.L.; Monsch, A.U.; Kressig, R.W.; Patel, C.; McCord, K.A.; Ademi, Z.; Tomonaga, Y.; Schwenkglenks, M.; Bucher, H.C.; et al. Acetylcholinesterase inhibitors combined with memantine for moderate to severe Alzheimer’s disease: A meta-analysis. Swiss Med. Wkly. 2019, 149, w20093. [Google Scholar] [CrossRef]
  10. Kabir, M.T.; Uddin, M.S.; Mamun, A.A.; Jeandet, P.; Aleya, L.; Mansouri, R.A.; Ashraf, G.M.; Mathew, B.; Bin-Jumah, M.N.; Abdel-Daim, M.M. Combination Drug Therapy for the Management of Alzheimer’s Disease. Int. J. Mol. Sci. 2020, 21, 3272. [Google Scholar] [CrossRef]
  11. Mateev, E.; Kondeva-Burdina, M.; Georgieva, M.; Zlatkov, A. Repurposing of FDA-approved drugs as dual-acting MAO-B and AChE inhibitors against Alzheimer’s disease: An in silico and in vitro study. J. Mol. Graph. Model. 2023, 122, 108471. [Google Scholar] [CrossRef]
  12. Chaib, S.; Vidal, B.; Bouillot, C.; Depoortere, R.; Newman-Tancredi, A.; Zimmer, L.; Levigoureux, E. Multimodal imaging study of the 5-HT1A receptor biased agonist, NLX-112, in a model of L-DOPA-induced dyskinesia. NeuroImage Clin. 2023, 39, 103497. [Google Scholar] [CrossRef]
  13. WHO Depressive Disorder (Depression)—Key Facts. Available online: https://www.who.int/news-room/fact-sheets/detail/depression (accessed on 10 October 2023).
  14. WHO Mental Disorders—Key Facts. Available online: https://www.who.int/news-room/fact-sheets/detail/mental-disorders (accessed on 10 October 2023).
  15. Paul, S.M.; Potter, W.Z. Finding new and better treatments for psychiatric disorders. Neuropsychopharmacology 2023, 49, 3–9. [Google Scholar] [CrossRef]
  16. Li, Z.; Ruan, M.; Chen, J.; Fang, Y. Major Depressive Disorder: Advances in Neuroscience Research and Translational Applications. Neurosci. Bull. 2021, 37, 863–880. [Google Scholar] [CrossRef] [PubMed]
  17. Nikayin, S.; Murphy, E.; Krystal, J.H.; Wilkinson, S.T. Long-term safety of ketamine and esketamine in treatment of depression. Expert Opin. Drug Saf. 2022, 21, 777–787. [Google Scholar] [CrossRef] [PubMed]
  18. Johnston, J.N.; Henter, I.D.; Zarate, C.A. The antidepressant actions of ketamine and its enantiomers. Pharmacol. Ther. 2023, 246, 108431. [Google Scholar] [CrossRef] [PubMed]
  19. Henssler, J.; Alexander, D.; Schwarzer, G.; Bschor, T.; Baethge, C. Combining Antidepressants vs Antidepressant Monotherapy for Treatment of Patients with Acute Depression—A Systematic Review and Meta-Analysis. JAMA Psychiatry 2022, 79, 300–312. [Google Scholar] [CrossRef] [PubMed]
  20. Garakani, A.; Murrough, J.W.; Freire, R.C.; Thom, R.P.; Larkin, K.; Buono, F.D.; Iosifescu, D.V. Pharmacotherapy of Anxiety Disorders: Current and Emerging Treatment Options. Front. Psychiatry 2020, 11, 1412. [Google Scholar] [CrossRef]
  21. Gupta, P.R.; Prabhavalkar, K. Combination therapy with neuropeptides for the treatment of anxiety disorder. Neuropeptides 2021, 86, 102127. [Google Scholar] [CrossRef]
  22. Jagaran, K.; Singh, M. Lipid Nanoparticles: Promising Treatment Approach for Parkinson’s Disease. Int. J. Mol. Sci. 2022, 23, 9361. [Google Scholar] [CrossRef]
  23. Pulgar, V.M. Transcytosis to Cross the Blood Brain Barrier, New Advancements and Challenges. Front. Neurosci. 2019, 12, 1019. [Google Scholar] [CrossRef]
  24. Puris, E.; Fricker, G.; Gynther, M. Targeting Transporters for Drug Delivery to the Brain: Can We Do Better? Pharm. Res. 2022, 39, 1415–1455. [Google Scholar] [CrossRef]
  25. Zhang, W.; Liu, Q.Y.; Haqqani, A.S.; Leclerc, S.; Liu, Z.; Fauteux, F.; Baumann, E.; Delaney, C.E.; Ly, D.; Star, A.T.; et al. Differential expression of receptors mediating receptor-mediated transcytosis (RMT) in brain microvessels, brain parenchyma and peripheral tissues of the mouse and the human. Fluids Barriers CNS 2020, 17, 47. [Google Scholar] [CrossRef]
  26. Wong, A.D.; Ye, M.; Levy, A.F.; Rothstein, J.D.; Bergles, D.E.; Searson, P.C. The blood-brain barrier: An engineering perspective. Front. Neuroeng. 2013, 6, 7. [Google Scholar] [CrossRef]
  27. Lombardo, S.M.; Schneider, M.; Türeli, A.E.; Günday Türeli, N. Key for crossing the BBB with nanoparticles: The rational design. Beilstein J. Nanotechnol. 2020, 11, 866–883. [Google Scholar] [CrossRef] [PubMed]
  28. Sanità, G.; Carrese, B.; Lamberti, A. Nanoparticle Surface Functionalization: How to Improve Biocompatibility and Cellular Internalization. Front. Mol. Biosci. 2020, 7, 587012. [Google Scholar] [CrossRef] [PubMed]
  29. Thambiliyagodage, C. Ligand exchange reactions and PEG stabilization of gold nanoparticles. Curr. Res. Green Sustain. Chem. 2022, 5, 100245. [Google Scholar] [CrossRef]
  30. Smolensky, E.D.; Park, H.E.; Berquó, T.S.; Pierre, V.C. Surface functionalization of magnetic iron oxide nanoparticles for MRI applications—Effect of anchoring group and ligand exchange protocol. Contrast Media Mol. Imaging 2011, 6, 189–199. [Google Scholar] [CrossRef] [PubMed]
  31. Dinkel, R.; Braunschweig, B.; Peukert, W. Fast and Slow Ligand Exchange at the Surface of Colloidal Gold Nanoparticles. J. Phys. Chem. C 2016, 120, 1673–1682. [Google Scholar] [CrossRef]
  32. Wagner, S.; Zensi, A.; Wien, S.L.; Tschickardt, S.E.; Maier, W.; Vogel, T.; Worek, F.; Pietrzik, C.U.; Kreuter, J.; von Briesen, H. Uptake Mechanism of ApoE-Modified Nanoparticles on Brain Capillary Endothelial Cells as a Blood-Brain Barrier Model. PLoS ONE 2012, 7, e32568. [Google Scholar] [CrossRef]
  33. Rajora, M.A.; Ding, L.; Valic, M.; Jiang, W.; Overchuk, M.; Chen, J.; Zheng, G. Tailored theranostic apolipoprotein E3 porphyrin-lipid nanoparticles target glioblastoma. Chem. Sci. 2017, 8, 5371–5384. [Google Scholar] [CrossRef]
  34. Nicolle, L.; Journot, C.M.A.; Gerber-Lemaire, S. Chitosan Functionalization: Covalent and Non-Covalent Interactions and Their Characterization. Polymers 2021, 13, 4118. [Google Scholar] [CrossRef]
  35. Mahajan, G.; Kaur, M.; Gupta, R. Green functionalized nanomaterials: Fundamentals and future opportunities. In Green Functionalized Nanomaterials for Environmental Applications; Elsevier: Amsterdam, The Netherlands, 2022; pp. 21–41. [Google Scholar]
  36. Ye, Z.; Gastfriend, B.D.; Umlauf, B.J.; Lynn, D.M.; Shusta, E.V. Antibody-Targeted Liposomes for Enhanced Targeting of the Blood-Brain Barrier. Pharm. Res. 2022, 39, 1523–1534. [Google Scholar] [CrossRef]
  37. Schnyder, A.; Huwyler, J. Drug transport to brain with targeted liposomes. NeuroRX 2005, 2, 99–107. [Google Scholar] [CrossRef]
  38. Zhang, Y.; He, J.; Shen, L.; Wang, T.; Yang, J.; Li, Y.; Wang, Y.; Quan, D. Brain-targeted delivery of obidoxime, using aptamer-modified liposomes, for detoxification of organophosphorus compounds. J. Control. Release 2021, 329, 1117–1128. [Google Scholar] [CrossRef] [PubMed]
  39. Juhairiyah, F.; de Lange, E.C.M. Understanding Drug Delivery to the Brain Using Liposome-Based Strategies: Studies that Provide Mechanistic Insights Are Essential. AAPS J. 2021, 23, 114. [Google Scholar] [CrossRef] [PubMed]
  40. Suk, J.S.; Xu, Q.; Kim, N.; Hanes, J.; Ensign, L.M. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv. Drug Deliv. Rev. 2016, 99, 28–51. [Google Scholar] [CrossRef]
  41. Luo, Y.; Yang, H.; Zhou, Y.-F.; Hu, B. Dual and multi-targeted nanoparticles for site-specific brain drug delivery. J. Control. Release 2020, 317, 195–215. [Google Scholar] [CrossRef] [PubMed]
  42. 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. 2021, 18, 714–725. [Google Scholar] [CrossRef]
  43. Stalmans, S.; Bracke, N.; Wynendaele, E.; Gevaert, B.; Peremans, K.; Burvenich, C.; Polis, I.; De Spiegeleer, B. Cell-Penetrating Peptides Selectively Cross the Blood-Brain Barrier In Vivo. PLoS ONE 2015, 10, e0139652. [Google Scholar] [CrossRef]
  44. Neves, A.R.; van der Putten, L.; Queiroz, J.F.; Pinheiro, M.; Reis, S. Transferrin-functionalized lipid nanoparticles for curcumin brain delivery. J. Biotechnol. 2021, 331, 108–117. [Google Scholar] [CrossRef]
  45. Neves, A.R.; Queiroz, J.F.; Reis, S. Brain-targeted delivery of resveratrol using solid lipid nanoparticles functionalized with apolipoprotein E. J. Nanobiotechnology 2016, 14, 27. [Google Scholar] [CrossRef]
  46. Neves, A.R.; Queiroz, J.F.; Lima, S.A.C.; Reis, S. Apo E-Functionalization of Solid Lipid Nanoparticles Enhances Brain Drug Delivery: Uptake Mechanism and Transport Pathways. Bioconjug. Chem. 2017, 28, 995–1004. [Google Scholar] [CrossRef]
  47. Monge, M.; Fornaguera, C.; Quero, C.; Dols-Perez, A.; Calderó, G.; Grijalvo, S.; García-Celma, M.J.; Rodríguez-Abreu, C.; Solans, C. Functionalized PLGA nanoparticles prepared by nano-emulsion templating interact selectively with proteins involved in the transport through the blood-brain barrier. Eur. J. Pharm. Biopharm. 2020, 156, 155–164. [Google Scholar] [CrossRef]
  48. Nsairat, H.; Khater, D.; Sayed, U.; Odeh, F.; Al Bawab, A.; Alshaer, W. Liposomes: Structure, composition, types, and clinical applications. Heliyon 2022, 8, e09394. [Google Scholar] [CrossRef] [PubMed]
  49. Spuch, C.; Navarro, C. Liposomes for Targeted Delivery of Active Agents against Neurodegenerative Diseases (Alzheimer’s Disease and Parkinson’s Disease). J. Drug Deliv. 2011, 2011, 469679. [Google Scholar] [CrossRef] [PubMed]
  50. 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]
  51. Salarpour, S.; Barani, M.; Pardakhty, A.; Khatami, M.; Pal Singh Chauhan, N. The application of exosomes and Exosome-nanoparticle in treating brain disorders. J. Mol. Liq. 2022, 350, 118549. [Google Scholar] [CrossRef]
  52. Li, X.-H.; Zhang, J.; Li, D.-F.; Wu, W.; Xie, Z.-W.; Liu, Q. Physiological and pathological insights into exosomes in the brain. Zool. Res. 2020, 41, 365–372. [Google Scholar] [CrossRef]
  53. Rotman, M.; Welling, M.M.; Bunschoten, A.; de Backer, M.E.; Rip, J.; Nabuurs, R.J.A.; 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]
  54. Mutlu, N.B.; Değim, Z.; Yılmaz, Ş.; Eşsiz, D.; Nacar, A. New perspective for the treatment of Alzheimer diseases: Liposomal rivastigmine formulations. Drug Dev. Ind. Pharm. 2011, 37, 775–789. [Google Scholar] [CrossRef]
  55. 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]
  56. Ismail, M.F.; ElMeshad, A.N.; Salem, N.A.H. Potential therapeutic effect of nanobased formulation of rivastigmine on rat model of Alzheimer’s disease. Int. J. Nanomed. 2013, 8, 393–406. [Google Scholar] [CrossRef]
  57. Li, W.; Zhou, Y.; Zhao, N.; Hao, B.; Wang, X.; Kong, P. Pharmacokinetic behavior and efficiency of acetylcholinesterase inhibition in rat brain after intranasal administration of galanthamine hydrobromide loaded flexible liposomes. Environ. Toxicol. Pharmacol. 2012, 34, 272–279. [Google Scholar] [CrossRef]
  58. Phachonpai, W.; Wattanathorn, J.; Muchimapura, S.; Tong-Un, T.; Preechagoon, D. Neuroprotective Effect of Quercetin Encapsulated Liposomes: A Novel Therapeutic Strategy against Alzheimer’s Disease. Am. J. Appl. Sci. 2010, 7, 480–485. [Google Scholar] [CrossRef]
  59. Xiang, Y.; Wu, Q.; Liang, L.; Wang, X.; Wang, J.; Zhang, X.; Pu, X.; Zhang, Q. Chlorotoxin-modified stealth liposomes encapsulating levodopa for the targeting delivery against the Parkinson’s disease in the MPTP-induced mice model. J. Drug Target. 2012, 20, 67–75. [Google Scholar] [CrossRef] [PubMed]
  60. Migliore, M.M.; Ortiz, R.; Dye, S.; Campbell, R.B.; Amiji, M.M.; Waszczak, B.L. Neurotrophic and neuroprotective efficacy of intranasal GDNF in a rat model of Parkinson’s disease. Neuroscience 2014, 274, 11–23. [Google Scholar] [CrossRef]
  61. Kahana, M.; Weizman, A.; Gabay, M.; Loboda, Y.; Segal-Gavish, H.; Gavish, A.; Barhum, Y.; Offen, D.; Finberg, J.; Allon, N.; et al. Liposome-based targeting of dopamine to the brain: A novel approach for the treatment of Parkinson’s disease. Mol. Psychiatry 2021, 26, 2626–2632. [Google Scholar] [CrossRef]
  62. Chen, J.; Liu, Q.; Wang, Y.; Guo, Y.; Xu, X.; Huang, P.; Lian, B.; Zhang, R.; Chen, Y.; Ha, Y. Protective effects of resveratrol liposomes on mitochondria in substantia nigra cells of parkinsonized rats. Ann. Palliat. Med. 2021, 10, 2458–2468. [Google Scholar] [CrossRef] [PubMed]
  63. Marino, A.; Battaglini, M.; Desii, A.; Lavarello, C.; Genchi, G.; Petretto, A.; Ciofani, G. Liposomes loaded with polyphenol-rich grape pomace extracts protect from neurodegeneration in a rotenone-based in vitro model of Parkinson’s disease. Biomater. Sci. 2021, 9, 8171–8188. [Google Scholar] [CrossRef] [PubMed]
  64. Moreno, L.C.; da Silva Oliveira, G.Z.; Cavalcanti, I.M.F.; Santos-Magalhães, N.S.; Rolim, H.M.L.; de Freitas, R.M. Development and evaluation of liposomal formulation containing nimodipine on anxiolytic activity in mice. Pharmacol. Biochem. Behav. 2014, 116, 64–68. [Google Scholar] [CrossRef] [PubMed]
  65. Siyal, F.J.; Siddiqui, R.A.; Memon, Z.; Aslam, Z.; Nisar, U.; Imad, R.; Shah, M.R. Eugenol and its liposome-based nano carrier reduce anxiety by inhibiting glyoxylase-1 expression in mice. Brazilian J. Biol. 2023, 83, e251219. [Google Scholar] [CrossRef] [PubMed]
  66. Siyal, F.J.; Memon, Z.; Siddiqui, R.A.; Aslam, Z.; Nisar, U.; Imad, R.; Shah, M.R. Eugenol and liposome-based nanocarriers loaded with eugenol protect against anxiolytic disorder via down regulation of neurokinin-1 receptors in mice. Pak. J. Pharm. Sci. 2020, 33, 2275–2284. [Google Scholar] [CrossRef]
  67. Tong-Un, T.; Wannanon, P.; Wattanatho, J.; Phachonpai, W. Quercetin Liposomes via Nasal Administration Reduce Anxiety and Depression-like Behaviors and Enhance Cognitive Performances in Rats. Am. J. Pharmacol. Toxicol. 2010, 5, 80–88. [Google Scholar] [CrossRef]
  68. Diniz, D.M.; Franze, S.; Homberg, J.R. Crossing the Blood-Brain-Barrier: A bifunctional liposome for BDNF gene delivery—A Pilot Study. bioRxiv 2020. [Google Scholar] [CrossRef]
  69. Moreno, L.C.; Rolim, H.M.L.; Freitas, R.M.; Santos-Magalhães, N.S. Antidepressant-like activity of liposomal formulation containing nimodipine treatment in the tail suspension test, forced swim test and MAOB activity in mice. Brain Res. 2016, 1646, 235–240. [Google Scholar] [CrossRef] [PubMed]
  70. Chauhan, T.; Rani, V.; Sahu, B.; Sharma, A.; Chand Kheruka, S.; Gambhir, S.; Dube, V.; Aggarwal, L.M.; Chawla, R. Negatively charged liposomes of sertraline hydrochloride: Formulation, characterization and pharmacokinetic studies. J. Drug Deliv. Sci. Technol. 2020, 58, 101780. [Google Scholar] [CrossRef]
  71. Priprem, A.; Chonpathompikunlert, P.; Sutthiparinyanont, S.; Wattanathorn, J. Antidepressant and cognitive activities of intranasal piperine-encapsulated liposomes. Adv. Biosci. Biotechnol. 2011, 02, 108–116. [Google Scholar] [CrossRef]
  72. Zhang, Y.; Liu, Y.; Liu, H.; Tang, W.H. Exosomes: Biogenesis, biologic function and clinical potential. Cell Biosci. 2019, 9, 19. [Google Scholar] [CrossRef] [PubMed]
  73. Saeedi, S.; Israel, S.; Nagy, C.; Turecki, G. The emerging role of exosomes in mental disorders. Transl. Psychiatry 2019, 9, 122. [Google Scholar] [CrossRef]
  74. Hornung, S.; Dutta, S.; Bitan, G. CNS-Derived Blood Exosomes as a Promising Source of Biomarkers: Opportunities and Challenges. Front. Mol. Neurosci. 2020, 13, 38. [Google Scholar] [CrossRef]
  75. Xia, X.; Wang, Y.; Huang, Y.; Zhang, H.; Lu, H.; Zheng, J.C. Exosomal miRNAs in central nervous system diseases: Biomarkers, pathological mediators, protective factors and therapeutic agents. Prog. Neurobiol. 2019, 183, 101694. [Google Scholar] [CrossRef]
  76. Satapathy, M.K.; Yen, T.-L.; Jan, J.-S.; Tang, R.-D.; Wang, J.-Y.; Taliyan, R.; Yang, C.-H. Solid Lipid Nanoparticles (SLNs): An Advanced Drug Delivery System Targeting Brain through BBB. Pharmaceutics 2021, 13, 1183. [Google Scholar] [CrossRef]
  77. Wang, H.; Sui, H.; Zheng, Y.; Jiang, Y.; Shi, Y.; Liang, J.; Zhao, L. Curcumin-primed exosomes potently ameliorate cognitive function in AD mice by inhibiting hyperphosphorylation of the Tau protein through the AKT/GSK-3β pathway. Nanoscale 2019, 11, 7481–7496. [Google Scholar] [CrossRef] [PubMed]
  78. Qi, Y.; Guo, L.; Jiang, Y.; Shi, Y.; Sui, H.; Zhao, L. Brain delivery of quercetin-loaded exosomes improved cognitive function in AD mice by inhibiting phosphorylated tau-mediated neurofibrillary tangles. Drug Deliv. 2020, 27, 745–755. [Google Scholar] [CrossRef] [PubMed]
  79. Alvarez-Erviti, L.; Seow, Y.; Yin, H.; Betts, C.; Lakhal, S.; Wood, M.J.A. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 2011, 29, 341–345. [Google Scholar] [CrossRef]
  80. Qu, M.; Lin, Q.; Huang, L.; Fu, Y.; Wang, L.; He, S.; Fu, Y.; Yang, S.; Zhang, Z.; Zhang, L.; et al. Dopamine-loaded blood exosomes targeted to brain for better treatment of Parkinson’s disease. J. Control. Release 2018, 287, 156–166. [Google Scholar] [CrossRef] [PubMed]
  81. Haney, M.J.; Klyachko, N.L.; Zhao, Y.; Gupta, R.; Plotnikova, E.G.; He, Z.; Patel, T.; Piroyan, A.; Sokolsky, M.; Kabanov, A.V.; et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J. Control. Release 2015, 207, 18–30. [Google Scholar] [CrossRef]
  82. Cooper, J.M.; Wiklander, P.B.O.; Nordin, J.Z.; Al-Shawi, R.; Wood, M.J.; Vithlani, M.; Schapira, A.H.V.; Simons, J.P.; El-Andaloussi, S.; Alvarez-Erviti, L. Systemic exosomal siRNA delivery reduced alpha-synuclein aggregates in brains of transgenic mice. Mov. Disord. 2014, 29, 1476–1485. [Google Scholar] [CrossRef] [PubMed]
  83. Ren, X.; Zhao, Y.; Xue, F.; Zheng, Y.; Huang, H.; Wang, W.; Chang, Y.; Yang, H.; Zhang, J. Exosomal DNA Aptamer Targeting α-Synuclein Aggregates Reduced Neuropathological Deficits in a Mouse Parkinson’s Disease Model. Mol. Ther. Nucleic Acids 2019, 17, 726–740. [Google Scholar] [CrossRef]
  84. 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]
  85. 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]
  86. Dhawan, S.; Kapil, R.; Singh, B. Formulation development and systematic optimization of solid lipid nanoparticles of quercetin for improved brain delivery. J. Pharm. Pharmacol. 2011, 63, 342–351. [Google Scholar] [CrossRef]
  87. Yusuf, M.; Khan, M.; Khan, R.A.; Ahmed, B. Preparation, characterization, in vivo and biochemical evaluation of brain targeted Piperine solid lipid nanoparticles in an experimentally induced Alzheimer’s disease model. J. Drug Target. 2013, 21, 300–311. [Google Scholar] [CrossRef] [PubMed]
  88. Smith, A.; Giunta, B.; Bickford, P.C.; Fountain, M.; Tan, J.; Shytle, R.D. Nanolipidic particles improve the bioavailability and α-secretase inducing ability of epigallocatechin-3-gallate (EGCG) for the treatment of Alzheimer’s disease. Int. J. Pharm. 2010, 389, 207–212. [Google Scholar] [CrossRef]
  89. Kakkar, V.; Kaur, I.P. Evaluating potential of curcumin loaded solid lipid nanoparticles in aluminium induced behavioural, biochemical and histopathological alterations in mice brain. Food Chem. Toxicol. 2011, 49, 2906–2913. [Google Scholar] [CrossRef] [PubMed]
  90. Tsai, M.-J.; Huang, Y.-B.; Wu, P.-C.; Fu, Y.-S.; Kao, Y.-R.; Fang, J.-Y.; Tsai, Y.-H. Oral Apomorphine Delivery from Solid Lipid Nanoparticles with Different Monostearate Emulsifiers: Pharmacokinetic and Behavioral Evaluations. J. Pharm. Sci. 2011, 100, 547–557. [Google Scholar] [CrossRef] [PubMed]
  91. Pardeshi, C.V.; Rajput, P.V.; Belgamwar, V.S.; Tekade, A.R.; Surana, S.J. Novel surface modified solid lipid nanoparticles as intranasal carriers for ropinirole hydrochloride: Application of factorial design approach. Drug Deliv. 2013, 20, 47–56. [Google Scholar] [CrossRef] [PubMed]
  92. Megala, M.; Shivaramakrishnan, B.; Kishore, R.M.; Balashanmugam, K. Neuroprotective potential of Naringenin-loaded solid-lipid nanoparticles against rotenone-induced Parkinson’s disease model. J. Appl. Pharm. Sci. 2020, 11, 19–28. [Google Scholar] [CrossRef]
  93. Vitorino, C.; Silva, S.; Gouveia, F.; Bicker, J.; Falcão, A.; Fortuna, A. QbD-driven development of intranasal lipid nanoparticles for depression treatment. Eur. J. Pharm. Biopharm. 2020, 153, 106–120. [Google Scholar] [CrossRef]
  94. Nirale, P.; Paul, A.; Yadav, K.S. Nanoemulsions for targeting the neurodegenerative diseases: Alzheimer’s, Parkinson’s and Prion’s. Life Sci. 2020, 245, 117394. [Google Scholar] [CrossRef]
  95. Misra, S.K.; Pathak, K. Nose-to-Brain Targeting via Nanoemulsion: Significance and Evidence. Colloids Interfaces 2023, 7, 23. [Google Scholar] [CrossRef]
  96. Singh, Y.; Meher, J.G.; Raval, K.; Khan, F.A.; Chaurasia, M.; Jain, N.K.; Chourasia, M.K. Nanoemulsion: Concepts, development and applications in drug delivery. J. Control. Release 2017, 252, 28–49. [Google Scholar] [CrossRef]
  97. Kaur, A.; Nigam, K.; Srivastava, S.; Tyagi, A.; Dang, S. Memantine nanoemulsion: A new approach to treat Alzheimer’s disease. J. Microencapsul. 2020, 37, 355–365. [Google Scholar] [CrossRef] [PubMed]
  98. Kaur, A.; Nigam, K.; Bhatnagar, I.; Sukhpal, H.; Awasthy, S.; Shankar, S.; Tyagi, A.; Dang, S. Treatment of Alzheimer’s diseases using donepezil nanoemulsion: An intranasal approach. Drug Deliv. Transl. Res. 2020, 10, 1862–1875. [Google Scholar] [CrossRef]
  99. Jiang, Y.; Liu, C.; Zhai, W.; Zhuang, N.; Han, T.; Ding, Z. The Optimization Design of Lactoferrin Loaded HupA Nanoemulsion for Targeted Drug Transport via Intranasal Route. Int. J. Nanomed. 2019, 14, 9217–9234. [Google Scholar] [CrossRef] [PubMed]
  100. Kotta, S.; Mubarak Aldawsari, H.; Badr-Eldin, S.M.; Alhakamy, N.A.; Md, S. Coconut oil-based resveratrol nanoemulsion: Optimization using response surface methodology, stability assessment and pharmacokinetic evaluation. Food Chem. 2021, 357, 129721. [Google Scholar] [CrossRef] [PubMed]
  101. Nasr, M. Development of an optimized hyaluronic acid-based lipidic nanoemulsion co-encapsulating two polyphenols for nose to brain delivery. Drug Deliv. 2016, 23, 1444–1452. [Google Scholar] [CrossRef]
  102. Kumar, S.; Ali, J.; Baboota, S. Design Expert ® supported optimization and predictive analysis of selegiline nanoemulsion via the olfactory region with enhanced behavioural performance in Parkinson’s disease. Nanotechnology 2016, 27, 435101. [Google Scholar] [CrossRef] [PubMed]
  103. Das, S.S.; Sarkar, A.; Chabattula, S.C.; Verma, P.R.P.; Nazir, A.; Gupta, P.K.; Ruokolainen, J.; Kesari, K.K.; Singh, S.K. Food-Grade Quercetin-Loaded Nanoemulsion Ameliorates Effects Associated with Parkinson’s Disease and Cancer: Studies Employing a Transgenic C. elegans Model and Human Cancer Cell Lines. Antioxidants 2022, 11, 1378. [Google Scholar] [CrossRef]
  104. Gaba, B.; Khan, T.; Haider, M.F.; Alam, T.; Baboota, S.; Parvez, S.; Ali, J. Vitamin E Loaded Naringenin Nanoemulsion via Intranasal Delivery for the Management of Oxidative Stress in a 6-OHDA Parkinson’s Disease Model. Biomed Res. Int. 2019, 2019, 2382563. [Google Scholar] [CrossRef]
  105. da Silva Campelo, M.; Câmara Neto, J.F.; de Souza, Á.L.; Ferreira, M.K.A.; dos Santos, H.S.; Gramosa, N.V.; de Aguiar Soares, S.; Ricardo, N.M.P.S.; de Menezes, J.E.S.A.; Ribeiro, M.E.N.P. Clove volatile oil-loaded nanoemulsion reduces the anxious-like behavior in adult zebrafish. DARU J. Pharm. Sci. 2023, 31, 183–192. [Google Scholar] [CrossRef]
  106. Kumar, A.; Jain, S.K. Preliminary studies for the development of intranasal nanoemulsion containing CNS agent: Emphasizing the utilization of cut and weigh method. Artif. Cells Nanomed. Biotechnol. 2017, 45, 515–521. [Google Scholar] [CrossRef]
  107. Boche, M.; Pokharkar, V. Quetiapine Nanoemulsion for Intranasal Drug Delivery: Evaluation of Brain-Targeting Efficiency. AAPS PharmSciTech 2017, 18, 686–696. [Google Scholar] [CrossRef]
  108. Pandey, Y.R.; Kumar, S.; Gupta, B.K.; Ali, J.; Baboota, S. Intranasal delivery of paroxetine nanoemulsion via the olfactory region for the management of depression: Formulation, behavioural and biochemical estimation. Nanotechnology 2016, 27, 025102. [Google Scholar] [CrossRef] [PubMed]
  109. Zielińska, A.; Carreiró, F.; Oliveira, A.M.; Neves, A.; Pires, B.; Venkatesh, D.N.; Durazzo, A.; Lucarini, M.; Eder, P.; Silva, A.M.; et al. Polymeric Nanoparticles: Production, Characterization, Toxicology and Ecotoxicology. Molecules 2020, 25, 3731. [Google Scholar] [CrossRef] [PubMed]
  110. Elmowafy, M.; Shalaby, K.; Elkomy, M.H.; Alsaidan, O.A.; Gomaa, H.A.M.; Abdelgawad, M.A.; Mostafa, E.M. Polymeric Nanoparticles for Delivery of Natural Bioactive Agents: Recent Advances and Challenges. Polymers 2023, 15, 1123. [Google Scholar] [CrossRef]
  111. Wüpper, S.; Lüersen, K.; Rimbach, G. Cyclodextrins, Natural Compounds, and Plant Bioactives—A Nutritional Perspective. Biomolecules 2021, 11, 401. [Google Scholar] [CrossRef] [PubMed]
  112. Xu, W.; Li, X.; Wang, L.; Li, S.; Chu, S.; Wang, J.; Li, Y.; Hou, J.; Luo, Q.; Liu, J. Design of Cyclodextrin-Based Functional Systems for Biomedical Applications. Front. Chem. 2021, 9, 635507. [Google Scholar] [CrossRef]
  113. Ghitman, J.; Voicu, S.I. Controlled drug delivery mediated by cyclodextrin-based supramolecular self-assembled carriers: From design to clinical performances. Carbohydr. Polym. Technol. Appl. 2023, 5, 100266. [Google Scholar] [CrossRef]
  114. La Barbera, L.; Mauri, E.; D’Amelio, M.; Gori, M. Functionalization strategies of polymeric nanoparticles for drug delivery in Alzheimer’s disease: Current trends and future perspectives. Front. Neurosci. 2022, 16, 939855. [Google Scholar] [CrossRef] [PubMed]
  115. Nagpal, K.; Singh, S.K.; Mishra, D.N. Optimization of brain targeted chitosan nanoparticles of Rivastigmine for improved efficacy and safety. Int. J. Biol. Macromol. 2013, 59, 72–83. [Google Scholar] [CrossRef]
  116. Fazil, M.; Md, S.; Haque, S.; Kumar, M.; Baboota, S.; kaur Sahni, J.; Ali, J. Development and evaluation of rivastigmine loaded chitosan nanoparticles for brain targeting. Eur. J. Pharm. Sci. 2012, 47, 6–15. [Google Scholar] [CrossRef]
  117. Hanafy, A.S.; Farid, R.M.; Helmy, M.W.; ElGamal, S.S. Pharmacological, toxicological and neuronal localization assessment of galantamine/chitosan complex nanoparticles in rats: Future potential contribution in Alzheimer’s disease management. Drug Deliv. 2016, 23, 3111–3122. [Google Scholar] [CrossRef]
  118. Hassanzadeh, G.; Fallahi, Z.; Khanmohammadi, M.; Elmizadeh, H.; Sharifzadeh, M.; Nouri, K.; Heydarian, Z.; Mahakizadeh, S.; Zendedel, A.; Beyer, C.; et al. Effect of Magnetic Tacrine-Loaded Chitosan Nanoparticles on Spatial Learning, Memory, Amyloid Precursor Protein and Seladin-1 Expression in the Hippocampus of Streptozotocin-Exposed Rats. Int. Clin. Neurosci. J. 2016, 3, 25–31. [Google Scholar] [CrossRef]
  119. Yang, R.; Zheng, Y.; Wang, Q.; Zhao, L. Curcumin-loaded chitosan–bovine serum albumin nanoparticles potentially enhanced Aβ 42 phagocytosis and modulated macrophage polarization in Alzheimer’s disease. Nanoscale Res. Lett. 2018, 13, 330. [Google Scholar] [CrossRef]
  120. Elnaggar, Y.S.R.; 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]
  121. Alam, S.; Mustafa, G.; Khan, Z.I.; Islam, F.; Bhatnagar, A.; Ahmad, F.J.; Kumar, M. Development and evaluation of thymoquinone-encapsulated chitosan nanoparticles for nose-to-brain targeting: A pharmacoscintigraphic study. Int. J. Nanomed. 2012, 7, 5705–5718. [Google Scholar] [CrossRef] [PubMed]
  122. Sánchez-López, E.; Ettcheto, M.; Egea, M.A.; Espina, M.; Cano, A.; Calpena, A.C.; Camins, A.; Carmona, N.; Silva, A.M.; Souto, E.B.; et al. Memantine loaded PLGA PEGylated nanoparticles for Alzheimer’s disease: In vitro and in vivo characterization. J. Nanobiotechnology 2018, 16, 32. [Google Scholar] [CrossRef]
  123. Quitschke, W.W.; Steinhauff, N.; Rooney, J. The effect of cyclodextrin-solubilized curcuminoids on amyloid plaques in Alzheimer transgenic mice: Brain uptake and metabolism after intravenous and subcutaneous injection. Alzheimer’s Res. Ther. 2013, 5, 16. [Google Scholar] [CrossRef]
  124. Wong, K.H.; Xie, Y.; Huang, X.; Kadota, K.; Yao, X.-S.; Yu, Y.; Chen, X.; Lu, A.; Yang, Z. Delivering Crocetin across the Blood-Brain Barrier by Using γ-Cyclodextrin to Treat Alzheimer’s Disease. Sci. Rep. 2020, 10, 3654. [Google Scholar] [CrossRef] [PubMed]
  125. Bi, C.; Wang, A.; Chu, Y.; Liu, S.; Mu, H.; Liu, W.; Wu, Z.; Sun, K.; Li, Y. Intranasal delivery of rotigotine to the brain with lactoferrin-modified PEG-PLGA nanoparticles for Parkinson’s disease treatment. Int. J. Nanomed. 2016, 11, 6547–6559. [Google Scholar] [CrossRef] [PubMed]
  126. Ahmad, N. Rasagiline-encapsulated chitosan-coated PLGA nanoparticles targeted to the brain in the treatment of parkinson’s disease. J. Liq. Chromatogr. Relat. Technol. 2017, 40, 677–690. [Google Scholar] [CrossRef]
  127. Chen, T.; Liu, W.; Xiong, S.; Li, D.; Fang, S.; Wu, Z.; Wang, Q.; Chen, X. Nanoparticles Mediating the Sustained Puerarin Release Facilitate Improved Brain Delivery to Treat Parkinson’s Disease. ACS Appl. Mater. Interfaces 2019, 11, 45276–45289. [Google Scholar] [CrossRef] [PubMed]
  128. Barros, M.C.F.; Ribeiro, A.C.F.; Esteso, M.A. Cyclodextrins in Parkinson’s Disease. Biomolecules 2018, 9, 3. [Google Scholar] [CrossRef] [PubMed]
  129. Bari, N.K.; Fazil, M.; Hassan, M.Q.; Haider, M.R.; Gaba, B.; Narang, J.K.; Baboota, S.; Ali, J. Brain delivery of buspirone hydrochloride chitosan nanoparticles for the treatment of general anxiety disorder. Int. J. Biol. Macromol. 2015, 81, 49–59. [Google Scholar] [CrossRef] [PubMed]
  130. Mahmoud, K.Y.; Elhesaisy, N.A.; Rashed, A.R.; Mikhael, E.S.; Fadl, M.I.; Elsadek, M.S.; Mohamed, M.A.; Mostafa, M.A.; Hassan, M.A.; Halema, O.M.; et al. Exploring the potential of intranasally administered naturally occurring quercetin loaded into polymeric nanocapsules as a novel platform for the treatment of anxiety. Sci. Rep. 2023, 13, 510. [Google Scholar] [CrossRef] [PubMed]
  131. Jani, P.; Vanza, J.; Pandya, N.; Tandel, H. Formulation of polymeric nanoparticles of antidepressant drug for intranasal delivery. Ther. Deliv. 2019, 10, 683–696. [Google Scholar] [CrossRef]
  132. Cayero-Otero, M.D.; Gomes, M.J.; Martins, C.; Álvarez-Fuentes, J.; Fernández-Arévalo, M.; Sarmento, B.; Martín-Banderas, L. In vivo biodistribution of venlafaxine-PLGA nanoparticles for brain delivery: Plain vs. functionalized nanoparticles. Expert Opin. Drug Deliv. 2019, 16, 1413–1427. [Google Scholar] [CrossRef]
  133. Haque, S.; Md, S.; Fazil, M.; Kumar, M.; Sahni, J.K.; Ali, J.; Baboota, S. Venlafaxine loaded chitosan NPs for brain targeting: Pharmacokinetic and pharmacodynamic evaluation. Carbohydr. Polym. 2012, 89, 72–79. [Google Scholar] [CrossRef]
  134. Haque, S.; Md, S.; Sahni, J.K.; Ali, J.; Baboota, S. Development and evaluation of brain targeted intranasal alginate nanoparticles for treatment of depression. J. Psychiatr. Res. 2014, 48, 1–12. [Google Scholar] [CrossRef]
  135. Tong, G.-F.; Qin, N.; Sun, L.-W. Development and evaluation of Desvenlafaxine loaded PLGA-chitosan nanoparticles for brain delivery. Saudi Pharm. J. 2017, 25, 844–851. [Google Scholar] [CrossRef]
  136. Singh, D.; Rashid, M.; Hallan, S.S.; Mehra, N.K.; Prakash, A.; Mishra, N. Pharmacological evaluation of nasal delivery of selegiline hydrochloride-loaded thiolated chitosan nanoparticles for the treatment of depression. Artif. Cells Nanomed. Biotechnol. 2015, 44, 865–877. [Google Scholar] [CrossRef]
  137. Aree, T. Advancing insights on β-cyclodextrin inclusion complexes with SSRIs through lens of X-ray diffraction and DFT calculation. Int. J. Pharm. 2021, 609, 121113. [Google Scholar] [CrossRef] [PubMed]
  138. Martins, P.M.; Lima, A.C.; Ribeiro, S.; Lanceros-Mendez, S.; Martins, P. Magnetic Nanoparticles for Biomedical Applications: From the Soul of the Earth to the Deep History of Ourselves. ACS Appl. Bio Mater. 2021, 4, 5839–5870. [Google Scholar] [CrossRef] [PubMed]
  139. Zhao, J.; Xu, N.; Yang, X.; Ling, G.; Zhang, P. The roles of gold nanoparticles in the detection of amyloid-β peptide for Alzheimer’s disease. Colloid Interface Sci. Commun. 2022, 46, 100579. [Google Scholar] [CrossRef]
  140. Mao, X.; Xu, J.; Cui, H. Functional nanoparticles for magnetic resonance imaging. WIREs Nanomed. Nanobiotechnol. 2016, 8, 814–841. [Google Scholar] [CrossRef]
  141. Singh, A.V.; Bansod, G.; Mahajan, M.; Dietrich, P.; Singh, S.P.; Rav, K.; Thissen, A.; Bharde, A.M.; Rothenstein, D.; Kulkarni, S.; et al. Digital Transformation in Toxicology: Improving Communication and Efficiency in Risk Assessment. ACS Omega 2023, 8, 21377–21390. [Google Scholar] [CrossRef]
  142. Wei, H.; Wiśniowska, A.; Fan, J.; Harvey, P.; Li, Y.; Wu, V.; Hansen, E.C.; Zhang, J.; Kaul, M.G.; Frey, A.M.; et al. Single-nanometer iron oxide nanoparticles as tissue-permeable MRI contrast agents. Proc. Natl. Acad. Sci. USA 2021, 118, e2102340118. [Google Scholar] [CrossRef] [PubMed]
  143. Avasthi, A.; Caro, C.; Pozo-Torres, E.; Leal, M.P.; García-Martín, M.L. Magnetic Nanoparticles as MRI Contrast Agents. Top. Curr. Chem. 2020, 378, 40. [Google Scholar] [CrossRef]
  144. Yang, H.; Wang, H.; Wen, C.; Bai, S.; Wei, P.; Xu, B.; Xu, Y.; Liang, C.; Zhang, Y.; Zhang, G.; et al. Effects of iron oxide nanoparticles as T2-MRI contrast agents on reproductive system in male mice. J. Nanobiotechnol. 2022, 20, 98. [Google Scholar] [CrossRef]
  145. Korchinski, D.J.; Taha, M.; Yang, R.; Nathoo, N.; Dunn, J.F. Iron Oxide as an Mri Contrast Agent for Cell Tracking: Supplementary Issue. Magn. Reson. Insights 2015, 8s1, MRI.S23557. [Google Scholar] [CrossRef]
  146. Oberdick, S.D.; Jordanova, K.V.; Lundstrom, J.T.; Parigi, G.; Poorman, M.E.; Zabow, G.; Keenan, K.E. Iron oxide nanoparticles as positive T1 contrast agents for low-field magnetic resonance imaging at 64 mT. Sci. Rep. 2023, 13, 11520. [Google Scholar] [CrossRef]
  147. Tang, T.; Valenzuela, A.; Petit, F.; Chow, S.; Leung, K.; Gorin, F.; Louie, A.Y.; Dhenain, M. In Vivo MRI of Functionalized Iron Oxide Nanoparticles for Brain Inflammation. Contrast Media Mol. Imaging 2018, 2018, 3476476. [Google Scholar] [CrossRef] [PubMed]
  148. Yu, M.; Huang, S.; Yu, K.J.; Clyne, A.M. Dextran and Polymer Polyethylene Glycol (PEG) Coating Reduce Both 5 and 30 nm Iron Oxide Nanoparticle Cytotoxicity in 2D and 3D Cell Culture. Int. J. Mol. Sci. 2012, 13, 5554–5570. [Google Scholar] [CrossRef] [PubMed]
  149. Cheng, K.K.; Chan, P.S.; Fan, S.; Kwan, S.M.; Yeung, K.L.; Wáng, Y.-X.J.; Chow, A.H.L.; Wu, E.X.; Baum, L. Curcumin-conjugated magnetic nanoparticles for detecting amyloid plaques in Alzheimer’s disease mice using magnetic resonance imaging (MRI). Biomaterials 2015, 44, 155–172. [Google Scholar] [CrossRef] [PubMed]
  150. Fernández, T.; Martínez-Serrano, A.; Cussó, L.; Desco, M.; Ramos-Gómez, M. Functionalization and Characterization of Magnetic Nanoparticles for the Detection of Ferritin Accumulation in Alzheimer’s Disease. ACS Chem. Neurosci. 2018, 9, 912–924. [Google Scholar] [CrossRef] [PubMed]
  151. Kim, K.Y.; Chang, K.-A. Therapeutic Potential of Magnetic Nanoparticle-Based Human Adipose-Derived Stem Cells in a Mouse Model of Parkinson’s Disease. Int. J. Mol. Sci. 2021, 22, 654. [Google Scholar] [CrossRef] [PubMed]
  152. Sivaji, K.; Kannan, R.R. Polysorbate 80 Coated Gold Nanoparticle as a Drug Carrier for Brain Targeting in Zebrafish Model. J. Clust. Sci. 2019, 30, 897–906. [Google Scholar] [CrossRef]
  153. Sardjono, R.E.; Khoerunnisa, F.; Musthopa, I.; Akasum, N.S.M.M.; Rachmawati, R. Synthesize, characterization, and anti-Parkinson activity of silver-Indonesian velvet beans (Mucuna pruriens) seed extract nanoparticles (AgMPn). J. Phys. Conf. Ser. 2018, 1013, 012195. [Google Scholar] [CrossRef]
  154. Kim, M.J.; Rehman, S.U.; Amin, F.U.; Kim, M.O. Enhanced neuroprotection of anthocyanin-loaded PEG-gold nanoparticles against Aβ1-42-induced neuroinflammation and neurodegeneration via the NF-KB /JNK/GSK3β signaling pathway. Nanomed. Nanotechnol. Biol. Med. 2017, 13, 2533–2544. [Google Scholar] [CrossRef]
  155. Khadrawy, Y.A.; Hosny, E.N.; Magdy, M.; Mohammed, H.S. Antidepressant effects of curcumin-coated iron oxide nanoparticles in a rat model of depression. Eur. J. Pharmacol. 2021, 908, 174384. [Google Scholar] [CrossRef]
  156. Lu, H.; Zhang, S.; Wang, J.; Chen, Q. A Review on Polymer and Lipid-Based Nanocarriers and Its Application to Nano-Pharmaceutical and Food-Based Systems. Front. Nutr. 2021, 8, 783831. [Google Scholar] [CrossRef]
  157. van der Koog, L.; Gandek, T.B.; Nagelkerke, A. Liposomes and Extracellular Vesicles as Drug Delivery Systems: A Comparison of Composition, Pharmacokinetics, and Functionalization. Adv. Healthc. Mater. 2022, 11, 2100639. [Google Scholar] [CrossRef]
  158. Akel, H.; Csóka, I.; Ambrus, R.; Bocsik, A.; Gróf, I.; Mészáros, M.; Szecskó, A.; Kozma, G.; Veszelka, S.; Deli, M.A.; et al. In Vitro Comparative Study of Solid Lipid and PLGA Nanoparticles Designed to Facilitate Nose-to-Brain Delivery of Insulin. Int. J. Mol. Sci. 2021, 22, 13258. [Google Scholar] [CrossRef] [PubMed]
  159. Kasina, V.; Mownn, R.J.; Bahal, R.; Sartor, G.C. Nanoparticle delivery systems for substance use disorder. Neuropsychopharmacology 2022, 47, 1431–1439. [Google Scholar] [CrossRef] [PubMed]
  160. Ross, A.M.; Kennedy, T.; McNulty, D.; Leahy, C.I.; Walsh, D.R.; Murray, P.; Grabrucker, A.M.; Mulvihill, J.J.E. Comparing nanoparticles for drug delivery: The effect of physiological dispersion media on nanoparticle properties. Mater. Sci. Eng. C 2020, 113, 110985. [Google Scholar] [CrossRef] [PubMed]
  161. Kaushik, A.; Jayant, R.D.; Bhardwaj, V.; Nair, M. Personalized nanomedicine for CNS diseases. Drug Discov. Today 2018, 23, 1007–1015. [Google Scholar] [CrossRef] [PubMed]
  162. Li, L.; He, R.; Yan, H.; Leng, Z.; Zhu, S.; Gu, Z. Nanotechnology for the diagnosis and treatment of Alzheimer’s disease: A bibliometric analysis. Nano Today 2022, 47, 101654. [Google Scholar] [CrossRef]
  163. Alghamdi, M.A.; Fallica, A.N.; Virzì, N.; Kesharwani, P.; Pittalà, V.; Greish, K. The Promise of Nanotechnology in Personalized Medicine. J. Pers. Med. 2022, 12, 673. [Google Scholar] [CrossRef]
  164. Foulkes, R.; Man, E.; Thind, J.; Yeung, S.; Joy, A.; Hoskins, C. The regulation of nanomaterials and nanomedicines for clinical application: Current and future perspectives. Biomater. Sci. 2020, 8, 4653–4664. [Google Scholar] [CrossRef]
  165. EMA. Withrawal Assessment Report for Sinerem (EMEA/CHMP/11527/2008); European Medicines Agency: London, UK, 2008. [Google Scholar]
  166. National Library of Medicine. ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ (accessed on 20 November 2023).
  167. Herdiana, Y.; Wathoni, N.; Shamsuddin, S.; Muchtaridi, M. Scale-up polymeric-based nanoparticles drug delivery systems: Development and challenges. OpenNano 2022, 7, 100048. [Google Scholar] [CrossRef]
Figure 1. Most common types of nanoparticles and functionalization strategies for brain disorder diagnostics and treatment. Created using BioRender.com (accessed on 15 October 2023).
Figure 1. Most common types of nanoparticles and functionalization strategies for brain disorder diagnostics and treatment. Created using BioRender.com (accessed on 15 October 2023).
Pharmaceuticals 16 01721 g001
Figure 2. Transport mechanism across the BBB. Created using BioRender.com (accessed on 20 November 2023).
Figure 2. Transport mechanism across the BBB. Created using BioRender.com (accessed on 20 November 2023).
Pharmaceuticals 16 01721 g002
Table 2. Examples of drugs loaded into exosomes for neurodegenerative disorder treatment.
Table 2. Examples of drugs loaded into exosomes for neurodegenerative disorder treatment.
Target/DiseaseDrug(s)Nanoparticle Type CompositionFunctionalizationDiameter of the Optimized Formulation(s) (nm)ObservationsRef.
ADCurcuminExosomeMacrophage RAW264.7 cells treated with curcumin Exosome inherited the lymphocyte function-associated antigen 1 from their parental cells, a protein that interacts with the endothelial intercellular adhesion molecule 1 117Exosome improved the solubility and bioavailability of curcumin and increased drug penetration across the BBB. Exosomes derived from curcumin-treated RAW264.7 cells relieved the symptoms of AD by inhibiting phosphorylation of the tau protein via activating the AKT/GSK-3β pathway.[77]
ADQuercetinExosomeIsolated from whole blood of SD ratsInherited heat shock protein 70150The bioavailability of quercetin was enhanced via loading into the exosome, relieving the symptoms of AD in okadaic-acid-induced AD mice, by inhibiting the cyclin-dependent-kinase-5-mediated phosphorylation of tau and reducing the formation of insoluble neurofibrillary tangles (NFTs). [78]
ADGAPDH small interfering RNAExosomeBone marrow from inbred C57BL/6 mice. Dendritic cells
with granulocyte/macrophage-colony stimulating factor were selected
Three different peptides—RVG
peptide, a muscle-specific peptide (MSP), and a FLAG
epitope—were cloned into Lamp2b
80Intravenously injected RVG-targeted exosomes delivered glyceraldehyde 3-phosphate dehydrogenase siRNA specifically to neurons, microglia, and oligodendrocytes in the brain, resulting in strong mRNA and protein knockdown of BACE1.[79]
PDDopamine (DA)ExosomeBlood samples from the orbit venous plexus of Kunming mice containing CD9, CD63, and CD81 marker proteins-40–200
(70% in the range of 70–100)
In vitro and in vivo studies demonstrated the successful delivery of DA to the brain using exosomes, including the striatum and substantia nigra. Brain distribution of DA increased >15-fold by using the blood exosomes as a delivery system. DA-loaded exosomes showed much better therapeutic efficacy in 6-OHDA-treated mice and lower systemic toxicity than free DA after intravenous administration.[80]
PDCatalaseExosomeRaw 264.7 macrophages-100–200Exosomes were readily taken up by neuronal cells in vitro. A considerable number of exosomes was detected in 6-OHDA-treated mice brains following intranasal administration, providing significant neuroprotective effects in in vitro and in vivo models of PD.[81]
PDSynuclein small interfering RNAExosomeMurine dendritic cells harvested from bone marrowExpressing RVG100In normal and in S129D α-syn transgenic mice, the authors detected significantly reduced α-synuclein mRNA and protein levels throughout the brain after treatment with RVG exosomes loaded with siRNA for α-synuclein. [82]
PDDNA aptamer targeting α-synuclein aggregatesExosomeHEK293T cellsExpressing RVG-Lamp2b-The aptamer-loaded RVG exosomes significantly reduced the α-synuclein preformed fibril-induced pathological aggregates and rescued synaptic protein (synapsin II and SNAP25) loss and neuronal death. Additionally, intraperitoneal administration of these exosomes into mice treated intra-striatally with α-synuclein preformed fibrils reduced the pathological α-synuclein aggregates and improved motor impairments.[83]
Alzheimer’s disease (AD), amyloid beta (Aβ), AKT/glycogen synthase kinase-3β (AKT/GSK-3β), beta-secretase (BACE1), blood–brain barrier (BBB), dioleoyl-3-trimethylammonium-propane chloride (DOTAP), 6-hydroxydopamine (6-OHDA), human kidney embryonic cell (HEK293), muscle-specific peptide (MSP), neurofibrillary tangles (NFTs), Parkinson’s disease (PD), rabies virus glycoprotein peptide (RVG), gene associated with brain diseases (SNAP25), α-syn transgenic mice (S129D), synaptosome-associated protein 25 (SNAP25).
Table 3. Examples of drugs loaded into solid lipid nanoparticles (SLN) for neurodegenerative and neuropsychiatric disorder treatment.
Table 3. Examples of drugs loaded into solid lipid nanoparticles (SLN) for neurodegenerative and neuropsychiatric disorder treatment.
Target/DiseaseDrug(s)Nanoparticle Type CompositionFunctionalizationDiameter of the Optimized Formulation(s) (nm)ObservationsRef.
ADNicotinamideSLNStearic acid and phospholipon® 90 G (as the oil phase) and sodium taurocholate and ultrapure water (as the water phase)Polysorbate 80, phosphatidylserine or phosphatidic acid 112, 124, 137The results of Morris water maze and also histopathological and biochemical tests demonstrated the effectiveness of intraperitoneal injection of phosphatidylserine-functionalized SLNs in improving cognition, preserving the neuronal cells, and reducing tau hyperphosphorylation in a rat model of AD.[84]
ADGalantamine hydrobromideSLNCompritol (glyceryl behnate), pluronic F-127 (250 mg) as surfactant and Tween 80 as co-surfactant-88–221In vivo assays demonstrated a significant memory restoration capability in cognitive deficit rats in comparison with free drug. The developed carriers offered approximately twice the bioavailability of the free drug.[85]
ADQuercetinSLNCompritol and Tween 80 as the surfactant-159In all the in vivo behavioral (spatial navigation task, elevated plus maze paradigm, gross behavioral activity) and biochemical (lipid peroxidation, glutathione levels, and nitrite levels) experiments, the rats treated with SLN-encapsulated quercetin showed markedly better memory retention than free quercetin-treated rats.[86]
ADPiperineSLNGlycerol mono stearate, Epikuron 200, Tween 80, Tween 20Polysorbate-80 coating312Ibotenic acid-stimulated lesions of basalis magnacellularis were developed in albino Wister rats. SLNs containing piperine reduced the SOD values, increased the AChE values, reduced immobility in the forced swimming test, and showed superior results than donepezil. Histopathology studies revealed reduced plaques and tangles.[87]
ADEpigallocatechin-3-O-gallate (EGCG)SLN--50Nano-EGCG more than doubled the oral bioavailability of EGCG in rats and also was more effective at promoting α-secretase activity in vitro.[88]
ADCurcuminSLNSoy lecithin and waterPolysorbate 80135Young male Lacca mice were treated with AlCl3 to induce alterations in the brain histopathology, loss of cognition, and oxidative damage. Behavioral assessments using a Morris water maze test, biochemical measurements (AChE activity, lipid peroxidation, reduced glutathione, superoxide dismutase, catalase, blood lipid profile), and histopathological analysis were performed. Curcumin SLNs completely reversed the induced alterations. [89]
PDApomorphineSLNGlyceryl monostearate (GMS) or polyethylene glycol monostearate (PMS)-155, 636-OHDA-lesioned rats were orally administered with apomorphine. The total number of rotations increased from 20 to 94 and from 20 to 115 when the drug was administered from SLNs containing GMS and PMS, respectively.[90]
PDRopinirole hydrochlorideSLNPluronic F68, stearylamine-66Physicochemical characterization and pharmacodynamic studies.[91]
PDNaringeninSLNGlycerol monostearate, Tween 80, and F68 non-ionic surfactant-135Neuroprotective activity of naringenin–SLN was evaluated using a rotenone-induced PD model in Wistar rats. Results of behavioral observations (in the Rota rod test) and biomarkers (reduced gluthatione, SOD, CAT, lipid peroxidation) showed that naringenin loaded SLN can exert neuroprotective effects.[92]
Depression/anxietyFluoxetineSLNPrecirol®, lauroglycol™, tween80-128–158The intranasal delivery of the optimal lipid nanoparticle formulation reduced both depressive and anxiety-like behaviors in adult CD1 mice (assessment using a mice marble-burying test (MBT) and mice forced swimming test (FST))[93]
Alzheimer’s disease (AD), acetylcholinesterase (AChE), catalase (CAT), epigallocatechin-3-O-gallate (EGCG), mice forced swimming test (FST), Glyceryl monostearate (GMS), 6-hydroxydopamine (6-OHDA), mice marble-burying test (MBT), Parkinson’s disease (PD), polyethylene glycol monostearate (PMS), superoxide dismutase (SOD), solid liquid nanoparticle (SLN).
Table 4. Examples of drugs loaded into nanoemulsions (NEs) for neurodegenerative and neuropsychiatric disorder treatment.
Table 4. Examples of drugs loaded into nanoemulsions (NEs) for neurodegenerative and neuropsychiatric disorder treatment.
Target/DiseaseDrug(s)Nanoparticle Type CompositionFunctionalizationGlobule Size of the Optimized Formulation(s) (nm)ObservationsRef.
ADMemantine hydrochlorideNELabrasol, Tween 20, PEG-~11NE displayed antioxidant activity (FRAP and DPPH), and 98% cell viability in the Neuro 2a cell line. Biodistribution results showed NE uptake in the brains of rats when administered intranasally.[97]
ADDonepezilNELabrasol, cetyl pyridinium chloride, glycerol-65Cell viability in the Neuro 2a cell line was 76.3% for the NE and 85% for donepezil solution. In vivo assays showed that rats intranasally administered with the NE showed maximum distribution in the brain and it remained in the target site until 24 h, while no brain uptake was seen in the rats orally administered with the NE. The rats intravenously administered with the NE or intranasally administered with donepezil solution showed only trace amounts of drug distribution in the brain. [98]
ADHuperzine ANEIsopropylmyristate, Capryol 90, Cremophor EL + LabrasolLactoferrin (Lf)15 (without Lf), 17 (with Lf)Lf–huperzine A-NE showed better uptake for the hCMEC/D3 cell line than unmodified huperzine A-NE. Similar results were found in in vivo assays.[99]
ADResveratrolNECoconut oil, pluronic and cremophor EL-111In Wistar rats, the brain-targeted efficiency was higher after intranasal administration of resveratrol NE (2 mg/kg) compared with that of resveratrol suspension.[100]
ADCurcumin + resveratrolNELabrafac lipophile/cremophor 40, with hyaluronic acid 115 NEs displayed antioxidant activity (DPPH method). Moreover, the integrity of the lining epithelium of the nasal cavity was maintained after repeated administration of hyaluronic-acid-based NEs co-encapsulating curcumin and resveratrol, with no inflammatory cellular infiltration in the lamina propria of the mucosal lining. NEs successfully increased the amount of both polyphenols in the brain of male Albino rats.[101]
PDSelegilineNEGrape seed oil, Sefsol 218®, Tween 80, lauroglycol 90-61Behavior studies (FST, locomotor activity test, catalepsy, muscle coordination test, akinesia test, pole test) were undertaken. Rats were treated with haloperidol to induce PD. Selegiline NE (administered intranasally) showed significant improvement in behavioral activities in comparison to the orally administered free drug.[102]
PDQuercetinNECapmul MCM NF and cremophor RH 40-~50In vivo results show that quercetin-loaded NEs potentially reduced α-synuclein aggregation, increased mitochondrial and fat content, and improved the lifespan in transgenic C. elegans strain NL5901. Moreover, quercetin NEs significantly downregulated the reactive oxygen species (ROS) levels in wild-type C. elegans strain N2 more efficiently than pure quercetin.[103]
PDNaringeninNEVitamin E: Capryol 90 (1:1), Tween 80, transcutol-HP, and water-39Behaviors in 6-OHDA-treated rats were successfully reversed after intranasal administration of naringenin NE along with the levodopa. Naringenin NE + levodopa also induced higher levels of GSH and SOD and lower levels of MDA.[104]
AnxietyClove volatile oil (contains eugenol)NECoconut oil, polysaccharides from Agaricus blazei Murill mushroom, Tween 80, and water-227 to 333Clove volatile oil (CVO), eugenol, a CVO NE, and an empty NE presented low acute toxicity in zebrafish. The CVO NE reduced the anxious-like behavior of adult zebrafish without affecting their locomotor activity. However, CVO and eugenol displayed anxiolytic activity but reduced animal locomotion similarly to benzodiazepines (diazepam). The observed anxiolytic activity of the CVO, eugenol, and CVO NE is linked to the GABAergic pathway.[105]
DepressionFluoxetineNECapmul MCM, labrasol, transcutol-P--Nasal ciliotoxicity studies were performed to evaluate any potential toxic effects of excipients used in the NE formulation on the nasal mucosa. The blank NE displayed no damage to nasal mucosa showing their potential for intranasal delivery.[106]
DepressionQuetiapine fumarateNECapmul MCM, Tween 80, transcutol P, PEG, water-144In vivo studies were performed in male Wistar rats after intravenous and intranasal administration of quetiapine pure drug and an NE containing quetiapine. Superiority of the NE for intranasal delivery was observed.[107]
DepressionParoxetineNECapmul MCM, Solutol HS 15, and PEG-59Permeation studies revealed increased permeation of the paroxetine NE (2.57-fold) compared to the paroxetine suspension. Behavioral studies (FST and locomotor activity test) in Wistar rats showed that treatment with paroxetine NE (administered intranasally) significantly improved the behavioral activities in comparison to paroxetine suspension (orally administered). The NEs were also effective in counterbalancing oxidative stress.[108]
Alzheimer’s disease (AD), clove volatile oil (CVO), 2,2-diphenyl-1-picrylhydrazyl (DPPH), Ferric-Reducing Antioxidant Power Assay (FRAP), forced swimming test (FST), gamma-aminobutyric acid (GABA), reduced glutathione (GSH), lactoferrin (Lf), nanoemulsion (NE), 6-hydroxydopamine (6-OHDA), polyethylene glycol (PEG), Parkinson’s disease (PD), reactive oxygen species (ROS), superoxide dismutase (SOD).
Table 5. Examples of drugs loaded into polymeric nanoparticles for neurodegenerative and neuropsychiatric disorder treatment.
Table 5. Examples of drugs loaded into polymeric nanoparticles for neurodegenerative and neuropsychiatric disorder treatment.
Target/DiseaseDrug(s)Nanoparticle Type CompositionFunctionalizationDiameter of the Optimized Formulation(s) (nm)ObservationsRef.
ADRivastigmineChitosan-based polymeric nanoparticlesChitosan, tri-polyphosphate pentasodiumTween 80®154Compared to both pure rivastigmine and uncoated nanoparticles, Tween 80®-coated nanoparticles induced significant reversal of scopolamine-induced effects. Coated nanoparticles also increased the maximum tolerable dose of rivastigmine. [115]
ADRivastigmineChitosan-based polymeric nanoparticlesChitosan, sodium tripolyphosphate-164Intranasal administration showed higher brain targeting of rivastigmine using chitosan nanoparticles compared to other tested formulations.[116]
ADGalantamineChitosan-based polymeric nanoparticlesChitosan, sodium tripolyphosphateTween 80®48–68Nasal administration of chitosan nanoparticles exhibited a significant decrease in the AChE protein level and activity in rat brains compared to the oral and nasal free galantamine solutions.[117]
ADTacrineChitosan-based polymeric nanoparticlesChitosan--The application of tacrine-loaded chitosan nanoparticles selectively increased the tacrine concentration in the brain tissue. Tacrine-loaded non-magnetic and tacrine-loaded magnetic chitosan nanoparticles improved spatial learning and memory after streptozotocin treatment in Wistar rats, with magnetic nanoparticles being the most effective. Tacrine-loaded chitosan nanoparticles increased seladin-1 and reduced APP gene expression.[118]
ADCurcuminChitosan-based polymeric nanoparticlesChitosan Bovine serum albumin (BSA)144Curcumin-loaded chitosan/BSA nanoparticles effectively increased drug penetration through the BBB, promoted the activation ofmicroglia, and further accelerated the phagocytosis of the Aβ peptide. They also inhibited the TLR4-MAPK/NF-κB signaling pathway and further downregulated M1 macrophage polarization.[119]
ADPiperineChitosan-based polymeric nanoparticlesChitosan, sodium tripolyphosphate, poloxamer 188 -249Piperine nanoparticles could significantly improve cognitive functions as efficiently as the standard drug (donepezil injection) with additional advantages of a dual mechanism of action (AChE inhibition and antioxidant effect). [120]
ADThymoquinoneChitosan-based polymeric nanoparticlesChitosan, sodium tripolyphosphate-172–281Intranasal thymoquinone-loaded nanoparticles were more effective in brain targeting compared to intravenous and intranasal thymoquinone solutions.[121]
ADMemantinePLGA-based polymeric nanoparticlesPoly(lactic-co-glycolic acid) (PLGA)PEG153Nanoparticles were able to cross the BBB. Behavioral tests in transgenic APPswe/PS1dE9 mice demonstrated that nanoparticles decreased memory impairment in comparison to the free drug solution. Memantine–PEG–PLGA nanoparticles reduced Aβ plaques and the associated inflammation characteristics of AD.[122]
ADBACE1-ASshRNA + D-peptideDendrigraft poly L-lysinesDendrigraft poly-l-lysines, α-malemidyl-ω-N-hydroxysuccinimidyl polyethyleneglycolTwo peptides—RVG-29 and D-peptide97, 110Downregulation of the key enzyme in Aβ formation was achieved by delivering non-coding RNA plasmid. Simultaneous delivery of the therapeutic gene and peptide into the brain led to a reduction in neurofibrillary tangles. Meanwhile, memory loss rescue in AD mice was also observed.[7]
ADCurcuminoidsCyclodextrin2-hydroxypropyl-cyclodextrin--Curcuminoids are rapidly metabolized after intravenous injection into APPSWE/PS1dE9 transgenic mice, and their effect on reducing the plaque load associated with AD may be dependent on the frequency of administration.[123]
ADCrocetinCyclodextrinγ-Cyclodextrin--A water-soluble crocetin-γ-cyclodextrin inclusion complex was nontoxic to normal neuroblastoma cells (N2a cells and SH-SY5Y cells) and AD model cells (7PA2 cells). Furthermore, it showed a stronger ability to downregulate the expression of C-terminus fragments and the level of Aβ in 7PA2 cells compared to the crocetin free drug. Both the inclusion complex and crocetin were able to prevent SH-SY5Y cell death from H2O2-induced toxicity.[124]
PDRotigotinePLGA-based polymeric nanoparticlesPLGAPEG, lactoferrin122Qualitative and quantitative cellular uptake studies demonstrated that accumulation of rotigotine was greater when using lactoferrin coating. In addition, intranasal delivery of rotigotine was much more effective with lactoferrin-coated nanoparticles than with uncoated nanoparticles. Rotigotine concentration was higher in the striatum, the primary region affected in PD.[125]
PDRasagilineChitosan coated PLGA- polymeric nanoparticlesPLGAChitosan122Intranasal delivery of mucoadhesive nanocarrier showed significant enhancement of bioavailability in brain, after administration of the rasagiline-chitosan-PLGA-nanoparticles which could be a substantial achievement of direct nose to brain targeting in PD therapy and related brain disorders. [126]
PDPuerarinPLGA-based polymeric nanoparticless-PLGAD-α-tocopherol poly(ethylene glycol)1000 succinate 88Relative to puerarin alone, encapsulated puerarin exhibited significantly improved cellular internalization, permeation, and neuroprotective effects in zebrafish, rats, and MPTP-treated mice. [127]
PDL-DOPACyclodextrinβ-cyclodextrin, 2-hydroxypropyl-β-cyclodextrin---[128]
AnxietyBuspirone hydrochlorideChitosan-based polymeric nanoparticlesthiolated
chitosan, chitosan, sodium tripolyphosphate, sodium
alginate
-227The brain concentration achieved after intranasal administration of buspirone/chitosan nanoparticles was higher than that achieved with intravenous or with intranasal administration of free buspirone.[129]
AnxietyQuercetinPolycaprolactone-based polymeric nanoparticlesPolycaprolactone, Span 80, Capryol 90, Tween 80® or poloxamer 188228Behavioral tests demonstrated the superiority of quercetin-loaded polymeric nanocapsules administered intranasally compared to quercetin solution administered both orally and intranasally.[130]
DepressionAgomelatinePLGA-based polymeric nanoparticlesPLGA, poloxamer 407-116Pharmacodynamic studies showed a significant reduction in immobility time in forced swimming tests in rats intranasally treated with the formulation, which indicated the antidepressant activity of the formulation.[131]
DepressionVenlafaxinePLGA-based polymeric nanoparticlesPLGA Transferrin or a specific peptide against transferrin receptor230In vivo studies showed that non-functionalized nanoparticles reached the brain more efficiently than functionalized ones after intranasal administration. Probably, plain NPs travel via the direct nose-to-brain route whereas functionalized NPs reach the brain via receptor-mediated endocytosis.[132]
DepressionVenlafaxineChitosan-based polymeric nanoparticlesChitosan, sodium tripolyphosphate-168The higher drug transport efficiency and intranasal direct transport percentage of venlafaxine/chitosan nanoparticles compared to other formulations (non-encapsulated venlafaxine via oral and intranasal route) suggest their better efficacy in the treatment of depression.[133]
DepressionVenlafaxineAlginate and chitosan-based polymeric nanoparticlesAlginate, chitosan -174Intranasal administration of venlafaxine-alginate nanoparticles delivered greater amounts of venlafaxine to the brain in comparison to the free drug solution.[134]
DepressionDesvenlafaxinePLGA-chitosan polymeric nanoparticlesPLGA, chitosan-173The optimized desvenlafaxine-loaded PLGA/chitosan nanoparticles on intranasal administration significantly reduced the symptoms of depression and enhanced the level of monoamines in the brain in Wistar rats in comparison with orally administered desvenlafaxine. Intranasal delivery of desvenlafaxine PLGA/chitosan nanoparticles also enhanced the pharmacokinetic profile of desvenlafaxine in the brain.[135]
DepressionSelegiline hydrochlorideChitosan-based polymeric nanoparticles Thiolated chitosan-215Forced swimming and tail suspension tests were used to evaluate the antidepressant activity, in which elevated immobility time was found to be reduced upon treatments. Thiolated chitosan nanoparticles seem to be promising candidates for nose-to-brain delivery in the evaluation of antidepressant activity.[136]
DepressionSertraline hydrochlorideCyclodextrinβ-Cyclodextrin--Physicochemical characterization of the nanoparticles.[137]
DepressionFluoxetine hydrochlorideCyclodextrinβ-Cyclodextrin--Physicochemical characterization of the nanoparticles.[137]
Alzheimer’s disease (AD), amyloid beta (Aβ), amyloid precursor protein (APP), , acetylcholinesterase (AChE), blood–brain barrier (BBB), bovine serum albumin (BSA), Cellosaurus cell line CHO 7PA2 (7PA2), 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), nanoparticles (NPs), Parkinson’s disease (PD), polyethylene glycol (PEG), poly(lactic-co-glycolic acid) (PLGA), rabies virus glycoprotein peptide (RVG), neuroblastoma cell line (SH-SY5Y), Toll-like receptor 4-MAP kinase/ nuclear factor kappa (TLR4-MAPK/NF-κB).
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

Grosso, C.; Silva, A.; Delerue-Matos, C.; Barroso, M.F. Single and Multitarget Systems for Drug Delivery and Detection: Up-to-Date Strategies for Brain Disorders. Pharmaceuticals 2023, 16, 1721. https://doi.org/10.3390/ph16121721

AMA Style

Grosso C, Silva A, Delerue-Matos C, Barroso MF. Single and Multitarget Systems for Drug Delivery and Detection: Up-to-Date Strategies for Brain Disorders. Pharmaceuticals. 2023; 16(12):1721. https://doi.org/10.3390/ph16121721

Chicago/Turabian Style

Grosso, Clara, Aurora Silva, Cristina Delerue-Matos, and Maria Fátima Barroso. 2023. "Single and Multitarget Systems for Drug Delivery and Detection: Up-to-Date Strategies for Brain Disorders" Pharmaceuticals 16, no. 12: 1721. https://doi.org/10.3390/ph16121721

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