Next Article in Journal
Structure and Formation of Z-DNA and Z-RNA
Previous Article in Journal
Novel 9-Benzylaminoacridine Derivatives as Dual Inhibitors of Phosphodiesterase 5 and Topoisomerase II for the Treatment of Colon Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Management of Brain Cancer and Neurodegenerative Disorders with Polymer-Based Nanoparticles as a Biocompatible Platform

by
Mehdi Bazi Alahri
1,
Alhawarin Jibril Ibrahim
2,
Mahmood Barani
3,*,
Hassan Arkaban
4,
Seyedeh Malahat Shadman
4,
Soodeh Salarpour
5,
Payam Zarrintaj
6,
Javad Jaberi
4 and
Abduladheem Turki Jalil
7
1
Department of Clinical Psychology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1971653313, Iran
2
Department of Chemistry, Faculty of Science, Al-Hussein Bin Talal University, Ma’an 71111, Jordan
3
Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman 7616913555, Iran
4
Department of Chemistry, University of Isfahan, Isfahan 8174673441, Iran
5
Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7616913555, Iran
6
School of Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, OK 74078, USA
7
Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla 51001, Iraq
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(2), 841; https://doi.org/10.3390/molecules28020841
Submission received: 7 December 2022 / Revised: 27 December 2022 / Accepted: 10 January 2023 / Published: 14 January 2023
(This article belongs to the Special Issue The Application of Nanomaterials in Cancer Diagnostics and Therapy)

Abstract

:
The blood–brain barrier (BBB) serves as a protective barrier for the central nervous system (CNS) against drugs that enter the bloodstream. The BBB is a key clinical barrier in the treatment of CNS illnesses because it restricts drug entry into the brain. To bypass this barrier and release relevant drugs into the brain matrix, nanotechnology-based delivery systems have been developed. Given the unstable nature of NPs, an appropriate amount of a biocompatible polymer coating on NPs is thought to have a key role in reducing cellular cytotoxicity while also boosting stability. Human serum albumin (HSA), poly (lactic-co-glycolic acid) (PLGA), Polylactide (PLA), poly (alkyl cyanoacrylate) (PACA), gelatin, and chitosan are only a few of the significant polymers mentioned. In this review article, we categorized polymer-coated nanoparticles from basic to complex drug delivery systems and discussed their application as novel drug carriers to the brain.

Graphical Abstract

1. Introduction

Technology and medicine have gone hand in hand for many years. Consistent advances in pharmaceuticals and the medical field have saved millions of lives and improved many others [1]. Neurodegenerative diseases (NDs) are dangerous diseases that are defined as progressive neuronal damage in some regions of the nervous system which affect cognitive function, motor activity, and mental damage and consequently become irrecoverable neuronal loss [2,3,4,5]. Nowadays, we witness longevity growth in developed countries due to improving hygiene and quality of life, which is closely associated with remarkable growth in the prevalence of neurodegenerative (ND) illnesses in the older population. Neuroinflammation, stress, mitochondrial dysfunction, and metabolic problems are also prevalent reasons for the pathogenesis of ND diseases. Hence, they will have adverse emotional, social, and financial burdens on the healthcare system in the future. Accordingly, efforts to find an effective theranostic strategy have been focused on continuing to treat ND disorders at an earlier stage, which would enable the prevention of cognitive impairment and irreversible neuronal damage [6,7].
There are essential interfaces between the central nervous system (CNS) and the enclosed cells and blood vessels, in which endothelial cells are connected by tight junctions (TJ) and adherens junctions (AJ) [8]. The main function of the blood–brain barrier (BBB) consists of protecting neuron cells and maintaining the brain’s internal environment. Therefore, BBB breakdown assists the entrance into the brain of neurotoxic blood-derived products, cells, and pathogens and is related to immune responses which lead to various neurodegenerative issues. The main reason for this problem in the body is that many physiological obstacles, especially BBB, hamper effective therapeutic substances from reaching the site of action [9].
Systemic chemotherapy (using the free drug) is currently the main strategy for the treatment of cancer. The systemic chemotherapy method has many drawbacks including fast blood clearance, poor bioavailability, and low tumor targeting. To overcome these drawbacks, a high drug dose should be administered to cause the required anti-tumor effect which, in turn, causes severe adverse side effects. Therefore, some superparticles and drug delivery systems have been developed to enhance the accumulation of the therapeutic as well as the diagnostic agents in the tumors, decrease their side effects, and improve their efficiency, stability, polarity, and bioavailability [10,11].
Nanotechnology research has found applications in many fields, from medicine and drug research to aeronautics and automobiles [12,13]. Active nanotech-based research has yielded several new advancements with wide-ranging applications [14,15]. New therapeutic efforts have been concentrated on the design of nanocarriers like polymeric nanoparticles [16,17], inorganic nanoparticles [18,19,20,21], green synthesized NPs [22,23,24], liposomes [25], nanofibers [26], hydrogels [27,28,29], Niosomes [30,31,32], and microemulsions [33,34] to transport therapeutic and diagnostic agents. In light of this fact, the above-mentioned nanoparticles (NPs) have attracted much attention thanks to their relatively high drug loading ability, lower or no systematic toxicity, controlled drug release, extended blood circulation time, capability of targeted delivery, and physical–chemical stability [35,36,37,38]. However, these properties for drug delivery rely on the type, size, surface chemistry, and polarity of the particles (Figure 1). On the other hand, molecule transformation is closely controlled by entering the brain parenchyma, then NPs penetrate the neuron cells by reaching the BBB, attending the surfactant in NP component and disrupting the TJ [39,40,41,42].
More importantly, the use of biopolymers in medical treatment has been regarded to make vigorous principles for peptides and proteins, as well as genes [44]. NPs coated in biopolymers in drug delivery permit them to simply penetrate the cells and release insoluble drugs to prevent their degradation. NPs can also be loaded with drugs by encapsulation within the polymer matrix adsorption, increasing the efficient loading and release of drugs in cells [45,46,47,48].
In this review, we try to focus on the problems of the BBB which appear in neurodegenerative diseases and how to design biocompatible polymer-coated and colloidally stable NPs to cure these disorders.

2. Brain Disorders

The World Health Organization (WHO) has recently declared that brain disorders are one of the primary morality causes in the world, mainly stemming from brain cancer or neurodegenerative diseases [49]. As most brain disorder diagnoses are highly invasive, time consuming, and expensive, it is imperative to develop non-invasive, cost-effective, and efficient tools for brain disorder characterization to reduce brain damage and improve longevity [50]. Surely, drug delivery in brain-based NPs coated in polymers opens a new avenue for solving these problems [51,52].

2.1. Brain Cancer

Tumors of the CNS are one of the challenges in medical care. They are typically located in the brain, which is a primary metastatic site for other organs’ tumors. The most prevalent types of brain tumors are gliomas. The different cell phenotypes of glia (astrocytes, oligodendrocytes, microglia, ependymal cells) can be the origin of gliomas. Based on the degree of cancer aggressiveness, gliomas are categorized from grade I to IV, in which the most malignant is IV [53]. Glioblastoma multiforme (GBM) is one of the most dangerous brain tumors in the world and kills many humans every year. The heterogeneity of the cellular and molecular properties of GBM complicates pharmacological interventions. GBM is a very invasive and recurring disease; furthermore, its clinical prognosis is poor, and finally, its mortality rates are high. Conventional therapeutic approaches like radiotherapy, chemotherapy, and surgical resection cannot be completely effective. The median overall survival time remains just 12–15 months after diagnosis, with less than 5% of people surviving longer than five years [54]. The complexity and delicacy of the structure and the primary function of the brain limit complete surgical resection. Due to the infiltrative growth behavior of GBM cells, the residual tumor cells can grow in the surrounding brain tissue. As the BBB protects these infiltrative cells from chemotherapy, resulting in high rates of cancer recurrence. Despite the conventional therapeutic regimen, alternative and targeted drug delivery are very essential to achieve better treatments for brain cancers [55].

2.2. Neurodegenerative Diseases

Neurodegenerative diseases are a group of neurological disorders in which the nervous function and anatomy in which both the peripheral and central nervous systems are gradually destroyed. These diseases are age-related; therefore, the number of cases is expected to rise as lifespans in many countries continue to increase. As the world grows older, unfortunately, the number of people with neurodegenerative diseases will increase. Furthermore, the importance of these diseases quickly becomes more serious than other diseases. They have relatively long durations and high treatment costs, so neurodegenerative diseases become a serious challenge for both patients and community healthcare. The etiology of these disorders is complex, but combinations of genetic factors and environmental conditions as contributing causes may play roles in neuronal dysfunctions. Alzheimer’s disease (AD) and Parkinson’s disease (PD) are two of the most common [56,57].

2.2.1. Alzheimer’s Disease (AD)

Alzheimer’s disease (AD) is a neurological illness that induces progressive dementia. It is a progressive, long-term, and age-related neurodegenerative illness that manifests symptoms in three stages: severe, mild, and moderate dementia. Short-term memory loss is typical in AD, which is followed by cognitive decline, mental illnesses, behavioral abnormalities, and coordination problems, finally leading to death [7].
Many aspects of the neuropathogenesis of AD have not yet been completely elucidated due to the diversity of AD pathogenesis. Numerous clinical data suggested hypotheses concerning AD, which is a progressive accumulation of protein aggregation in the brain that causes neurodegeneration [58]. The Aß amyloid protein cascade hypothesis, oxidative stress theory, mitochondrial dysfunction, tau protein hyperphosphorylation theory, and the neuroinflammatory response are among the ideas hypothesized for the neuropathogenesis of AD. Other pathogenic pathways for AD have been identified as melatonin deficit, metal ion metabolism disturbance, cholinergic damage, immunological abnormalities, and so on [7,58,59]. The hypothesis of AD pathogenesis asserting that the impairment of cholinergic neurotransmitter systems may be the cause of the suppression of acetylcholine by Acetylcholinesterase (AChE) activity and the activation of the glutamatergic system has been considered to develop drugs based on these mechanisms [60].

2.2.2. Parkinson’s Disease (PD)

Parkinson’s disease (PD) is the second most common progressive neurodegenerative disease. The pathophysiology of PD in human postmortem studies recognizes the neurodegeneration or death of neurons in locus ceruleus, the substantia nigra par compacta, and other neuronal populations. Based on the results of scientists’ research, the environment/lifestyle, and genetics (PARK-SNCA, PARK-PArkiN, etc.), several molecular events and hallmarks are considered the main causes of this disease. Mitochondrial dysfunction, α-synuclein misfolding, neuroinflammation, oxidative stress, aggregation, and impaired calcium homeostasis are involved in the pathogenesis of PD. Lewy bodies and Lewy neurites are intracellular disease-related protein aggregates and neuropathological hallmarks of PD. They are mostly present in pigmented neurons in the substantia nigra; however, they have been found in other central and peripheral neuronal populations [61,62].
The neuronal dysfunctions appearing in PD present with motor and non-motor symptoms. The main motor symptoms are tremors at rest, muscle rigidity, and bradykinesia, which can be related to the damage of dopaminergic neurons. On the other hand, the loss of non-dopaminergic neurons causes other motor symptoms relating to walking, balance, and posture. The quality of life in Parkinson’s patients is significantly reduced due to these various complications. Besides these motor symptoms, the non-motor symptoms affect the PD patients’ life, such as their mood conditions, sleep disorders, cognitive dysfunction, and dysautonomia [7].

3. The Challenge of the BBB

The application of drugs used for brain disorders is restricted, since they must overcome barriers. The BBB is a vasculature of the CNS separating the circulating blood from the brain’s extracellular fluid. The BBB is a semi-permeable and highly selective membrane barrier that imposes various obstacles to the transfer of many molecules. The BBB consists of a continuous layer of endothelial cells joined by tight junctions, transport proteins (efflux pumps), and extracellular and intracellular enzymes in the brain parenchyma which are called the physical barrier, transport barrier, and, enzymatic barrier, respectively [63]. This structure can control the transport of all substances and differentiate between molecules with specified components. Some essential substances such as glucose, amino acids, and hormones are transferred by transporters through the blood to the brain and vice versa. The influx of substances into the brain’s capillary endothelial cells and choroid plexus epithelial cells is facilitated by uptake transporters, while the materials are exported from the cells by efflux transporters. The efflux transporters on the blood-facing membrane of the BBB can be a threat to return drugs to blood circulation [7].
In transporting drugs to the brain, many factors influence the delivery and effectiveness of therapeutic agents through the BBB. For example, if drug molecules bind to non-transporters, they cannot be effective and pass through the BBB. Furthermore, the solubility and the molecular weight of each structure have an important role, since the lipophilic low-molecular-weight molecules that are not ionized at physiological pH can cross the BBB by diffusion. Moreover, other various factors could be effective for the delivery of drugs, e.g., the membrane or luminal surface of the brain capillary, the composition of cerebrospinal fluid (CSF) or interstitial fluid, and the functional groups or surface charge of therapeutic structures, so the prediction of drug transportation through the BBB may be very complicated [64].
According to the brain’s protective mechanisms and the importance of drug transportation to this organ, various strategies have been studied to overcome or bypass these obstacles. Therefore, maintaining normal body functions and reducing the post-delivery toxicity of the drugs is highly essential. Increasing the bioavailability and targetability of therapeutic substances in CNS-related diseases could be achieved by drug delivery systems, but some of them demonstrate invasive or nonspecific pathways [7,64].

4. Coating Polymers

Many types of nanocarriers have been developed for treating brain disorders [65]. Polymer-based therapeutic agents have been explored for the treatment of neurodegenerative diseases due to various fascinating advantages of polymers such as great biocompatibility, nontoxicity, controllable degradation rate, tunable architectures, the possibility of multiple interactions between amyloidogenic protein/peptide and polymer, and excellent in vivo stability [66].
In this section, we will look at some of the most commonly used coating polymers for neurodegenerative disorders.

4.1. Polysorbate (PS)

PSs are nonionic synthetic surfactants. This group of surfactants with uncharged head groups is an important subgroup of surfactants made up of poly ethoxy Sorbitan fatty acid esters. PSs have long been used in pharmaceuticals and food additives like emulsifiers and stabilizers [67,68].
PSs have a Sorbitan ring with polyethylene oxide (PEO) attached to hydroxyl groups as their backbone structure. The four hydroxyl groups of the Sorbitan ring are conjugated to various numbers of ethylene oxide subunits. PSs are made chemically in two steps: Sorbitan is ethoxylated, then esterified with fatty acids. Figure 2 depicts the PS synthesis scheme, and Figure 3 shows the type of fatty acid ester connected with the PEO Sorbitan portion of the molecule, with the number following PS. PSs contain hydrophilic PEO segments and hydrophobic fatty acid segments. As a result, amphiphilic PSs can be used as effective surfactants [69,70,71]. PSs have been used as solubilizers and emulsifiers for hydrophobic pharmaceuticals, as well as protein stabilizing agents, and as drug carriers for both hydrophilic and hydrophobic medications [72,73,74,75]. PSs have also pulled in awesome consideration as brain-targeting coating materials that can encourage the transport of drug carriers over the blood–brain barrier (BBB). PSs have various points of interest, such as commercial plenitude and ease of chemical alteration, which render them reasonable for different biomedical applications. Hence, PSs have been broadly utilized as emulsifiers and stabilizers in different drug formulations and nourishment additives [76]. Additionally, PSs exhibit high biocompatibility and low toxicity and are thus advantageous for use as drug carriers. Owing to their amphiphilic nature, PSs have been used as solubilizers of hydrophobic drugs, proteins, and inorganic nanoparticles. Therefore, PS-based drug carriers can carry multiple drugs for combination therapy. Particularly, PS-drug conjugates and PS-coated drug carriers have great potential to treat central nervous system-related diseases because PS coatings can improve the crossing of the BBB [77].
PS 80 (Tween 80) has been used in the formulation of therapeutic monoclonal antibodies due to its unique properties. These properties include low toxicity, good protein stabilization, and biocompatibility. PSs have been used in vivo and in vitro to improve the permeability of various drugs. PS 80 is an emulsifier that facilitates drugs crossing through the BBB [78]. PS80 increases BBB crossing in epithelial BBB cells by adsorbing apolipoprotein onto nanoparticles, resulting in LDL receptor-mediated transcytosis [79,80]. PS 80-coated R-HCl loaded nanoparticles can also be an efficient drug delivery system in the treatment of Parkinson’s disease to revert the neurodegeneration with the strategy of coating the chitosan nanoparticles which enhance the brain targeting of the encapsulated drug [81].

4.2. Polyethylene Glycol (PEG)

PEGs are also known as Macrogols [82]. PEG has become well known due to its great structural flexibility, lack of steric hindrances, amphiphilicity, biocompatibility, and high hydration capacity and has lots on interest in drug delivery applications [83,84]. PEGs have extremely active functional terminals and are electrically neutral at all pH levels [85].
The FDA has approved PEG for intravenous, oral, and cutaneous use in humans [86, 87]. As a result, biocompatibility, circulation time, and aggregation are all improved [88]. PEG has been used to coat various polymeric nanoparticle systems. Surprisingly, the length of the PEG chain has an impact on the penetration of polymeric nanoparticles into the extracellular region of the brain. PEG-coated nanoparticles have recently grabbed great attention in the treatment of AD due to the vast biological efficiencies of PEG [17,89]. PEG is a polymer of choice in drug delivery systems and is popular due to its tunable properties and well-established safety profile. PEG coating of NPs can make a highly suitable strategy for improving efficiency in the systemic delivery of therapeutic agents and studying how the properties of PEG coatings influence NP biodistribution and the ability to penetrate the brain [16,90].

4.3. Chitosan

Chitosan is a linear polysaccharide, and because of its low cost, availability across a wide variety of molecular weights, and biodegradability, it is one of the most commonly used natural polymer nanoparticles for medication delivery [91]. It also has special biological features like anticancer, antibacterial, and antioxidant capabilities [92]. Chitin, a natural polymer derived from crustaceans or fungi, is partially N-deacetylated to synthesize chitosan [93]. Chitosan contains three different kinds of functional groups (amine, primary, and secondary hydroxyl) which could be used to make a variety of chemical modifications. The molecular weight, degree of deacetylation, and chemical modifications can all affect its biodegradability [92]. Chemical cross-linking, ionic gelation, and microfluidic synthesis are all ways that can be used to prepare chitosan nanoparticles [94,95]. These nanoparticles have shown great potential in brain drug delivery because of their positive charge, which increases cell uptake and makes them appropriate for loading with negatively charged therapeutics [96]. As an example, antibody-modified PEG–chitosan nanoparticles showed significant brain absorption, which was attributed to the antibody’s synergy with the positive chitosan charge [97]. Despite this, chitosan nanoparticles have drawbacks, including inadequate drug loading effectiveness on hydrophobic substrates and poor molecular weight control [98]. However, many advantages, including its ability to improve the bioavailability of drugs, efficiency in targeted drug delivery, reducing side effects, increasing drug effects at the target site, and increasing drug stability have attracted many researchers to the design of brain delivery systems in this way. Importantly, chitosan can open TJs with its ability to cross the BBB to provide sufficient chitosan-based DDSs for brain drug delivery applications and represent a better alternative to conventional drug formulations in brain diseases [99,100,101].

4.4. Poly-Ɛ-Caprolactone (PCL)

PCL is a biodegradable, FDA-approved polyester that has been utilized in a variety of applications, including sutures, implants, contraception devices, and drug delivery systems [102,103]. PCL is made up of repeated hexanoate units and can be broken down in the body by hydrolysis into 6-hydroxy caproic acid [104], which can subsequently be converted into adipate [105] and catalyzed to CO2 [106]. The ring-opening polymerization of 𝜖-caprolactone or the condensation polymerization of 6-hydroxyhexanoic acid is used to prepare it. Due to PCL’s insolubility in water, di-block PEG-b-PCL copolymers are commonly used to prepare PCL-based nanoparticles. Standard procedures such as film dehydration, microfluidics, emulsion, and solvent displacement can be used to prepare these nanoparticles [107]. Drug delivery for neurological diseases has also been examined using PCL-based nanoparticles [108]. For example, in an intracranial glioma tumor-bearing in vivo model, peptide-functionalized PEG–PCL micelles demonstrated significantly better transport ratios and increased accumulation in an in vitro BBB model [109]. PCL, on the other hand, has a low degradation rate, making it inappropriate for use as a drug delivery method [110]. Changing the molar mass or coating it with alternative polymers, such as PLA, might alleviate this problem [111]. Hence, it has been used as a suitable coating in the design of the brain delivery system [52,112,113].

4.5. Polyacrylic Acid (PAA)

PAA, also known as poly 1-carboxyethylene, is a high-molecular-weight synthetic polymer produced from acrylic acid monomers. PAA is typically formed with the use of an initiator and free radical polymerization (Figure 4). Poly (1-carboxyethylene) is a low-cost polymer that has been commercialized [114,115,116].
PLA, a common pH-responsive polymer [117], has traditionally been used as a hydrophilic section in amphiphilic or amphipathic block copolymers with a wide range of properties [118]. PAA is an acrylic acid polymer containing a carboxylic group (–COOH) on each monomer unit end which is attached to the vinyl group (Figure 4). PAA, a thermoplastic polymer with many carboxyl groups, has a high bioavailability and can be used as a surface modification for biological nanomaterials [119]. PAA is a biocompatible, nontoxic, and biodegradable polymer that has attracted a lot of attention in recent years [120,121,122]. PAA nano-derivatives can be made by chemically modifying carboxyl groups, and they have better chemical characteristics than untreated PAA. PAA is used in a variety of industries, including adhesives, coatings, packaging, pharmacology, and other medical and biological fields [123]. PAA offers excellent medication storage and delivery capabilities due to its nontoxicity and absorption qualities. According to this, this polymer can be a suitable choice for the brain delivery system [124,125].
Figure 4. Synthesis and chemical structure of poly sodium acrylate (NaPAA) and PAA, adapted from [126], MDPI, 2020.
Figure 4. Synthesis and chemical structure of poly sodium acrylate (NaPAA) and PAA, adapted from [126], MDPI, 2020.
Molecules 28 00841 g004

4.6. Poly (Lactic-co-Glycolic Acid) (PLGA)

PLGA is a type of linear copolymer that can be synthesized in a variety of ratios of the two monomers lactic acid and glycolic acid [127]. The FDA has approved PLGA for medical applications like drug delivery devices and biomaterials. The hydrolytic de-esterification of the PLGA copolymers is followed by the clearing of their monomeric anions, lactate, and glycolate, making them nontoxic and biodegradable [128,129]. The transformation of glycolic acid and lactic acid can alter the rate of degradation, degree of crystallinity, and mechanical strength, and therefore release kinetics and drug loading. Polylactic acid (PLA) is a crystalline hydrophobic polymer because of its methyl side chains, whereas polyglycolic acid (PGA) is a rigid and hydrophilic polymer with low mechanical strength [130]. As a result, PLGA copolymers with a higher PGA:PLA ratio are more hydrophobic, which means they degrade and release drugs at a slower rate [131]. PLGA can be prepared by using several methods: Segmer assembly polymerization [132], ring-opening polymerization [133], and the polycondensation process [131]. PLGA nanoparticles can be prepared from PLGA copolymers, utilizing processes like emulsion, nanoprecipitation, solvent co-evaporation, and spray-drying [134]. Soft lithography can also be used to synthesize non-spherical nanoparticles (e.g., cylindrical shapes) [135]. The terminal carboxylic acid groups can be used to introduce required surface modifications [136]. As a result, a wide range of pharmacological compounds, including anti-inflammatory medicines, antibiotics, chemotherapeutics, and proteins, have been integrated into PLGA nanoparticles [134]. For crossing the BBB, a variety of PLGA formulations have been investigated [137]. PLGA may be a well-known choice as a biodegradable medication carrier. The debasement rate of PLGA and the discharge of typified drugs can be controlled by the physicochemical properties of the polymer such as atomic weight, hydrophilicity, and lactide (LA) to glycolide (GA) proportion. So, PLGA-based nanoparticles have a higher effective half-life, bioavailability, and efficacy, and can be efficiently delivered to the brain by intranasal administration as well [138,139,140].

4.7. Hyaluronic Acid (HA)

Hyaluronic acid (HA), a biocompatible, biodegradable, and chemically adaptable molecule, has received more and more interest in the biomedical community during the past ten years. The extracellular matrix (ECM), which contains a significant amount of HA, is crucial for preserving cellular homeostasis and interaction [141,142]. Although HA is a naturally occurring substance with favorable cell interactions, low immunogenicity, mild antigenic characteristics, and the ability to be chemically modified, it has poor mechanical qualities, is expensive to produce, and is not very reproducible [143,144]. It has been demonstrated that HA is involved in tissue development, embryonic development, angiogenesis, cell migration, and proliferation. With polar and apolar moieties in the polymer structure, HA is naturally hydrophilic and allows for chemical interaction with a variety of chemical agents (Figure 5). Its molecular weight (MW), which in turn relies on the source, determines its structural, physicochemical, degradable, and biological features [145]. The HA-based polymer has been a terrific idea for the examination of tumors, especially for brain tumors, and is ideal for generating a more biocompatible system indicative of a healthy condition. In the CNS, HA plays a significant role in a variety of cell activities, including cell migration, proliferation, differentiation, and others. Medication thickening, the formation of conjugates when HA and its derivatives are utilized as carriers, sustained release, and improved drug targeting have all been demonstrated [146]. Therefore, this polymer may be a promising option for treating neurodegenerative diseases and brain tumors [147].

4.8. Cyclodextrins (CDs)

CDs are cyclic oligosaccharides made from starch by enzymatically cleaving the amylose helix. These ring-shaped molecules are very hydrophilic because their many hydroxyl moieties face outward. On the other hand, due to the glucosidic oxygen linkages, the inner side of the cavity is less hydrophilic. With the help of this structure, CDs can incorporate other less hydrophilic compounds (guests) into the cavity, creating what are known as host–guest inclusion complexes. Because of the CD cavity’s molecular size, the majority of medications, tastes, cosmetic compounds, insecticides, etc., can be molecularly enclosed there. The size of the guest affects the complex’s stoichiometry. Through particular synthetic processes, the many hydroxyl groups are easily changed to create different CD derivatives [148] (Figure 6).
The potential of cyclodextrins to actively remove lipids from cell membranes and to offer an appropriate carrier system for drug delivery has led to an ongoing increase in interest in CDs as therapeutic agents. The goal of creating new CD derivatives is to enhance CD bioavailability, biocompatibility, and therapeutic results. Due to their ability to reduce cell toxicity and hide the bitterness of many active compounds or adjuvants, CDs have the potential to improve medication solubility, bioavailability, and chemical stability. This has been linked to the fact that CDs’ physicochemical structure makes it possible for drug candidates to be trapped inside of their cavities, enabling the administration of hydrophilic or hydrophobic medications into bodily tissue. Many CD derivatives have been created to increase these molecules’ water solubility relative to their natural counterparts [149]. There are two key benefits to the CDs’ capacity to build inclusion complexes. In order to assist their aqueous dissolution, they first aid in solubilizing poorly hydrophobic substances by employing CD as a so-called carrier partner. The hydrophobic interior of the CDs makes it simple to encapsulate hydrophobic molecules of interest, while the hydrophilic outside of the CDs renders the entire complex water soluble. The second benefit of inclusion complex creation is that it significantly alters the properties of the target molecule (in our example, “drugs”). This includes altering the drug’s stability, bioavailability, oral absorption, and interactions with biological membranes and cells. CDs are widely used as drug delivery carriers through nasal mucosae and ocular, dermal, intestinal, and brain barriers because they improve the delivery and bioavailability of hydrophilic, hydrophobic, and lipophilic drugs. CDs have also been widely used to improve biocompatibility and bioavailability when combined with active drug compounds, thereby increasing drug efficacy [150].

4.9. Human Serum Albumin (HSA)

The most prevalent protein in plasma, HSA, is a monomeric multidomain molecule that serves as both the primary moderator of fluid flow across bodily compartments and the primary determinant of plasma oncotic pressure. HSA has a remarkable ability for complex formation, acting as a store and transporter for a variety of endogenous and foreign substances. Indeed, HSA serves as the primary carrier for fatty acids, influences the pharmacokinetics of many medications, allows for the metabolic modification of some ligands, neutralizes potential toxins, makes up the majority of the antioxidant capacity in human plasma, and exhibits (pseudo-)enzymatic properties [151]. HSA is an excellent choice for nanoparticle preparation because it has been demonstrated to be biodegradable, nontoxic, simple to purify, and soluble in water. This makes it easy to administer via injection. It has been demonstrated that adding the right medications to nanoparticles can prevent the pharmacologically active compounds from prematurely degrading or inactivating after injection as well as during storage [152]. 3D structure of HSA is shown in Figure 7.

5. Polymer-Coated Nanoparticles

Nanotechnology is helping to considerably improve, even revolutionize, many technologies and industry sectors. Nanomedicine is the medical application of nanotechnology. Nanomedicine ranges from the medical applications of nanomaterials and biological devices to nanoelectronic biosensors, and even possible future applications of molecular nanotechnology such as biological machines [154,155,156]. Nanoparticles are extensively used to increase the efficacy of a drug and reduce side effects through direct targeting and site-specific controlled delivery [157]. However, due to the toxicity of inorganic nanoparticles, polymers are generally used as nanoparticle carriers to reduce toxicity and increase cellular uptake. Table 1 summarizes some advantages and disadvantages of four common nanoparticles in brain delivery for neurodegenerative disorders.
Even though the already specified drugs are compelling within the treatment of neurodegenerative diseases (NDDs), the blood–brain barrier impedes their treatment and determination given its nearness to the central CNS illness [116,161,162,163]. Due to their low bioavailability and pharmacokinetic characteristics, traditional medicines, such as protein macromolecules, are limited in their therapeutic applicability. Subsequently, the consideration of NPs with neurotrophic impacts has become a modern hotspot within the NDDs study field [126]. Nanotechnology’s rapid growth and wide range of uses necessitate a strategy for creating safe NPs [164]. The architecture of the surface is a key role in the physicochemical properties of metallic particles that influences their mechanism of action in organic or natural environments [10].
Graphene is one of the prominent compounds that is used frequently in the therapy and diagnosis of neurodegenerative disorders. For the better function of graphene, the surface chemistry of graphene was modified with hydrocarbon, hydroxyl, aldehyde, and carboxyl groups and was studied by means of computational chemistry [165]. Paola Ginestra illustrated porous framework generation by electrospinning an arrangement containing PCL and graphene powder with completely different percentages. The effect of graphene on filament shape and mechanical properties in pliable testing was investigated, as well as the effect of electrospun substrate composition on neural stem cell separation. The researchers refined rat stem cells on fibrous platforms to study the effect of filament action on cell morphology and neuron divergence [166]. Rossana Rauti et al. demonstrated that slight graphene oxide nanosheets (s-GO) target specific presynaptic vesicle release. They propose that s-GO chips diminish the accessibility of the transmitter, using its advancing quick release and ensuing exhaustion, driving a decline in glutamatergic neurotransmission. After injecting s-GO into the hippocampus in vivo, ex vivo patch clamp recordings from brain slices revealed a substantial decrease in glutamatergic synaptic activity compared to saline infusions 48 h later [167].
Chlorogenic acid (CGA) is a polyphenolic compound that has potential anti-inflammatory and antioxidative features and has risen as a promising compound in ameliorating NDDs. Owing to its destitute steadiness, bioavailability, and release kinetics, CGA required an appropriate nanocarrier-based pharmaceutical plan for targeting NDDs. Vinayak Agarwal et al. considered the in-silico approval of CGA as a successful therapeutic agent targeting different NDDs, through the creation of a polymeric nanoparticle-based carrier framework to overcome its pharmacological confinements and progress its stability. A fruitful in silico approval was conducted utilizing atomic docking strategies at the side synthesis of polymeric nanoparticles loaded with CGA (CGA-NPs) by ionic gelation strategy. The results displayed a mean size of 101.9 ± 1.5 nm with a polydispersibility of 0.065 and a ZP of −17.4 mV (Figure 8). The release kinetics information from in vitro also appeared to support the release of CGA from the NPs after taking the first-order kinetics, recommending the suitable planning of nanoformulation [168].
Igor M. Pongrac and colleagues evaluated the effect of different surface coatings on the cellular uptake and toxicity of Ag NPs against murine neural stem cells (mNSCs). Some silver NPs were prepared with different surface coatings including cetyltrimethylammonium bromide (CTAB), poly-L-lysine (PLL), poly(vinylpyrrolidone) (PVP), sodium bis (2-Ethylhexyl)-sulfosuccinate (AOT), and bovine serum egg whites (BSA). The findings demonstrated that Ag NPs stabilized with various surface coatings had various toxicities and uptake effects in MNCs. For all of the Ag NP types tested, macropinocytosis was determined to be the most widely used tool in mNSCs. These findings add to the body of knowledge necessary for assessing the security of new nanostructures [169].
Liu et al. prepared PS NPs and attached an iron chelator to them, demonstrating their ability to protect neurons in vitro. The developed NPs have the potential to apply as a transfection vector in Parkinson’s treatment, as well as an elective infection to reduce the immune response in blood circulation [170].
Trapani et al. used chitosan nanoparticles for the encapsulation of dopamine acid (DA) as a safe vector to pass through the blood–brain barrier. Furthermore, the sum of DA and chitosan NPs within the striatum is greater than that of DA alone [171]. In a separate study, Javier Garcia et al. reported the production of a family of polymeric NPs and the DA loaded within NPs by its reversible coordination complexation. The results revealed a loading efficiency of up to 60%. DA nanoparticle coordination polymers (DA-NCPs) have worse toxicity, degradation kinetics, and absorption by BE (2)-M17 DA in vitro with glutamatergic cells and free DA. Direct particle mixing in the ventricle of rats in vivo appears to result in rapid transport within the brain of normal rats, increasing striatal DA levels. More importantly, after 4 days of nasal doses of DA-NCPs equivalent to 200 g of free medication each day, the duration and number of apomorphine-induced rotations were meaningfully fewer than in either vehicle or DA-treated rats used for comparability [172].
Linying Liu and colleagues demonstrated a straightforward technique using a switchable poly (carboxy betaine) gold nanoparticle that delivered genes and chemical medicines programmatically (gene–chem). The particle is replaceable between the delivery route and specific infected cells, allowing for enhanced synergistic neuronal recovery and also computed tomography (CT) imaging. The precise transformation delivery framework will serve as an effective gene–chem co-delivery stage to enable precise brain malady therapy [173].
The effects of polymer-coated silica NPs utilized in laser tissue soldering (LTS) on human brain endothelial cells (ECs) and BBB models were studied by Aniela Bittner et al. In the co-culture scenario with pericytes and ECs, only the cell type directly exposed to NPs demonstrated a time-dependent internalization. There was no evidence of NP transfer between the two cell types. The length and concentration of NP introduction had a moderate effect on cell reasonability. Following NP injection, protein expression of the nuclear factor k-light-chain-enhancer in various endothelial grip particles and activated B cells revealed no irritation or EC enactment. The established endothelial layer reduced the section of judgment tracer particles. The addition of NPs had no effect on junctional proteins, BBB arrangement, or decision making [174].
PLA-coated MSNs were tested for brain targeting with low-density lipoprotein receptor (LDLR) as a ligand. These targeted particles were capable of crossing the BBB and delivering resveratrol to the CNS. PLA was explored in this research as a stimuli-responsive watchman. It was demonstrated that PLA fundamentally impairs medication discharge in PBS, but in the presence of superoxide (ROS), sedate discharge increased due to PLA’s accelerated decomposition [175].
PEG-coated MSNs were used to deliver flavonoids (Figure 9) with significant antioxidant effects (quercetin, myricetin, and myricitrin) to increase their water solubility, release characteristics, and stability. Flavonoids with high drug loading (up to 27 weight percent) were found to be effective at inhibiting H2O2-induced changes in the elasticity and morphology of a model lipid membrane. The findings suggested that flavonoid-loaded MSNs had neuroprotective properties and that atomic force microscopy could be a useful technology for monitoring drug-induced effects at the membrane level, which could be expanded to the cellular level [176].
Cerium oxide nanoparticles appear to mimic oxidoreductase proteins by stimulating the decomposition of reactive oxygen species and natural substrates. Hydrogen peroxides and superoxide radicals, which are damaging particles produced in oxidative stress-related diseases, exhibit this similarity [177].
By affixing 7.8 nm cerium oxide centers to four PEG-grafted copolymers and two poly (sodium acrylate) with various terminal or tying down conclusion bunches, such as phosphonic acids, Victor Baldim et al. created six polymer-coated cerium oxide nanoparticles. The coated nanoparticles’ superoxide dismutase, peroxidase, catalase, and oxidase-like catalytic works were usefully considered. The polymer coatings do not disrupt oxidase-like, superoxide, dismutase-like, or catalase-like, catalytic activities of cerium oxide nanoparticles, but surprisingly boost peroxidase-like catalytic activities. It is also demonstrated that the particles coated with PEG-grafted copolymers work better as oxidoreductase-like proteins than the PAA-coated particles, a finding that supports the use of phosphonic acids as securing bunches at the molecular surface [177].
Xiaoming Chen and colleagues coated PLA microspheres with cationic polymers such as chitosan chloride (CSC), chitosan (CS), and polyethyleneimine (PEI) to form positively charged surfaces. By raising the surface charge of coated microspheres, they appeared to boost antigen adsorption capacity. HBsAg adsorbed on the surface of cationic microspheres improved antigen take-up and increased CD86, MHC I, and MHC II expression, as well as IL-1, IL-6, TNF-, and IL-12 release in macrophages. In antigen-attached cationic microsphere formations, antigens were more likely to relocate independently of lysosomes following phagocytosis. Furthermore, they found that microspheres coated with cationic polymers with somewhat high positive charges and greater antigen adsorption stimulated the Th1 response effectively. In summation, cationic polymer-coated PLA microspheres might serve as a vehicle for recombinant antigen delivery to initiate solid cell and humoral-resistant reactions [178].
Fornaguera C. and coworkers designed novel galantamine-loaded poly (lactic-co-glycolic acid) NPs for the first time by using the nano-emulsification method. They displayed that these NPs have the appropriate features to become advanced drug delivery systems for the therapy of neurodegenerative diseases. Loading proficiency was higher than 90 wt% with a supported medicate release profile. The enzymatic activity of the NPs was kept at 80% after its loading in nanoparticles [179].
Meenakshi Malhotra and colleagues produced nanoparticles for siRNA conveyance in ND using an underutilized peptide-tagged PEG chitosan polymer. They specifically advertised a simple chemo-specific conjugation of monomethoxy PEG at the C2 hydroxyl gather of chitosan polymer, with PEG conjugation to a cell-penetrating peptide. In an in vitro demonstration of an NDD Spinocerebellar ataxia (SCA1) over-expressing ataxin protein, the nanoparticles attempted to transport a useful siRNA against the Ataxin-1 quality. The results show that the SCA1 protein is successfully hidden after 48 h of transfection [159]. The structure and synthesis method of the coated NPs are shown in Table 2. Various polymer-coated nanoparticles for brain-targeted delivery are summarized in Table 3.

6. Challenges

Unlike significant achievements in the biomedical application of nanomedicine, there are some problems which have not yet been reported in clinical treatment, and these are limited in experimental data [189]. Importantly, a clear pathway leading to a cure for these diseases remains elusive, and the potential toxicity of NPs and their effect on the human brain are considered some of the main challenges leading to complicated molecular pathogenesis procedures, useless medical protocol, asymptomatic presentation, and the heterogeneous nature of the diseases [189,190]. Many factors including the size, shape, surface potential, surface functionalization, impurity attached during synthesis, and chemical composition have a direct bearing on the toxicity of nanomaterials [191]. In addition, some metal NPs block the protein transformation of heavy-chain ferritin and form complex structures that adversely affect the function of neuroprotection [192].
Accordingly, it seems that there are still unclear and complex challenges for the application of nanomedicine which are necessary to attend to in clinical translation.

7. Conclusions and Future Prospective

The cells of the BBB produce prostaglandins, nitric oxide, and cytokines that affect central nervous system (CNS) function, and dysfunction of the BBB causes some neurodegenerative disorders. It is essential to present a sufficient procedure in drug delivery and design suitable nanocarriers which are able to surround drugs to protect them, improve their circulation time, and provide a temporal and spatial controlled release in these areas through some strategies such as the usage of certain ligands on NP surface, exploiting their shape, and nanocarrier-based biocompatible polymers. To date, there has not been a reported case in clinical trials for neurodegenerative disorders, and it seems that it is the best occasion for NP developed in pre-clinical tests to pave the way for treating these diseases. Thus, a NP must be designed that behaves specifically and decreases side effects on other organs and cells. To sum up, the design of compatible, suitable polymer-coated nanoparticles and a combination of favorable therapeutic and imaging agents will be necessary to open the horizon in the treatment of more effective, non-evasive, and brain-directed treatments in neurodegenerative diseases which improve longevity and health.

Author Contributions

M.B.A.: Conceptualization, Methodology, Software, Writing—original draft, Writing—review and editing; A.T.J.: Investigation, Methodology, Visualization, Writing—review and editing; M.B.: Data curation, Investigation, Project administration, Supervision, Writing—original draft, Writing—review and editing; H.A.: Visualization, Writing—original draft, Writing—review and editing; S.M.S.: Software, Validation, Writing—original draft; S.S.: Resources, Software, Validation; P.Z.: Investigation, Methodology, Supervision, Writing—original draft; J.J.: Software, Validation, Writing—original draft, Writing—review and editing; A.J.I.: Investigation, Supervision, Writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sikdar, S.; Guha, S. Advancements of healthcare technologies: Paradigm towards smart healthcare systems. In Recent Trends in Image and Signal Processing in Computer Vision; Springer: Berlin/Heidelberg, Germany, 2020; pp. 113–132. [Google Scholar]
  2. Liu, Z.; Zhou, T.; Ziegler, A.C.; Dimitrion, P.; Zuo, L. Oxidative stress in neurodegenerative diseases: From molecular mechanisms to clinical applications. Oxid. Med. Cell. Longev. 2017, 2017, 2525967. [Google Scholar] [CrossRef]
  3. Wyss-Coray, T. Ageing, neurodegeneration and brain rejuvenation. Nature 2016, 539, 180–186. [Google Scholar] [CrossRef] [Green Version]
  4. Cicero, C.E.; Mostile, G.; Vasta, R.; Rapisarda, V.; Santo Signorelli, S.; Ferrante, M.; Zappia, M.; Nicoletti, A. Metals and neurodegenerative diseases. A systematic review. Environ. Res. 2017, 159, 82–94. [Google Scholar] [CrossRef]
  5. Sabir, F.; Ain, Q.U.; Rahdar, A.; Yang, Z.; Barani, M.; Bilal, M.; Bhalla, N. Correction to: Functionalized Nanoparticles in Drug Delivery: Strategies to Enhance Direct Nose-to-Brain Drug Delivery via Integrated Nerve Pathways. In Synthesis and Applications of Nanoparticles; Springer: Berlin/Heidelberg, Germany, 2022; p. C1. [Google Scholar]
  6. Cattaneo, A.; Cattane, N.; Galluzzi, S.; Provasi, S.; Lopizzo, N.; Festari, C.; Ferrari, C.; Guerra, U.P.; Paghera, B.; Muscio, C. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol. Aging 2017, 49, 60–68. [Google Scholar] [CrossRef] [Green Version]
  7. Md, S.; Bhattmisra, S.K.; Zeeshan, F.; Shahzad, N.; Mujtaba, M.A.; Meka, V.S.; Radhakrishnan, A.; Kesharwani, P.; Baboota, S.; Ali, J. Nano-carrier enabled drug delivery systems for nose to brain targeting for the treatment of neurodegenerative disorders. J. Drug Deliv. Sci. Technol. 2018, 43, 295–310. [Google Scholar] [CrossRef]
  8. Saraiva, C.; Praça, C.; Ferreira, R.; Santos, T.; Ferreira, L.; Bernardino, L. Nanoparticle-mediated brain drug delivery: Overcoming blood–brain barrier to treat neurodegenerative diseases. J. Control. Release 2016, 235, 34–47. [Google Scholar] [CrossRef] [Green Version]
  9. Salarpour, S.; Barani, M.; Pardakhty, A.; Khatami, M.; Chauhan, N.P.S. The application of exosomes and exosome-nanoparticle in treating brain disorders. J. Mol. Liq. 2022, 350, 118549. [Google Scholar] [CrossRef]
  10. Arkaban, H.; Shervedani, R.K.; Yaghoobi, F.; Kefayat, A.; Ghahremani, F. Imaging and therapeutic capabilities of the AuNPs@ MnCO3/Mn3O4, coated with PAA and integrated with folic acid, doxorubicin and propidium iodide for murine breast cancer. J. Drug Deliv. Sci. Technol. 2022, 67, 102818. [Google Scholar] [CrossRef]
  11. Liu, C.; Wang, Y.; Li, L.; He, D.; Chi, J.; Li, Q.; Wu, Y.; Zhao, Y.; Zhang, S.; Wang, L. Engineered extracellular vesicles and their mimetics for cancer immunotherapy. J. Control. Release 2022, 349, 679–698. [Google Scholar] [CrossRef]
  12. Roostaee, M.; Sheikhshoaie, I. Fabrication of a sensitive sensor for determination of xanthine in the presence of uric acid and ascorbic acid by modifying a carbon paste sensor with Fe3O4@ Au core–shell and an ionic liquid. J. Food Meas. Charact. 2022, 16, 731–739. [Google Scholar] [CrossRef]
  13. Sonbol, H.; AlYahya, S.; Ameen, F.; Alsamhary, K.; Alwakeel, S.; Al-Otaibi, S.; Korany, S. Bioinspired synthesize of CuO nanoparticles using Cylindrospermum stagnale for antibacterial, anticancer and larvicidal applications. Appl. Nanosci. 2021, in press. [Google Scholar] [CrossRef]
  14. Zeng, Q.; Bie, B.; Guo, Q.; Yuan, Y.; Han, Q.; Han, X.; Chen, M.; Zhang, X.; Yang, Y.; Liu, M. Hyperpolarized Xe NMR signal advancement by metal-organic framework entrapment in aqueous solution. Proc. Natl. Acad. Sci. USA 2020, 117, 17558–17563. [Google Scholar] [CrossRef]
  15. He, X.; Zhu, Y.; Yang, L.; Wang, Z.; Wang, Z.; Feng, J.; Wen, X.; Cheng, L.; Zhu, R. MgFe-LDH nanoparticles: A promising leukemia inhibitory factor replacement for self-renewal and pluripotency maintenance in cultured mouse embryonic stem cells. Adv. Sci. 2021, 8, 2003535. [Google Scholar] [CrossRef]
  16. Rabiee, N.; Ahmadi, S.; Afshari, R.; Khalaji, S.; Rabiee, M.; Bagherzadeh, M.; Fatahi, Y.; Dinarvand, R.; Tahriri, M.; Tayebi, L. Polymeric nanoparticles for nasal drug delivery to the brain: Relevance to Alzheimer’s disease. Adv. Ther. 2021, 4, 2000076. [Google Scholar] [CrossRef]
  17. Zhang, W.; Mehta, A.; Tong, Z.; Esser, L.; Voelcker, N.H. Development of polymeric nanoparticles for blood–brain barrier transfer—Strategies and challenges. Adv. Sci. 2021, 8, 2003937. [Google Scholar] [CrossRef]
  18. Meena, J.; Gupta, A.; Ahuja, R.; Singh, M.; Bhaskar, S.; Panda, A.K. Inorganic nanoparticles for natural product delivery: A review. Environ. Chem. Lett. 2020, 18, 2107–2118. [Google Scholar] [CrossRef]
  19. Akbarizadeh, M.R.; Sarani, M.; Darijani, S. Study of antibacterial performance of biosynthesized pure and Ag-doped ZnO nanoparticles. Rend. Lincei. Sci. Fis. E Nat. 2022, 33, 613–621. [Google Scholar] [CrossRef]
  20. Akbarizadeh, M.R.; Naderifar, M.; Mousazadeh, F.; Zafarnia, N.; Sarani, M. Cytotoxic activity and Magnetic Behavior of green synthesized iron oxide nanoparticles on brain glioblastoma cells. Nanomed. Res. J. 2022, 7, 99–106. [Google Scholar]
  21. Mortezagholi, B.; Movahed, E.; Fathi, A.; Soleimani, M.; Forutan Mirhosseini, A.; Zeini, N.; Khatami, M.; Naderifar, M.; Abedi Kiasari, B.; Zareanshahraki, M. Plant-mediated synthesis of silver-doped zinc oxide nanoparticles and evaluation of their antimicrobial activity against bacteria cause tooth decay. Microsc. Res. Tech. 2022, 85, 3553–3564. [Google Scholar] [CrossRef]
  22. Satarzadeh, N.; Shakibaie, M.; Adeli-Sardou, M.; Jabari-Morouei, F.; Forootanfar, H.; Sadeghi-Dousari, A. Facile microwave-assisted biosynthesis of arsenic nanoparticles and evaluation their antioxidant properties and cytotoxic effects: A preliminary in vitro study. J. Clust. Sci. 2022, in press. [Google Scholar] [CrossRef]
  23. Mohanta, Y.K.; Panda, S.K.; Syed, A.; Ameen, F.; Bastia, A.K.; Mohanta, T.K. Bio-inspired synthesis of silver nanoparticles from leaf extracts of Cleistanthus collinus (Roxb.): Its potential antibacterial and anticancer activities. IET Nanobiotechnol. 2018, 12, 343–348. [Google Scholar] [CrossRef]
  24. Sonbol, H.; Ameen, F.; AlYahya, S.; Almansob, A.; Alwakeel, S. Padina boryana mediated green synthesis of crystalline palladium nanoparticles as potential nanodrug against multidrug resistant bacteria and cancer cells. Sci. Rep. 2021, 11, 5444. [Google Scholar] [CrossRef]
  25. Kumar, B.; Pandey, M.; Pottoo, F.H.; Fayaz, F.; Sharma, A.; Sahoo, P. Liposomes: Novel drug delivery approach for targeting Parkinson’s disease. Curr. Pharm. Des. 2020, 26, 4721–4737. [Google Scholar] [CrossRef]
  26. Ozkizilcik, A.; Williams, R.; Tian, Z.R.; Muresanu, D.F.; Sharma, A.; Sharma, H.S. Synthesis of biocompatible titanate nanofibers for effective delivery of neuroprotective agents. In Neurotrophic Factors; Springer: Berlin/Heidelberg, Germany, 2018; pp. 433–442. [Google Scholar]
  27. Lai, W.-F.; Wong, W.-T. Property-tuneable microgels fabricated by using flow-focusing microfluidic geometry for bioactive agent delivery. Pharmaceutics 2021, 13, 787. [Google Scholar] [CrossRef]
  28. Lai, W.-F. Development of hydrogels with self-healing properties for delivery of bioactive agents. Mol. Pharm. 2021, 18, 1833–1841. [Google Scholar] [CrossRef]
  29. Obireddy, S.R.; Lai, W.-F. Multi-component hydrogel beads incorporated with reduced graphene oxide for ph-responsive and controlled co-delivery of multiple agents. Pharmaceutics 2021, 13, 313. [Google Scholar] [CrossRef]
  30. Barani, M.; Mirzaei, M.; Torkzadeh-Mahani, M.; Nematollahi, M.H. Lawsone-loaded Niosome and its antitumor activity in MCF-7 breast Cancer cell line: A Nano-herbal treatment for Cancer. DARU J. Pharm. Sci. 2018, 26, 11–17. [Google Scholar] [CrossRef]
  31. Hajizadeh, M.R.; Maleki, H.; Barani, M.; Fahmidehkar, M.A.; Mahmoodi, M.; Torkzadeh-Mahani, M. In vitro cytotoxicity assay of D-limonene niosomes: An efficient nano-carrier for enhancing solubility of plant-extracted agents. Res. Pharm. Sci. 2019, 14, 448. [Google Scholar]
  32. Barani, M.; Mirzaei, M.; Torkzadeh-Mahani, M.; Adeli-Sardou, M. Evaluation of carum-loaded niosomes on breast cancer cells: Physicochemical properties, in vitro cytotoxicity, flow cytometric, DNA fragmentation and cell migration assay. Sci. Rep. 2019, 9, 7139. [Google Scholar] [CrossRef] [Green Version]
  33. Yin, Y.; Wang, J.; Yang, M.; Du, R.; Pontrelli, G.; McGinty, S.; Wang, G.; Yin, T.; Wang, Y. Penetration of the blood–brain barrier and the anti-tumour effect of a novel PLGA-lysoGM1/DOX micelle drug delivery system. Nanoscale 2020, 12, 2946–2960. [Google Scholar] [CrossRef]
  34. Rahdar, A.; Taboada, P.; Hajinezhad, M.R.; Barani, M.; Beyzaei, H. Effect of tocopherol on the properties of Pluronic F127 microemulsions: Physico-chemical characterization and in vivo toxicity. J. Mol. Liq. 2019, 277, 624–630. [Google Scholar] [CrossRef]
  35. Muntoni, E.; Martina, K.; Marini, E.; Giorgis, M.; Lazzarato, L.; Salaroglio, I.C.; Riganti, C.; Lanotte, M.; Battaglia, L. Methotrexate-loaded solid lipid nanoparticles: Protein functionalization to improve brain biodistribution. Pharmaceutics 2019, 11, 65. [Google Scholar] [CrossRef]
  36. Kheirkhah, P.; Denyer, S.; Bhimani, A.D.; Arnone, G.D.; Esfahani, D.R.; Aguilar, T.; Zakrzewski, J.; Venugopal, I.; Habib, N.; Gallia, G.L. Magnetic drug targeting: A novel treatment for intramedullary spinal cord tumors. Sci. Rep. 2018, 8, 11417. [Google Scholar] [CrossRef] [Green Version]
  37. Liu, M.; Li, M.; Wang, G.; Liu, X.; Liu, D.; Peng, H.; Wang, Q. Heart-targeted nanoscale drug delivery systems. J. Biomed. Nanotechnol. 2014, 10, 2038–2062. [Google Scholar] [CrossRef]
  38. Peng, H.; Liu, X.; Wang, R.; Jia, F.; Dong, L.; Wang, Q. Emerging nanostructured materials for musculoskeletal tissue engineering. J. Mater. Chem. B 2014, 2, 6435–6461. [Google Scholar] [CrossRef]
  39. Wohlfart, S.; Gelperina, S.; Kreuter, J. Transport of drugs across the blood–brain barrier by nanoparticles. J. Control. Release 2012, 161, 264–273. [Google Scholar] [CrossRef]
  40. Poovaiah, N.; Davoudi, Z.; Peng, H.; Schlichtmann, B.; Mallapragada, S.; Narasimhan, B.; Wang, Q. Treatment of neurodegenerative disorders through the blood–brain barrier using nanocarriers. Nanoscale 2018, 10, 16962–16983. [Google Scholar] [CrossRef] [Green Version]
  41. Barani, M.; Mukhtar, M.; Rahdar, A.; Sargazi, G.; Thysiadou, A.; Kyzas, G.Z. Progress in the Application of Nanoparticles and Graphene as Drug Carriers and on the Diagnosis of Brain Infections. Molecules 2021, 26, 186. [Google Scholar] [CrossRef]
  42. Mukhtar, M.; Bilal, M.; Rahdar, A.; Barani, M.; Arshad, R.; Behl, T.; Brisc, C.; Banica, F.; Bungau, S. Nanomaterials for diagnosis and treatment of brain cancer: Recent updates. Chemosensors 2020, 8, 117. [Google Scholar] [CrossRef]
  43. Jena, L.; McErlean, E.; McCarthy, H. Delivery across the blood-brain barrier: Nanomedicine for glioblastoma multiforme. Drug Deliv. Transl. Res. 2020, 10, 304–318. [Google Scholar] [CrossRef] [Green Version]
  44. Moghimi, S.M.; Hunter, A.C.; Murray, J.C. Long-circulating and target-specific nanoparticles: Theory to practice. Pharmacol. Rev. 2001, 53, 283–318. [Google Scholar]
  45. Calzoni, E.; Cesaretti, A.; Polchi, A.; Di Michele, A.; Tancini, B.; Emiliani, C. Biocompatible polymer nanoparticles for drug delivery applications in cancer and neurodegenerative disorder therapies. J. Funct. Biomater. 2019, 10, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Letchford, K.; Burt, H. A review of the formation and classification of amphiphilic block copolymer nanoparticulate structures: Micelles, nanospheres, nanocapsules and polymersomes. Eur. J. Pharm. Biopharm. 2007, 65, 259–269. [Google Scholar] [CrossRef]
  47. Joye, I.J.; McClements, D.J. Biopolymer-based nanoparticles and microparticles: Fabrication, characterization, and application. Curr. Opin. Colloid Interface Sci. 2014, 19, 417–427. [Google Scholar] [CrossRef]
  48. Lai, W.-F. Non-conjugated polymers with intrinsic luminescence for drug delivery. J. Drug Deliv. Sci. Technol. 2020, 59, 101916. [Google Scholar] [CrossRef]
  49. Miller, K.D.; Fidler-Benaoudia, M.; Keegan, T.H.; Hipp, H.S.; Jemal, A.; Siegel, R.L. Cancer statistics for adolescents and young adults, 2020. CA Cancer J. Clin. 2020, 70, 443–459. [Google Scholar] [CrossRef]
  50. Tandel, G.S.; Biswas, M.; Kakde, O.G.; Tiwari, A.; Suri, H.S.; Turk, M.; Laird, J.R.; Asare, C.K.; Ankrah, A.A.; Khanna, N. A review on a deep learning perspective in brain cancer classification. Cancers 2019, 11, 111. [Google Scholar] [CrossRef] [Green Version]
  51. Ansari, M.A.; Chung, I.-M.; Rajakumar, G.; Alzohairy, M.A.; Alomary, M.N.; Thiruvengadam, M.; Pottoo, F.H.; Ahmad, N. Current nanoparticle approaches in nose to brain drug delivery and anticancer therapy-a review. Curr. Pharm. Des. 2020, 26, 1128–1137. [Google Scholar] [CrossRef]
  52. Shakeri, S.; Ashrafizadeh, M.; Zarrabi, A.; Roghanian, R.; Afshar, E.G.; Pardakhty, A.; Mohammadinejad, R.; Kumar, A.; Thakur, V.K. Multifunctional polymeric nanoplatforms for brain diseases diagnosis, therapy and theranostics. Biomedicines 2020, 8, 13. [Google Scholar] [CrossRef] [Green Version]
  53. Parodi, A.; Rudzińska, M.; Deviatkin, A.A.; Soond, S.M.; Baldin, A.V.; Zamyatnin, A.A. Established and emerging strategies for drug delivery across the blood-brain barrier in brain cancer. Pharmaceutics 2019, 11, 245. [Google Scholar] [CrossRef] [Green Version]
  54. Wang, D.; Wang, C.; Wang, L.; Chen, Y. A comprehensive review in improving delivery of small-molecule chemotherapeutic agents overcoming the blood-brain/brain tumor barriers for glioblastoma treatment. Drug Deliv. 2019, 26, 551–565. [Google Scholar] [CrossRef]
  55. Gregory, J.V.; Kadiyala, P.; Doherty, R.; Cadena, M.; Habeel, S.; Ruoslahti, E.; Lowenstein, P.R.; Castro, M.G.; Lahann, J. Systemic brain tumor delivery of synthetic protein nanoparticles for glioblastoma therapy. Nat. Commun. 2020, 11, 5687. [Google Scholar] [CrossRef]
  56. Fayazi, N.; Sheykhhasan, M.; Soleimani Asl, S.; Najafi, R. Stem cell-derived exosomes: A new strategy of neurodegenerative disease treatment. Mol. Neurobiol. 2021, 58, 3494–3514. [Google Scholar] [CrossRef]
  57. Peplow, P.V.; Martinez, B.; Gennarelli, T.A. Prevalence, Needs, Strategies, and Risk Factors for Neurodegenerative Diseases. In Neurodegenerative Diseases Biomarkers; Springer: Berlin/Heidelberg, Germany, 2022; pp. 3–8. [Google Scholar]
  58. Ross, C.; Taylor, M.; Fullwood, N.; Allsop, D. Liposome delivery systems for the treatment of Alzheimer’s disease. Int. J. Nanomed. 2018, 13, 8507. [Google Scholar] [CrossRef] [Green Version]
  59. Ouyang, Q.; Meng, Y.; Zhou, W.; Tong, J.; Cheng, Z.; Zhu, Q. New advances in brain-targeting nano-drug delivery systems for Alzheimer’s disease. J. Drug Target. 2022, 30, 61–81. [Google Scholar] [CrossRef]
  60. Karthivashan, G.; Ganesan, P.; Park, S.-Y.; Kim, J.-S.; Choi, D.-K. Therapeutic strategies and nano-drug delivery applications in management of ageing Alzheimer’s disease. Drug Deliv. 2018, 25, 307–320. [Google Scholar] [CrossRef] [Green Version]
  61. Kaushik, A.C.; Bharadwaj, S.; Kumar, S.; Wei, D.-Q. Nano-particle mediated inhibition of Parkinson’s disease using computational biology approach. Sci. Rep. 2018, 8, 9169. [Google Scholar] [CrossRef] [Green Version]
  62. Jankovic, J.; Tan, E.K. Parkinson’s disease: Etiopathogenesis and treatment. J. Neurol. Neurosurg. Psychiatry 2020, 91, 795–808. [Google Scholar] [CrossRef]
  63. Chen, Y.-X.; Wei, C.-X.; Lyu, Y.-Q.; Chen, H.-Z.; Jiang, G.; Gao, X.-L. Biomimetic drug-delivery systems for the management of brain diseases. Biomater. Sci. 2020, 8, 1073–1088. [Google Scholar] [CrossRef]
  64. Upadhyay, R.K. Drug delivery systems, CNS protection, and the blood brain barrier. BioMed Res. Int. 2014, 2014, 869269. [Google Scholar] [CrossRef] [Green Version]
  65. Li, J.; Zhao, J.; Tan, T.; Liu, M.; Zeng, Z.; Zeng, Y.; Zhang, L.; Fu, C.; Chen, D.; Xie, T. Nanoparticle drug delivery system for glioma and its efficacy improvement strategies: A comprehensive review. Int. J. Nanomed. 2020, 15, 2563. [Google Scholar] [CrossRef] [Green Version]
  66. Ghosh, P.; Bera, A.; De, P. Current status, challenges and future directions in the treatment of neurodegenerative diseases by polymeric materials. J. Indian Chem. Soc. 2021, 98, 100011. [Google Scholar] [CrossRef]
  67. Tao, X.; Li, Y.; Hu, Q.; Zhu, L.; Huang, Z.; Yi, J.; Yang, X.; Hu, J.; Feng, X. Preparation and drug release study of novel nanopharmaceuticals with polysorbate 80 surface adsorption. J. Nanomater. 2018, 2018, 4718045. [Google Scholar] [CrossRef]
  68. Prieto, C.; Calvo, L. Performance of the biocompatible surfactant Tween 80, for the formation of microemulsions suitable for new pharmaceutical processing. J. Appl. Chem. 2013, 2013, 930356. [Google Scholar] [CrossRef] [Green Version]
  69. Sahu, A.K.; Mishra, J.; Mishra, A.K. Introducing Tween-curcumin niosomes: Preparation, characterization and microenvironment study. Soft Matter 2020, 16, 1779–1791. [Google Scholar] [CrossRef]
  70. Deng, L.-L.; Taxipalati, M.; Que, F.; Zhang, H. Physical characterization and antioxidant activity of thymol solubilized Tween 80 micelles. Sci. Rep. 2016, 6, 38160. [Google Scholar] [CrossRef]
  71. Hekmat, A.; Attar, H.; Seyf Kordi, A.A.; Iman, M.; Jaafari, M.R. New oral formulation and in vitro evaluation of docetaxel-loaded nanomicelles. Molecules 2016, 21, 1265. [Google Scholar] [CrossRef] [Green Version]
  72. Garidel, P.; Hoffmann, C.; Blume, A. A thermodynamic analysis of the binding interaction between polysorbate 20 and 80 with human serum albumins and immunoglobulins: A contribution to understand colloidal protein stabilisation. Biophys. Chem. 2009, 143, 70–78. [Google Scholar] [CrossRef] [Green Version]
  73. Jiao, J. Polyoxyethylated nonionic surfactants and their applications in topical ocular drug delivery. Adv. Drug Deliv. Rev. 2008, 60, 1663–1673. [Google Scholar] [CrossRef]
  74. El-Setouhy, D.A.; Basalious, E.B.; Abdelmalak, N.S. Bioenhanced sublingual tablet of drug with limited permeability using novel surfactant binder and microencapsulated polysorbate: In vitro/in vivo evaluation. Eur. J. Pharm. Biopharm. 2015, 94, 386–392. [Google Scholar] [CrossRef]
  75. Su, R.; Yang, L.; Wang, Y.; Yu, S.; Guo, Y.; Deng, J.; Zhao, Q.; Jin, X. Formulation, development, and optimization of a novel octyldodecanol-based nanoemulsion for transdermal delivery of ceramide IIIB. Int. J. Nanomed. 2017, 12, 5203. [Google Scholar] [CrossRef] [Green Version]
  76. Ravichandran, V.; Lee, M.; Nguyen Cao, T.G.; Shim, M.S. Polysorbate-based drug formulations for brain-targeted drug delivery and anticancer therapy. Appl. Sci. 2021, 11, 9336. [Google Scholar] [CrossRef]
  77. Torosantucci, R.; Furtmann, B.; Elshorst, B.; Pfeiffer-Marek, S.; Hartleb, T.; Andres, N.; Bussemer, T. Protein-excipient interactions evaluated via nuclear magnetic resonance studies in polysorbate-based multidose protein formulations: Influence on antimicrobial efficacy and potential study approach. J. Pharm. Sci. 2018, 107, 2531–2537. [Google Scholar] [CrossRef]
  78. Norouzi, M.; Yathindranath, V.; Thliveris, J.A.; Miller, D.W. Salinomycin-loaded iron oxide nanoparticles for glioblastoma therapy. Nanomaterials 2020, 10, 477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Kreuter, J. Nanoparticulate systems for brain delivery of drugs. Adv. Drug Deliv. Rev. 2001, 47, 65–81. [Google Scholar] [CrossRef]
  80. Tröster, S.; Müller, U.; Kreuter, J. Modification of the body distribution of poly (methyl methacrylate) nanoparticles in rats by coating with surfactants. Int. J. Pharm. 1990, 61, 85–100. [Google Scholar] [CrossRef]
  81. Ray, S.; Sinha, P.; Laha, B.; Maiti, S.; Bhattacharyya, U.K.; Nayak, A.K. Polysorbate 80 coated crosslinked chitosan nanoparticles of ropinirole hydrochloride for brain targeting. J. Drug Deliv. Sci. Technol. 2018, 48, 21–29. [Google Scholar] [CrossRef]
  82. Thomas, A.; Müller, S.S.; Frey, H. Beyond poly (ethylene glycol): Linear polyglycerol as a multifunctional polyether for biomedical and pharmaceutical applications. Biomacromolecules 2014, 15, 1935–1954. [Google Scholar] [CrossRef] [PubMed]
  83. Barani, M.; Mirzaei, M.; Torkzadeh-Mahani, M.; Lohrasbi-Nejad, A.; Nematollahi, M.H. A new formulation of hydrophobin-coated niosome as a drug carrier to cancer cells. Mater. Sci. Eng. C 2020, 113, 110975. [Google Scholar] [CrossRef] [PubMed]
  84. Davarpanah, F.; Khalili Yazdi, A.; Barani, M.; Mirzaei, M.; Torkzadeh-Mahani, M. Magnetic delivery of antitumor carboplatin by using PEGylated-Niosomes. DARU J. Pharm. Sci. 2018, 26, 57–64. [Google Scholar] [CrossRef]
  85. D’souza, A.A.; Shegokar, R. Polyethylene glycol (PEG): A versatile polymer for pharmaceutical applications. Expert Opin. Drug Deliv. 2016, 13, 1257–1275. [Google Scholar] [CrossRef] [PubMed]
  86. Jokerst, J.V.; Lobovkina, T.; Zare, R.N.; Gambhir, S.S. Nanoparticle PEGylation for imaging and therapy. Nanomedicine 2011, 6, 715–728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Schöttler, S.; Becker, G.; Winzen, S.; Steinbach, T.; Mohr, K.; Landfester, K.; Mailänder, V.; Wurm, F.R. Protein adsorption is required for stealth effect of poly (ethylene glycol)-and poly (phosphoester)-coated nanocarriers. Nat. Nanotechnol. 2016, 11, 372–377. [Google Scholar] [CrossRef] [PubMed]
  88. Knop, K.; Hoogenboom, R.; Fischer, D.; Schubert, U.S. Poly (ethylene glycol) in drug delivery: Pros and cons as well as potential alternatives. Angew. Chem. Int. Ed. 2010, 49, 6288–6308. [Google Scholar] [CrossRef] [PubMed]
  89. Howard, M.D.; Jay, M.; Dziubla, T.D.; Lu, X. PEGylation of nanocarrier drug delivery systems: State of the art. J. Biomed. Nanotechnol. 2008, 4, 133–148. [Google Scholar] [CrossRef]
  90. Nguyen, T.T.; Nguyen, T.T.D.; Nguyen, T.K.O.; Vo, T.K. Advances in developing therapeutic strategies for Alzheimer’s disease. Biomed. Pharmacother. 2021, 139, 111623. [Google Scholar] [CrossRef]
  91. Kean, T.; Thanou, M. Biodegradation, biodistribution and toxicity of chitosan. Adv. Drug Deliv. Rev. 2010, 62, 3–11. [Google Scholar] [CrossRef]
  92. Younes, I.; Hajji, S.; Frachet, V.; Rinaudo, M.; Jellouli, K.; Nasri, M. Chitin extraction from shrimp shell using enzymatic treatment. Antitumor, antioxidant and antimicrobial activities of chitosan. Int. J. Biol. Macromol. 2014, 69, 489–498. [Google Scholar] [CrossRef]
  93. Peluso, G.; Petillo, O.; Ranieri, M.; Santin, M.; Ambrosic, L.; Calabró, D.; Avallone, B.; Balsamo, G. Chitosan-mediated stimulation of macrophage function. Biomaterials 1994, 15, 1215–1220. [Google Scholar] [CrossRef]
  94. Majedi, F.S.; Hasani-Sadrabadi, M.M.; VanDersarl, J.J.; Mokarram, N.; Hojjati-Emami, S.; Dashtimoghadam, E.; Bonakdar, S.; Shokrgozar, M.A.; Bertsch, A.; Renaud, P. On-chip fabrication of paclitaxel-loaded chitosan nanoparticles for cancer therapeutics. Adv. Funct. Mater. 2014, 24, 432–441. [Google Scholar] [CrossRef]
  95. Mikušová, V.; Mikuš, P. Advances in chitosan-based nanoparticles for drug delivery. Int. J. Mol. Sci. 2021, 22, 9652–9673. [Google Scholar]
  96. Karlsson, J.; Vaughan, H.J.; Green, J.J. Biodegradable polymeric nanoparticles for therapeutic cancer treatments. Annu. Rev. Chem. Biomol. Eng. 2018, 9, 105–127. [Google Scholar] [CrossRef]
  97. Monsalve, Y.; Tosi, G.; Ruozi, B.; Belletti, D.; Vilella, A.; Zoli, M.; Vandelli, M.A.; Forni, F.; Lopez, B.L.; Sierra, L. PEG-g-chitosan nanoparticles functionalized with the monoclonal antibody OX26 for brain drug targeting. Nanomedicine 2015, 10, 1735–1750. [Google Scholar] [CrossRef] [PubMed]
  98. Yu, S.; Xu, X.; Feng, J.; Liu, M.; Hu, K. Chitosan and chitosan coating nanoparticles for the treatment of brain disease. Int. J. Pharm. 2019, 560, 282–293. [Google Scholar] [CrossRef] [PubMed]
  99. Pacheco, C.; Sousa, F.; Sarmento, B. Chitosan-based nanomedicine for brain delivery: Where are we heading? React. Funct. Polym. 2020, 146, 104430. [Google Scholar] [CrossRef]
  100. Wilson, B.; Alobaid, B.N.M.; Geetha, K.M.; Jenita, J.L. Chitosan nanoparticles to enhance nasal absorption and brain targeting of sitagliptin to treat Alzheimer’s disease. J. Drug Deliv. Sci. Technol. 2021, 61, 102176. [Google Scholar] [CrossRef]
  101. Manek, E.; Darvas, F.; Petroianu, G.A. Use of biodegradable, chitosan-based nanoparticles in the treatment of Alzheimer’s disease. Molecules 2020, 25, 4866. [Google Scholar] [CrossRef]
  102. Sinha, V.; Bansal, K.; Kaushik, R.; Kumria, R.; Trehan, A. Poly-ϵ-caprolactone microspheres and nanospheres: An overview. Int. J. Pharm. 2004, 278, 1–23. [Google Scholar] [CrossRef]
  103. Woodruff, M.; Hutmacher, D. The return of a forgotten polymer—Polycaprolactone in the 21st century. Prog. Polym. Sci. 2010, 35, 1217–1256. [Google Scholar] [CrossRef] [Green Version]
  104. Woodward, S.C.; Brewer, P.; Moatamed, F.; Schindler, A.; Pitt, C. The intracellular degradation of poly (ε-caprolactone). J. Biomed. Mater. Res. 1985, 19, 437–444. [Google Scholar] [CrossRef] [PubMed]
  105. Kanehisa, M.; Goto, S.; Kawashima, S.; Nakaya, A. The KEGG databases at GenomeNet. Nucleic Acids Res. 2002, 30, 42–46. [Google Scholar] [CrossRef] [Green Version]
  106. Wishart, D.S.; Feunang, Y.D.; Marcu, A.; Guo, A.C.; Liang, K.; Vázquez-Fresno, R.; Sajed, T.; Johnson, D.; Li, C.; Karu, N. HMDB 4.0: The human metabolome database for 2018. Nucleic Acids Res. 2018, 46, D608–D617. [Google Scholar] [CrossRef] [PubMed]
  107. Grossen, P.; Witzigmann, D.; Sieber, S.; Huwyler, J. PEG-PCL-based nanomedicines: A biodegradable drug delivery system and its application. J. Control. Release 2017, 260, 46–60. [Google Scholar] [CrossRef]
  108. Varan, C.; Bilensoy, E. Cationic PEGylated polycaprolactone nanoparticles carrying post-operation docetaxel for glioma treatment. Beilstein J. Nanotechnol. 2017, 8, 1446–1456. [Google Scholar] [CrossRef]
  109. Xin, H.; Jiang, X.; Gu, J.; Sha, X.; Chen, L.; Law, K.; Chen, Y.; Wang, X.; Jiang, Y.; Fang, X. Angiopep-conjugated poly (ethylene glycol)-co-poly (ε-caprolactone) nanoparticles as dual-targeting drug delivery system for brain glioma. Biomaterials 2011, 32, 4293–4305. [Google Scholar] [CrossRef] [PubMed]
  110. Ulery, B.D.; Nair, L.S.; Laurencin, C.T. Biomedical applications of biodegradable polymers. J. Polym. Sci. Part B Polym. Phys. 2011, 49, 832–864. [Google Scholar] [CrossRef] [Green Version]
  111. Huang, M.-H.; Li, S.; Vert, M. Synthesis and degradation of PLA–PCL–PLA triblock copolymer prepared by successive polymerization of ε-caprolactone and dl-lactide. Polymer 2004, 45, 8675–8681. [Google Scholar] [CrossRef]
  112. Mohamadpour, H.; Azadi, A.; Rostamizadeh, K.; Andalib, S.; Saghatchi Zanjani, M.R.; Hamidi, M. Preparation, Optimization, and Evaluation of Methoxy Poly (ethylene glycol)-co-poly (ε-caprolactone) Nanoparticles Loaded by Rivastigmine for Brain Delivery. ACS Chem. Neurosci. 2020, 11, 783–795. [Google Scholar] [CrossRef] [PubMed]
  113. Pohlmann, A.R.; Fonseca, F.N.; Paese, K.; Detoni, C.B.; Coradini, K.; Beck, R.C.; Guterres, S.S. Poly (ϵ-caprolactone) microcapsules and nanocapsules in drug delivery. Expert Opin. Drug Deliv. 2013, 10, 623–638. [Google Scholar] [CrossRef]
  114. Pavlinec, J.; Novák, I.; Rychlý, J.; Kleinová, A.; Nógellová, Z.; Preťo, J.; Vanko, V.; Žigo, O.; Chodák, I. Hot melt adhesives prepared by grafting of acrylic and crotonic acids onto metallocene ethylene–octene copolymers. J. Plast. Film Sheet. 2019, 35, 239–259. [Google Scholar] [CrossRef]
  115. Dashtizadeh, A.; Abdouss, M.; Khorassani, M.; Mahdavi, H. Preparation of silica-filled water-based acrylic nanocomposite paints with improved scratch resistance. J. Plast. Film Sheet. 2012, 28, 120–135. [Google Scholar] [CrossRef]
  116. Foroushani, M.S.; Zahmatkeshan, A.; Arkaban, H.; Shervedani, R.K.; Kefayat, A. A drug delivery system based on nanocomposites constructed from metal-organic frameworks and Mn3O4 nanoparticles: Preparation and physicochemical characterization for BT-474 and MCF-7 cancer cells. Colloids Surf. B Biointerfaces 2021, 202, 111712. [Google Scholar] [CrossRef] [PubMed]
  117. Mori, H.; Müller, A.H.; Klee, J.E. Intelligent colloidal hybrids via reversible pH-induced complexation of polyelectrolyte and silica nanoparticles. J. Am. Chem. Soc. 2003, 125, 3712–3713. [Google Scholar] [CrossRef] [PubMed]
  118. Shimizu, T.; Masuda, M.; Minamikawa, H. Supramolecular nanotube architectures based on amphiphilic molecules. Chem. Rev. 2005, 105, 1401–1444. [Google Scholar] [CrossRef]
  119. Xu, M.; Zhu, J.; Wang, F.; Xiong, Y.; Wu, Y.; Wang, Q.; Weng, J.; Zhang, Z.; Chen, W.; Liu, S. Improved in vitro and in vivo biocompatibility of graphene oxide through surface modification: Poly (acrylic acid)-functionalization is superior to PEGylation. ACS Nano 2016, 10, 3267–3281. [Google Scholar] [CrossRef] [PubMed]
  120. Kausar, A. Poly (acrylic acid) nanocomposites: Design of advanced materials. J. Plast. Film Sheet. 2021, 37, 409–428. [Google Scholar] [CrossRef]
  121. Jing, X.; Feng, P.; Chen, Z.; Xie, Z.; Li, H.; Peng, X.-F.; Mi, H.-Y.; Liu, Y. Highly stretchable, self-healable, freezing-tolerant, and transparent polyacrylic acid/nanochitin composite hydrogel for self-powered multifunctional sensors. ACS Sustain. Chem. Eng. 2021, 9, 9209–9220. [Google Scholar] [CrossRef]
  122. Ebrahimi, A.K.; Sheikhshoaie, I.; Salimi, S.; Arkaban, H. In-situ facile synthesis of superparamagnetic porous core-shell structure for dye adsorption. J. Mol. Struct. 2021, 1228, 129797. [Google Scholar] [CrossRef]
  123. Jiang, L.; Gao, L.; Wang, X.; Tang, L.; Ma, J. The application of mucoadhesive polymers in nasal drug delivery. Drug Dev. Ind. Pharm. 2010, 36, 323–336. [Google Scholar] [CrossRef]
  124. Borkar, N.; Mu, H.; Holm, R. Challenges and trends in apomorphine drug delivery systems for the treatment of Parkinson’s disease. Asian J. Pharm. Sci. 2018, 13, 507–517. [Google Scholar] [CrossRef]
  125. Durazo, S.A.; Kompella, U.B. Functionalized nanosystems for targeted mitochondrial delivery. Mitochondrion 2012, 12, 190–201. [Google Scholar] [CrossRef] [Green Version]
  126. Arkaban, H.; Barani, M.; Akbarizadeh, M.R.; Pal Singh Chauhan, N.; Jadoun, S.; Dehghani Soltani, M.; Zarrintaj, P. Polyacrylic acid nanoplatforms: Antimicrobial, tissue engineering, and cancer theranostic applications. Polymers 2022, 14, 1259. [Google Scholar] [CrossRef]
  127. Hines, D.J.; Kaplan, D.L. Poly (lactic-co-glycolic) acid-controlled-release systems: Experimental and modeling insights. Crit. Rev. Ther. Drug Carr. Syst. 2013, 30, 257–276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Jose, S.; Sowmya, S.; Cinu, T.; Aleykutty, N.; Thomas, S.; Souto, E. Surface modified PLGA nanoparticles for brain targeting of Bacoside-A. Eur. J. Pharm. Sci. 2014, 63, 29–35. [Google Scholar] [CrossRef] [PubMed]
  129. Arkaban, H.; Mirzaei, M.; Behzadi, M. Magnetic solid-phase extraction of lawsone using polyphenol-coated magnetic nanoparticles: Synthesis, characterization and examination. Chromatographia 2021, 84, 455–462. [Google Scholar] [CrossRef]
  130. Makadia, H.K.; Siegel, S.J. Poly lactic-co-glycolic acid (PLGA) as biodegradable controlled drug delivery carrier. Polymers 2011, 3, 1377–1397. [Google Scholar] [CrossRef] [PubMed]
  131. Gentile, P.; Chiono, V.; Carmagnola, I.; Hatton, P.V. An overview of poly (lactic-co-glycolic) acid (PLGA)-based biomaterials for bone tissue engineering. Int. J. Mol. Sci. 2014, 15, 3640–3659. [Google Scholar] [CrossRef]
  132. Stayshich, R.M.; Weiss, R.M.; Li, J.; Meyer, T.Y. Periodic incorporation of pendant hydroxyl groups in repeating sequence PLGA copolymers. Macromol. Rapid Commun. 2011, 32, 220–225. [Google Scholar] [CrossRef]
  133. Choi, S.H.; Park, T.G. Synthesis and characterization of elastic PLGA/PCL/PLGA tri-block copolymers. J. Biomater. Sci. Polym. Ed. 2002, 13, 1163–1173. [Google Scholar] [CrossRef] [Green Version]
  134. Rezvantalab, S.; Drude, N.I.; Moraveji, M.K.; Güvener, N.; Koons, E.K.; Shi, Y.; Lammers, T.; Kiessling, F. PLGA-based nanoparticles in cancer treatment. Front. Pharmacol. 2018, 9, 1260. [Google Scholar] [CrossRef] [Green Version]
  135. Chu, K.S.; Hasan, W.; Rawal, S.; Walsh, M.D.; Enlow, E.M.; Luft, J.C.; Bridges, A.S.; Kuijer, J.L.; Napier, M.E.; Zamboni, W.C. Plasma, tumor and tissue pharmacokinetics of Docetaxel delivered via nanoparticles of different sizes and shapes in mice bearing SKOV-3 human ovarian carcinoma xenograft. Nanomed. Nanotechnol. Biol. Med. 2013, 9, 686–693. [Google Scholar] [CrossRef] [Green Version]
  136. Zhang, K.; Tang, X.; Zhang, J.; Lu, W.; Lin, X.; Zhang, Y.; Tian, B.; Yang, H.; He, H. PEG-PLGA copolymers: Their structure and structure-influenced drug delivery applications. J. Control. Release 2014, 183, 77–86. [Google Scholar] [CrossRef]
  137. Hoyos-Ceballos, G.P.; Ruozi, B.; Ottonelli, I.; Da Ros, F.; Vandelli, M.A.; Forni, F.; Daini, E.; Vilella, A.; Zoli, M.; Tosi, G. PLGA-PEG-ANG-2 Nanoparticles for Blood–Brain Barrier Crossing: Proof-of-Concept Study. Pharmaceutics 2020, 12, 72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Gambaryan, P.; Kondrasheva, I.; Severin, E.; Guseva, A.; Kamensky, A. Increasing the Efficiency of Parkinson’s Disease Treatment Using a poly (lactic-co-glycolic acid) (PLGA) Based L-DOPA Delivery System. Exp. Neurobiol. 2014, 23, 246. [Google Scholar] [CrossRef] [PubMed]
  139. Huang, W.; Zhang, C. Tuning the size of poly (lactic-co-glycolic acid) (PLGA) nanoparticles fabricated by nanoprecipitation. Biotechnol. J. 2018, 13, 1700203. [Google Scholar] [CrossRef] [PubMed]
  140. Zeb, A.; Gul, M.; Nguyen, T.-T.-L.; Maeng, H.-J. Controlled release and targeted drug delivery with poly (lactic-co-glycolic acid) nanoparticles: Reviewing two decades of research. J. Pharm. Investig. 2022, 52, 683–724. [Google Scholar] [CrossRef]
  141. Popova, N.V.; Jücker, M. The role of mTOR signaling as a therapeutic target in cancer. Int. J. Mol. Sci. 2021, 22, 1743. [Google Scholar] [CrossRef]
  142. Su, M.; Soomro, S.H.; Jie, J.; Fu, H. Effects of the extracellular matrix on myelin development and regeneration in the central nervous system. Tissue Cell 2021, 69, 101444. [Google Scholar] [CrossRef]
  143. Bădilă, A.E.; Rădulescu, D.M.; Niculescu, A.-G.; Grumezescu, A.M.; Rădulescu, M.; Rădulescu, A.R. Recent advances in the treatment of bone metastases and primary bone tumors: An up-to-date review. Cancers 2021, 13, 4229. [Google Scholar] [CrossRef]
  144. Voci, S.; Fresta, M.; Cosco, D. Gliadins as versatile biomaterials for drug delivery applications. J. Control. Release 2021, 329, 385–400. [Google Scholar] [CrossRef] [PubMed]
  145. Browne, S.; Hossainy, S.; Healy, K. Hyaluronic Acid macromer molecular weight dictates the biophysical properties and in vitro cellular response to semisynthetic hydrogels. ACS Biomater. Sci. Eng. 2019, 6, 1135–1143. [Google Scholar] [CrossRef]
  146. Zakusilo, F.T.; O’Banion, M.K.; Gelbard, H.A.; Seluanov, A.; Gorbunova, V. Matters of size: Roles of hyaluronan in CNS aging and disease. Ageing Res. Rev. 2021, 72, 101485. [Google Scholar] [CrossRef] [PubMed]
  147. Grieco, M.; Ursini, O.; Palamà, I.E.; Gigli, G.; Moroni, L.; Cortese, B. HYDRHA: Hydrogels of hyaluronic acid. New biomedical approaches in cancer, neurodegenerative diseases, and tissue engineering. Mater. Today Bio 2022, 17, 100453. [Google Scholar] [CrossRef] [PubMed]
  148. Coisne, C.; Tilloy, S.; Monflier, E.; Wils, D.; Fenart, L.; Gosselet, F. Cyclodextrins as emerging therapeutic tools in the treatment of cholesterol-associated vascular and neurodegenerative diseases. Molecules 2016, 21, 1748. [Google Scholar] [CrossRef]
  149. Loftus, S.K.; Morris, J.A.; Carstea, E.D.; Gu, J.Z.; Cummings, C.; Brown, A.; Ellison, J.; Ohno, K.; Rosenfeld, M.A.; Tagle, D.A. Murine model of Niemann-Pick C disease: Mutation in a cholesterol homeostasis gene. Science 1997, 277, 232–235. [Google Scholar] [CrossRef] [PubMed]
  150. Carstea, E.D.; Morris, J.A.; Coleman, K.G.; Loftus, S.K.; Zhang, D.; Cummings, C.; Gu, J.; Rosenfeld, M.A.; Pavan, W.J.; Krizman, D.B. Niemann-Pick C1 disease gene: Homology to mediators of cholesterol homeostasis. Science 1997, 277, 228–231. [Google Scholar] [CrossRef] [Green Version]
  151. Fanali, G.; Di Masi, A.; Trezza, V.; Marino, M.; Fasano, M.; Ascenzi, P. Human serum albumin: From bench to bedside. Mol. Asp. Med. 2012, 33, 209–290. [Google Scholar] [CrossRef]
  152. Sebak, S.; Mirzaei, M.; Malhotra, M.; Kulamarva, A.; Prakash, S. Human serum albumin nanoparticles as an efficient noscapine drug delivery system for potential use in breast cancer: Preparation and in vitro analysis. Int. J. Nanomed. 2010, 5, 525. [Google Scholar]
  153. Larsen, M.T.; Kuhlmann, M.; Hvam, M.L.; Howard, K.A. Albumin-based drug delivery: Harnessing nature to cure disease. Mol. Cell. Ther. 2016, 4, 3. [Google Scholar] [CrossRef] [Green Version]
  154. Shabani-Nooshabadi, M.; Roostaee, M. Coupling of NiO nanoparticles and room temperature ionic liquid for fabrication of highly sensitive voltammetric sensor in tryptophan analysis. Anal. Bioanal. Electrochem. 2016, 8, 578–588. [Google Scholar]
  155. Vidhya, M.S.; Ameen, F.; Dawoud, T.; Yuvakkumar, R.; Ravi, G.; Kumar, P.; Velauthapillai, D. Anti-cancer applications of Zr, Co, Ni-doped ZnO thin nanoplates. Mater. Lett. 2021, 283, 128760. [Google Scholar] [CrossRef]
  156. Swathi, S.; Ameen, F.; Ravi, G.; Yuvakkumar, R.; Hong, S.; Velauthapillai, D.; AlKahtani, M.D.; Thambidurai, M.; Dang, C. Cancer targeting potential of bioinspired chain like magnetite (Fe3O4) nanostructures. Curr. Appl. Phys. 2020, 20, 982–987. [Google Scholar] [CrossRef]
  157. Ameen, F.; Al-Maary, K.S.; Almansob, A.; AlNadhari, S. Antioxidant, antibacterial and anticancer efficacy of Alternaria chlamydospora-mediated gold nanoparticles. Appl. Nanosci. 2022, in press. [Google Scholar] [CrossRef]
  158. Singh, P.; Pandit, S.; Mokkapati, V.; Garg, A.; Ravikumar, V.; Mijakovic, I. Gold nanoparticles in diagnostics and therapeutics for human cancer. Int. J. Mol. Sci. 2018, 19, 1979. [Google Scholar] [CrossRef] [PubMed]
  159. Malhotra, M.; Tomaro-Duchesneau, C.; Prakash, S. Synthesis of TAT peptide-tagged PEGylated chitosan nanoparticles for siRNA delivery targeting neurodegenerative diseases. Biomaterials 2013, 34, 1270–1280. [Google Scholar] [CrossRef] [PubMed]
  160. Tao, J.; Fei, W.; Tang, H.; Li, C.; Mu, C.; Zheng, H.; Li, F.; Zhu, Z. Angiopep-2-conjugated “core–shell” hybrid nanovehicles for targeted and pH-triggered delivery of arsenic trioxide into glioma. Mol. Pharm. 2019, 16, 786–797. [Google Scholar] [CrossRef] [PubMed]
  161. Calvo, P.; Gouritin, B.; Villarroya, H.; Eclancher, F.; Giannavola, C.; Klein, C.; Andreux, J.P.; Couvreur, P. Quantification and localization of PEGylated polycyanoacrylate nanoparticles in brain and spinal cord during experimental allergic encephalomyelitis in the rat. Eur. J. Neurosci. 2002, 15, 1317–1326. [Google Scholar] [CrossRef] [PubMed]
  162. Arkaban, H.; Ebrahimi, A.K.; Yarahmadi, A.; Zarrintaj, P.; Barani, M. Development of a multifunctional system based on CoFe2O4@ polyacrylic acid NPs conjugated to folic acid and loaded with doxorubicin for cancer theranostics. Nanotechnology 2021, 32, 305101. [Google Scholar] [CrossRef]
  163. Arkaban, H.; Shervedani, R.K.; Yaghoobi, F.; Kefayat, A. A nanocomposite theranostic system, consisting of AuNPs@ MnCO3/Mn3O4 coated with PAA and integrated with folic acid, doxorubicin, and propidium iodide: Synthesis, characterization and examination for capturing of cancer cells. Inorg. Chem. Commun. 2021, 128, 108566. [Google Scholar] [CrossRef]
  164. Rahim, M.; Iram, S.; Syed, A.; Ameen, F.; Hodhod, M.S.; Khan, M.S. Nutratherapeutics approach against cancer: Tomato-mediated synthesised gold nanoparticles. IET Nanobiotechnol. 2018, 12, 1–5. [Google Scholar] [CrossRef]
  165. Kolev, S.K.; Aleksandrov, H.A.; Atanasov, V.A.; Popov, V.N.; Milenov, T.I. Surface chemistry of reduced graphene oxide: H-atom transfer reactions. Appl. Surf. Sci. 2021, 567, 150815. [Google Scholar] [CrossRef]
  166. Ginestra, P. Manufacturing of polycaprolactone-Graphene fibers for nerve tissue engineering. J. Mech. Behav. Biomed. Mater. 2019, 100, 103387. [Google Scholar] [CrossRef] [Green Version]
  167. Rauti, R.; Medelin, M.; Newman, L.; Vranic, S.; Reina, G.; Bianco, A.; Prato, M.; Kostarelos, K.; Ballerini, L. Graphene oxide flakes tune excitatory neurotransmission in vivo by targeting hippocampal synapses. Nano Lett. 2019, 19, 2858–2870. [Google Scholar] [CrossRef] [PubMed]
  168. Agarwal, V.; Agarwal, S.; Kaur, R.; Pancham, P.; Kaur, H.; Bhardwaj, S.; Singh, M. In-silico validation and development of chlorogenic acid (CGA) loaded polymeric nanoparticle for targeting neurodegenerative disorders. J. Biomater. Nanobiotechnol. 2020, 11, 279. [Google Scholar] [CrossRef]
  169. Pongrac, I.M.; Ahmed, L.B.; Mlinarić, H.; Jurašin, D.D.; Pavičić, I.; Čermak, A.M.M.; Milić, M.; Gajović, S.; Vrček, I.V. Surface coating affects uptake of silver nanoparticles in neural stem cells. J. Trace Elem. Med. Biol. 2018, 50, 684–692. [Google Scholar] [CrossRef] [PubMed]
  170. Liu, G.; Men, P.; Kudo, W.; Perry, G.; Smith, M.A. Nanoparticle–chelator conjugates as inhibitors of amyloid-β aggregation and neurotoxicity: A novel therapeutic approach for Alzheimer disease. Neurosci. Lett. 2009, 455, 187–190. [Google Scholar] [CrossRef]
  171. Trapani, A.; De Giglio, E.; Cafagna, D.; Denora, N.; Agrimi, G.; Cassano, T.; Gaetani, S.; Cuomo, V.; Trapani, G. Characterization and evaluation of chitosan nanoparticles for dopamine brain delivery. Int. J. Pharm. 2011, 419, 296–307. [Google Scholar] [CrossRef]
  172. García-Pardo, J.; Novio, F.; Nador, F.; Cavaliere, I.; Suárez-García, S.; Lope-Piedrafita, S.; Candiota, A.P.; Romero-Gimenez, J.; Rodríguez-Galván, B.; Bové, J. Bioinspired theranostic coordination polymer nanoparticles for intranasal dopamine replacement in parkinson’s disease. ACS Nano 2021, 15, 8592–8609. [Google Scholar] [CrossRef]
  173. Liu, L.; Li, Y.; Liu, R.; Shen, Q.; Li, Y.; Shi, Z.; Shen, J.; Ji, W.; Zhang, X. Switchable nanoparticle for programmed gene-chem delivery with enhanced neuronal recovery and CT imaging for neurodegenerative disease treatment. Mater. Horiz. 2019, 6, 1923–1929. [Google Scholar] [CrossRef]
  174. Bittner, A.; Ducray, A.D.; Widmer, H.R.; Stoffel, M.H.; Mevissen, M. Effects of gold and PCL-or PLLA-coated silica nanoparticles on brain endothelial cells and the blood–brain barrier. Beilstein J. Nanotechnol. 2019, 10, 941–954. [Google Scholar] [CrossRef]
  175. Shen, Y.; Cao, B.; Snyder, N.R.; Woeppel, K.M.; Eles, J.R.; Cui, X.T. ROS responsive resveratrol delivery from LDLR peptide conjugated PLA-coated mesoporous silica nanoparticles across the blood–brain barrier. J. Nanobiotechnol. 2018, 16, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Mandić, L.; Sadžak, A.; Strasser, V.; Baranović, G.; Domazet Jurašin, D.; Dutour Sikirić, M.; Šegota, S. Enhanced protection of biological membranes during lipid peroxidation: Study of the interactions between flavonoid loaded mesoporous silica nanoparticles and model cell membranes. Int. J. Mol. Sci. 2019, 20, 2709. [Google Scholar] [CrossRef] [Green Version]
  177. Baldim, V.; Yadav, N.; Bia, N.; Graillot, A.; Loubat, C.; Singh, S.; Karakoti, A.S.; Berret, J.-F. Polymer-coated cerium oxide nanoparticles as oxidoreductase-like catalysts. ACS Appl. Mater. Interfaces 2020, 12, 42056–42066. [Google Scholar] [CrossRef]
  178. Chen, X.; Liu, Y.; Wang, L.; Liu, Y.; Zhang, W.; Fan, B.; Ma, X.; Yuan, Q.; Ma, G.; Su, Z. Enhanced humoral and cell-mediated immune responses generated by cationic polymer-coated PLA microspheres with adsorbed HBsAg. Mol. Pharm. 2014, 11, 1772–1784. [Google Scholar] [CrossRef] [PubMed]
  179. Fornaguera, C.; Feiner-Gracia, N.; Calderó, G.; García-Celma, M.; Solans, C. Galantamine-loaded PLGA nanoparticles, from nano-emulsion templating, as novel advanced drug delivery systems to treat neurodegenerative diseases. Nanoscale 2015, 7, 12076–12084. [Google Scholar] [CrossRef]
  180. Sun, W.; Xie, C.; Wang, H.; Hu, Y. Specific role of polysorbate 80 coating on the targeting of nanoparticles to the brain. Biomaterials 2004, 25, 3065–3071. [Google Scholar] [CrossRef] [PubMed]
  181. Li, W.; Qiu, J.; Li, X.-L.; Aday, S.; Zhang, J.; Conley, G.; Xu, J.; Joseph, J.; Lan, H.; Langer, R. BBB pathophysiology–independent delivery of siRNA in traumatic brain injury. Sci. Adv. 2021, 7, eabd6889. [Google Scholar] [CrossRef] [PubMed]
  182. Ren, T.; Xu, N.; Cao, C.; Yuan, W.; Yu, X.; Chen, J.; Ren, J. Preparation and therapeutic efficacy of polysorbate-80-coated amphotericin B/PLA-b-PEG nanoparticles. J. Biomater. Sci. Polym. Ed. 2009, 20, 1369–1380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Wilson, B.; Samanta, M.K.; Santhi, K.; Kumar, K.S.; Ramasamy, M.; Suresh, B. Chitosan nanoparticles as a new delivery system for the anti-Alzheimer drug tacrine. Nanomed. Nanotechnol. Biol. Med. 2010, 6, 144–152. [Google Scholar] [CrossRef]
  184. 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]
  185. Ahlschwede, K.M.; Curran, G.L.; Rosenberg, J.T.; Grant, S.C.; Sarkar, G.; Jenkins, R.B.; Ramakrishnan, S.; Poduslo, J.F.; Kandimalla, K.K. Cationic carrier peptide enhances cerebrovascular targeting of nanoparticles in Alzheimer’s disease brain. Nanomed. Nanotechnol. Biol. Med. 2019, 16, 258–266. [Google Scholar] [CrossRef]
  186. Cano, A.; Ettcheto, M.; Chang, J.-H.; Barroso, E.; Espina, M.; Kühne, B.A.; Barenys, M.; Auladell, C.; Folch, J.; Souto, E.B. Dual-drug loaded nanoparticles of Epigallocatechin-3-gallate (EGCG)/Ascorbic acid enhance therapeutic efficacy of EGCG in a APPswe/PS1dE9 Alzheimer’s disease mice model. J. Control. Release 2019, 301, 62–75. [Google Scholar] [CrossRef]
  187. Cano, A.; Ettcheto, M.; Espina, M.; Auladell, C.; Calpena, A.C.; Folch, J.; Barenys, M.; Sánchez-López, E.; Camins, A.; García, M.L. Epigallocatechin-3-gallate loaded PEGylated-PLGA nanoparticles: A new anti-seizure strategy for temporal lobe epilepsy. Nanomed. Nanotechnol. Biol. Med. 2018, 14, 1073–1085. [Google Scholar] [CrossRef] [PubMed]
  188. Kaur, S.; Manhas, P.; Swami, A.; Bhandari, R.; Sharma, K.K.; Jain, R.; Kumar, R.; Pandey, S.K.; Kuhad, A.; Sharma, R.K. Bioengineered PLGA-chitosan nanoparticles for brain targeted intranasal delivery of antiepileptic TRH analogues. Chem. Eng. J. 2018, 346, 630–639. [Google Scholar] [CrossRef]
  189. Mukherjee, S.; Madamsetty, V.S.; Bhattacharya, D.; Roy Chowdhury, S.; Paul, M.K.; Mukherjee, A. Recent advancements of nanomedicine in neurodegenerative disorders theranostics. Adv. Funct. Mater. 2020, 30, 2003054. [Google Scholar] [CrossRef]
  190. Lang, A.E. Clinical trials of disease-modifying therapies for neurodegenerative diseases: The challenges and the future. Nat. Med. 2010, 16, 1223–1226. [Google Scholar] [CrossRef] [PubMed]
  191. Teleanu, D.M.; Chircov, C.; Grumezescu, A.M.; Teleanu, R.I. Neurotoxicity of nanomaterials: An up-to-date overview. Nanomaterials 2019, 9, 96. [Google Scholar] [CrossRef] [Green Version]
  192. Venkataramani, V.; Doeppner, T.R.; Willkommen, D.; Cahill, C.M.; Xin, Y.; Ye, G.; Liu, Y.; Southon, A.; Aron, A.; Au-Yeung, H.Y. Manganese causes neurotoxic iron accumulation via translational repression of amyloid precursor protein and H-Ferritin. J. Neurochem. 2018, 147, 831–848. [Google Scholar] [CrossRef] [PubMed]
Figure 1. An overview of transport mechanisms across the blood–brain barrier (BBB), adapted from [43], Springer, 2020.
Figure 1. An overview of transport mechanisms across the blood–brain barrier (BBB), adapted from [43], Springer, 2020.
Molecules 28 00841 g001
Figure 2. PS synthesis of Sorbitan, adapted from [76], MDPI, 2021.
Figure 2. PS synthesis of Sorbitan, adapted from [76], MDPI, 2021.
Molecules 28 00841 g002
Figure 3. Structures of several PSs, adapted from [76], MDPI, 2021.
Figure 3. Structures of several PSs, adapted from [76], MDPI, 2021.
Molecules 28 00841 g003
Figure 5. Chemical structure of hyaluronic acid [146].
Figure 5. Chemical structure of hyaluronic acid [146].
Molecules 28 00841 g005
Figure 6. Chemical structure of cyclodextrins [148].
Figure 6. Chemical structure of cyclodextrins [148].
Molecules 28 00841 g006
Figure 7. The 3D structure of HSA [153].
Figure 7. The 3D structure of HSA [153].
Molecules 28 00841 g007
Figure 8. (a) Graph representing the particle size analysis and (b) Zeta potential of the optimized nanoparticles (CGA-NPs), adapted from [168], scientific research, 2020.
Figure 8. (a) Graph representing the particle size analysis and (b) Zeta potential of the optimized nanoparticles (CGA-NPs), adapted from [168], scientific research, 2020.
Molecules 28 00841 g008
Figure 9. Synthesis of the flavonoid-loaded mesoporous silica nanoparticles (MSNs). Stage A: PEGylation of propylamine MSN; Stage B: Loading of flavonoids, adapted from [176], MDPI, 2019.
Figure 9. Synthesis of the flavonoid-loaded mesoporous silica nanoparticles (MSNs). Stage A: PEGylation of propylamine MSN; Stage B: Loading of flavonoids, adapted from [176], MDPI, 2019.
Molecules 28 00841 g009
Table 1. The advantages and disadvantages of the common nanoparticles in brain delivery.
Table 1. The advantages and disadvantages of the common nanoparticles in brain delivery.
ComponentsAdvantagesDisadvantagesRef.
Cerium oxide
  • Manageability.
  • Cost-effectiveness.
  • Less time consuming.
  • Required less energy.
  • Poor ability to overcome the BBB.
[158]
Chitosan
  • Nontoxic.
  • Biodegradable.
  • Biocompatible polysaccharide.
  • Soluble in dilute acids.
  • Helping in complexing the negatively charged therapeutic molecules simply by ionic interactions.
  • Improving cellular uptake of chitosan-derived nanoparticles (proton sponge effect).
  • Poor water solubility.
  • Weak encapsulation efficiency of both hydrophilic and hydrophobic drugs.
[159]
[160]
Gold
  • Predominant optical properties fit for detection/imaging.
  • Well-established synthesis methods.
  • The potential capability to cross the BBB.
  • Preventing cognitive deficits, oxidative stress and inflammation in AD rat model.
  • Relative low cytotoxicity.
  • Finding easy synthesis.
  • Control over size and shape.
  • Improving colloidal stability, and the ability to tune the surface chemistry to achieve easy conjugation.
[78,158]
Silica
  • Large surface area.
  • High structural stability.
  • Easy surface functionalization.
  • Low cost of production.
  • Excellent biocompatibility.
  • Protracted circulation properties.
  • Pore size of mesoporous silica.
  • Decrease toxicity.
  • Tuning particle size, pore size, surface properties, and the porous structure.
  • Endowing MSNs with a large surface area for drug adsorption.
[78]
Table 2. The structure and synthesis method of the coated NPs.
Table 2. The structure and synthesis method of the coated NPs.
ComponentSynthesis MethodStructureRef.
GO/PCLelectrospinningporous framework[166]
GOhummernanosheets[167]
SiO2@PSAionic gelation methodcore-shell[168]
Ag NPsprecipitationcore-shell[169]
Polystyrene---nanosphere–chelator conjugate[170]
Chitosanionic gelationnanospheres[171]
Dopaminereversible coordination complexationcoordination polymer nanoparticles[172]
SiO2@ PLLAStobercore-shell[174]
Polymer-coated cerium oxideprecipitation−redispersionstair-like[177]
Cationic
polymer-coated PLA
emulsification and emulsion solvent evaporationmicrospheres[178]
Table 3. Various polymer-coated nanoparticles for brain-targeted delivery.
Table 3. Various polymer-coated nanoparticles for brain-targeted delivery.
Drug
Carriers
Therapeutic
Agents
DiseasesSurface
Coating
Study ModelReference
PLAFITC-dextranBBBPS 80Kunming
mice
[180]
PLGAsiRNATraumatic
brain injury
PS 80C57BL/6J mice[181]
PLA-b-PEGAmphotericin BCryptococcal
Meningitis
PS 80BALB/c
mice
[182]
ChitosanTacrineAlzheimer’s
Disease
PS 80in vitro release[183]
liposomesGalantamineAlzheimer’s disease and vascular dementiaPEGrat brains[184]
Silica-encapsulated liposomesArsenic trioxideGliomaPAArat brains[160]
MSNsResveratrolParkinson’s diseasePLArat brains[175]
PLGACurcuminAlzheimer’s diseaseChitosanTg2576 mice[185]
PLGAEpigallocatechin gallateAlzheimer’s diseasePEGAPP/PS1 mice[186]
PLGAEpigallocatechin gallateEpilepsyPEGKA–C57BL/6
mice
[187]
PLGATRH analogsEpilepsyChitosanrat brains[188]
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

Bazi Alahri, M.; Jibril Ibrahim, A.; Barani, M.; Arkaban, H.; Shadman, S.M.; Salarpour, S.; Zarrintaj, P.; Jaberi, J.; Turki Jalil, A. Management of Brain Cancer and Neurodegenerative Disorders with Polymer-Based Nanoparticles as a Biocompatible Platform. Molecules 2023, 28, 841. https://doi.org/10.3390/molecules28020841

AMA Style

Bazi Alahri M, Jibril Ibrahim A, Barani M, Arkaban H, Shadman SM, Salarpour S, Zarrintaj P, Jaberi J, Turki Jalil A. Management of Brain Cancer and Neurodegenerative Disorders with Polymer-Based Nanoparticles as a Biocompatible Platform. Molecules. 2023; 28(2):841. https://doi.org/10.3390/molecules28020841

Chicago/Turabian Style

Bazi Alahri, Mehdi, Alhawarin Jibril Ibrahim, Mahmood Barani, Hassan Arkaban, Seyedeh Malahat Shadman, Soodeh Salarpour, Payam Zarrintaj, Javad Jaberi, and Abduladheem Turki Jalil. 2023. "Management of Brain Cancer and Neurodegenerative Disorders with Polymer-Based Nanoparticles as a Biocompatible Platform" Molecules 28, no. 2: 841. https://doi.org/10.3390/molecules28020841

APA Style

Bazi Alahri, M., Jibril Ibrahim, A., Barani, M., Arkaban, H., Shadman, S. M., Salarpour, S., Zarrintaj, P., Jaberi, J., & Turki Jalil, A. (2023). Management of Brain Cancer and Neurodegenerative Disorders with Polymer-Based Nanoparticles as a Biocompatible Platform. Molecules, 28(2), 841. https://doi.org/10.3390/molecules28020841

Article Metrics

Back to TopTop