Back to Journals » International Journal of Nanomedicine » Volume 19

Brain Targeting Nanomedicines: Pitfalls and Promise

Authors Kakinen A , Jiang Y, Davis TP, Teesalu T , Saarma M

Received 12 December 2023

Accepted for publication 15 April 2024

Published 27 May 2024 Volume 2024:19 Pages 4857—4875

DOI https://doi.org/10.2147/IJN.S454553

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Professor Jie Huang



Aleksandr Kakinen,1,2 Yuhao Jiang,2 Thomas Paul Davis,2 Tambet Teesalu,3,4 Mart Saarma1

1Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland; 2Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia; 3Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia; 4Materials Research Laboratory, University of California Santa Barbara, Santa Barbara, CA, USA

Correspondence: Mart Saarma, Institute of Biotechnology, HiLIFE, University of Helsinki, Viikinkaari 5D, Helsinki, 00790, Finland, Tel +358505002726 ; +358294159378, Email [email protected] Aleksandr Kakinen, Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Building 75, Cnr College Road& Cooper Road, St Lucia, QLD, 4067, Australia, Tel +61 7 344 63152, Email [email protected]

Abstract: Brain diseases are the most devastating problem among the world’s increasingly aging population, and the number of patients with neurological diseases is expected to increase in the future. Although methods for delivering drugs to the brain have advanced significantly, none of these approaches provide satisfactory results for the treatment of brain diseases. This remains a challenge due to the unique anatomy and physiology of the brain, including tight regulation and limited access of substances across the blood-brain barrier. Nanoparticles are considered an ideal drug delivery system to hard-to-reach organs such as the brain. The development of new drugs and new nanomaterial-based brain treatments has opened various opportunities for scientists to develop brain-specific delivery systems that could improve treatment outcomes for patients with brain disorders such as Alzheimer’s disease, Parkinson’s disease, stroke and brain tumors. In this review, we discuss noteworthy literature that examines recent developments in brain-targeted nanomedicines used in the treatment of neurological diseases.

Keywords: brain delivery, blood-brain barrier, targeted delivery, nanoparticle, neurodegenerative diseases, stroke, cancer

Introduction

In 1885, Paul Ehrlich made a groundbreaking discovery: intravenous injection of dyes stained all organs except the brain.1 This discovery illuminated the existence of the highly restrictive biological barrier in the central nervous system (CNS) known as the blood-brain barrier (BBB).2 The BBB is essential for protecting the CNS from toxins, drugs, and pathogens. Most of the BBB surface consists of thin capillaries in which tight BBB connections occur between endothelial cells, preventing chemicals from entering the brain paracellularly.3 In addition to endothelial cells, brain cells such as pericytes and astrocytes build and maintain the BBB. The BBB can only be penetrated passively by lipid-soluble solutes that can readily diffuse across the capillary endothelial membrane.4 As a result, about 98% of small molecules, virtually all large molecules and most biological drugs are excluded from the BBB.5 The integrity of the BBB is critical for the proper functioning of the CNS. Conversely, disruption of the BBB is a key element in the development of many brain disorders, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), stroke and several others.6–9

As the world population ages, the global burden associated with neurological disorders increases worldwide, accounting for more than 6% of all diseases10 and costing more than US$1.7 trillion annually in the US and Europe alone.11,12 In recent years, CNS diseases have become a leading cause of disability-adjusted life years and one of the most common causes of death and disability in the world.13 Over the past 30 years, neurological diseases have increased disability-adjusted life years and mortality by 15% and 39%, respectively.14 The number of people with dementia is estimated to increase from ~57 million cases worldwide in 2019 to 152 million cases in 2050.15 AD, PD, stroke, multiple sclerosis and amyotrophic lateral sclerosis (ALS) are just some of the incurable CNS diseases. Most available medications only relieve symptoms and do not affect the progression of the disease. Thus, there is an urgent need for more effective drug delivery methods and disease-modifying drugs that can slow down or stop neurodegeneration and cure neurological disorders. The goal is to stop neurodegeneration and regenerate still alive neurons.

Drug delivery to the CNS remains a major therapeutic challenge. Delivery of therapeutics across the BBB is considered a minimally invasive strategy to target the brain. However, most preclinical and clinical studies to date show the relative lack of success that researchers in this field have achieved. Notably, only 3–5% of brain-directed pharmaceuticals have reached the market in recent years.16 The greatest challenges in development of systemically-administered CNS drugs include low brain delivery due to insignificant BBB permeability,17 increased BBB disruption due to the drug’s pathway of entry into the brain,18,19 and systemic toxicity due to poor drug selectivity. To overcome these challenges, several administration routes have been explored including intranasal and intracranial routes.20–22 Focusing on intravenous administration, nanosized carriers (nanoparticles (NPs), size >100 nm)23 are increasingly being developed for more efficient drug delivery across the BBB for the treatment and diagnosis of a wide range of CNS diseases, such as cancer and neurodegenerative diseases.5,24–26 In addition, different precision targeting strategies (eg, functionalization with antibodies, ligand proteins or homing peptides) have been explored to facilitate drug uptake into the brain.27,28 This review summarizes noteworthy literature examining recent advances in brain drug delivery using nanomaterials for the treatment of neurological diseases.

Current Approaches and Challenges in Brain Drug Delivery

In recent years, the challenges of drug delivery in brain diseases have been increasingly recognized and studied. Despite this rapid progress, barriers to drug delivery often vary by disease and delivery method. The delivery of the drug into the CNS through the cardiovascular system is prevented by physiological and physical obstacles, including the BBB at the level of the cerebrovascular bed and other specialized barriers, such as the blood-spinal cerebrospinal fluid barrier or the blood-tumor barrier. Consequently, even if a compound reaches the brain, its quantity remains below therapeutically relevant concentrations.29,30 One reason for this is the drug’s design, which limits the compound’s ability to reach the brain. The therapeutic agent very often interacts with macromolecules in the body, impairing drug’s stability, therapeutic activity, or penetration through the BBB.31,32 Another problem is the presence of enzymes in the plasma and the CNS tissue that can render the drug inactive.2,33 For example, brain microvessels have similar to liver metabolic activities due to the presence of enzymes like epoxide hydrolase and uridine 5’-diphospho-glucuronosyltransferase.33 In this section, we discuss the architecture of the BBB, as well as brain drug delivery strategies and challenges.

Blood-Brain Barrier (BBB)

The neurovascular interfaces in the brain, spinal cord, retina, and peripheral nerves are essential to maintain the health and functionality of these delicate tissues.34,35 These interfaces are safeguarded by specialized blood-tissue barriers that exclude harmful substances and immune cells while allowing necessary molecules and nutrients to pass through. The BBB is a highly restrictive barrier that is essential for normal hemostasis and optimal functioning of the CNS. Complex cellular and noncellular components work together to keep the BBB functioning. Low permeability of the BBB is controlled by complex crosstalk between brain microvascular endothelial cells (BMEC), pericytes, astrocytes (Figure 1A) with other cell types in the brain parenchyma (eg, microglia, neurons, oligodendrocytes).36 The transmembrane proteins in structures connecting endothelial cells known as tight junctions form a highly selective network that forces the majority of molecular traffic toward a transcellular route (ie, through cells), rather than the paracellular route (ie, between cells).36 Tight junctions are composed of a complex of ~40 different proteins including transmembrane cell adhesion proteins such as claudins, occludins, and junction adhesion molecules.37 Pericytes surrounding the BMEC are in close contact with the endothelial cells and communicate with them through gap and adherens junctions.38–40 Astrocytes are star-shaped cells of ectodermal origin, have processes known as perivascular endfeet that are applied to the walls of microvessels.41 Whereas the astrocytic endfoot cover of the blood vessel is not considered to be a critical element for the physical barrier of the BBB,42 these cells are essential for the formation and maintenance of the BBB by modulating the expression of transporters and receptors and providing factors that contribute to the formation and maintenance of tight junctions between the BMEC.

Figure 1 Schematic structure of the blood-brain barrier (BBB) (A) and transport mechanisms of nanoparticles (NPs) through the BBB (B). NPs can cross the BBB by paracellular pathway, passive transcellular diffusion, carrier-mediated transport, receptor-mediated transcytosis, adsorptive-mediated transcytosis, and cell-mediated transport.

The BBB exhibits distinct microanatomical and physiological features in different brain regions that are also reflected in differences in the BBB barrier function. For example, due to the higher synaptic and metabolic activity of neurons, grey matter has a higher capillary density than white matter.43 In addition, the morphology of astrocytes differs in grey and white matter. Specifically, the most common type of astrocytes in grey matter is protoplasmic, whereas the majority of astrocytes in white matter are fibrous.44 Regional differences are also observed in pericytes. For example, the highest pericyte coverage is present in the cortex, hippocampus and caudate nucleus, whereas the cervical, thoracic and lumbar spinal cord anterior horn capillaries have lower pericyte coverage and show higher permeability than the brain microvessels.45 High diversity of glial sub-populations throughout the CNS was also seen using single-cell transcriptomics.46 Furthermore, the cerebral endothelial cell gene expression diverges across the vascular tree (arterioles, capillaries, and venules). Notably, solute transport-related genes such as monocarboxylate transporter 1 and plasma membrane Ca2+ ATPase Type 2 exhibit higher expression in capillaries compared to venules.47 Cerebral and pial microvessels in the brain also display differences, with pial microvessels lacking astrocytic end feet showing lower barrier function.48

Molecular Transport Across the BBB

The brain cannot synthesize most of its required nutrients and thus relies on supplies from the circulation system. Due to the presence of tight junctions between the endothelial cells of the capillaries, which almost completely exclude the possibility of paracellular transport, most of the molecules essential for the brain cross the BBB through a transcellular route. Small lipophilic molecules with a mass of less than 500 Da can in most cases cross the BBB by simple diffusion.49 Most nutrients, however, do not meet size criteria for passive diffusion and rely on membrane transporters or carriers to extravasate and enter the brain.36,50 These transporters are either concentration-dependent (facilitated diffusion transporters) or energy-dependent (active transporters). The transporter-mediated uptake is typically ~10 times faster than transmembrane diffusion, saturable, and structure-specific. Larger macromolecules cannot follow this pathway and use receptor-mediated transport to pass into and out of the brain. Various receptors on the surface of BMEC bind specific ligands and trigger the uptake of the compounds into a vesicle, with subsequent receptor-mediated uptake and different traffic routes including lysosomal degradation, recycling back to the apical membrane or routing to the basolateral membrane where membrane fusion allows for the release of the vesicle content (transcytosis). The type of receptor and intravesicular environment determine whether endocytosis or transcytosis occurs.51 The heterogeneous distribution of receptors throughout the BBB allows for the transportation of the macromolecules to specific areas of the brain. In addition to physical BBB alteration techniques (eg, using osmotic agents, electromagnetic fields, or microbubbles that oscillate in response to the application of ultrasound, leading to transient opening of the BBB),52,53 the aforementioned transcellular pathways are used to deliver therapeutics into the brain parenchyma.

Strategies to Cross the BBB and Drug Delivery Challenges

Strategic therapeutic approaches for drug delivery to the brain can be divided into two broad categories: BBB bypass and BBB crossing. Perhaps one of the least invasive methods of bypassing the BBB is the intrathecal injection of therapeutic agents into the subarachnoid space of the spinal cord.18 Although the cerebrospinal fluid flows in opposite direction to desired drug delivery, this administration method has been shown to successfully deliver therapeutics to the brain tissue.54 To bypass the BBB, intracerebral administration can also be used, allowing release of the drug into the brain parenchyma through an implant or injection.55 This route can be used for brain delivery of a wide range of compounds and formulations. Although intracerebral administration has some advantages, such as reduced systemic toxicity, its significant disadvantages include poor drug diffusion, and the invasive and risky nature of the procedure that can cause infections. In addition, this method is expensive, requires complicated brain surgery and hospitalization. Intranasal administration is another method of bypassing the BBB. This method is non-invasive and does not require injections, however, clinical observations over the past 40 years have shown inconsistent results.56

In the context of trans-BBB delivery, drugs can enter the CNS via the paracellular pathway, passive transcellular diffusion, carrier-mediated transport, receptor-mediated transcytosis, adsorptive-mediated transcytosis, or cell-mediated transport (Figure 1B). Although, intracellular pores are normally only around 1 nm, in various brain pathologies or as a response to certain physicochemical stimuli, the BBB confluency may be disrupted, increasing drug extravasation and penetration into the brain.6,8,57 As disused above, passive transcellular diffusion is only available for small (below 500 Da) lipophilic molecules. Transporters that are involved in carrier-mediated transport are structure-specific and rarely transport drug analogues. As such, drug molecules must be chemically modified to mimic the normal ligands. Receptor-mediated transcytosis relies on BBB-expressed proteins/receptors to transport therapeutics from the blood to the brain. This delivery strategy may be applied to different targeting ligands such as antibodies, proteins, peptides, and aptamers. Being a non-invasive method, it can be used for repeated drug delivery. Receptor-mediated transcytosis is widely used for the delivery of the NPs across the BBB,58 and its application in targeted drug delivery is discussed in more detail below. Cell-mediated transport differs from previously discussed pathways and relies on the internalization of the therapeutic agent by immune cells such as monocytes or macrophages and subsequent transport across the BBB into the brain.59,60

Targeted Drug Delivery

Improvement of physicochemical or biological properties of drugs is usually achieved by structural alterations of the molecule or, in the case of nanomaterials, modifying the surface of the particle. The purpose of such functionalization can be improved pharmacokinetics (eg, longer half-life and prolonged systemic circulation), better penetration through biological barriers (eg, the BBB) or specific organ/tissue targeting (eg, brain or tumor). To extend the blood circulation half-life of the NP, one of the oldest practices is particle functionalization by polyethylene glycol (PEG).61 Furthermore, noncovalent functionalization of the particle surface with surfactants such as polysorbate 80 (Tween 80) or poloxamer 188 has been shown to guide NPs to the brain via adsorption of apolipoproteins,62 facilitating interaction with BMEC and transport of NPs across the BBB.30,63,64 By counteracting the nonspecific adhesion due to van der Waals interactions, PEGylation opens the opportunity for targeting of NPs with targeting moieties such as antibodies and homing peptides.65,66 These ligands either engage with the endocytosis or transcytosis receptors expressed on the BMEC surface (eg, transferrin, insulin, lactoferrin, and lipoprotein receptors), penetrate directly through cellular membranes (cell-penetrating peptides (CPP)),65,67,68 or used mixed pathways for BBB penetration. NP-CPP conjugates were the first BBB-crossing NPs that did not compromise the integrity of the BBB.69,70 CPP are typically endocytosed, with the conjugated therapeutic agents becoming captured inside endosomes.71,72 Endosomal escape is not fully understood,73,74 and represents a major barrier to the usage of CPP as delivery systems. Strategies to facilitate endosomal escape have been described in a recent review by Ghorai et al.27 Another significant drawback of CPP, lack of cellular selectivity, can be enhanced by the inclusion of receptor-specific modules to target specific organs, tissues or cells.75 Brain-targeting peptides can enhance drug or NP interactions with BMEC, increase uptake, and consequently transport therapeutic payloads across the BBB, delivering drug molecules to the brain.28,76–78 BBB targeting peptides are derived from various sources, such as viral proteins (human immunodeficiency virus (HIV) (HIV-1-TAT),79 rabies virus,80), phage biopanning screens (Pep-22, TGN, G23, T7, THR),81 venom neurotoxins (Apanin, MiniAp),82 and different endogenous proteins (regulon polypeptides, receptor-associated protein, Kunitz protease inhibitor domains).

Nano-Based Approaches Towards Drug Delivery to the Brain

Nanoparticles have been demonstrated to translocate across the BBB either by carrier-mediated transcytosis, adaptive-mediated transcytosis, or receptor-mediated transcytosis (Figure 1B). The ability of the nanoformulated drug to cross the BBB is determined by the physicochemical and biological properties of the NPs, and not by the chemical structure of the drug. The biodistribution of NPs is significantly influenced by their physicochemical characteristics, including particle size, shape, charge, and surface modifications. To improve NP transport across the BBB and enhance cellular uptake, NPs are often decorated with molecules, polymers, or targeting ligands such as small compounds, antibodies or homing peptides.82,83 For example, NPs coated with transferrin, apolipoproteins,64 and insulin-like growth factor II84 have been shown to be more effective at penetrating the BBB than nonfunctionalized NPs.85 Nanomedicine approaches to treat brain diseases are discussed in more detail below.

Application of Nanomaterials for Neurodegenerative Disease Therapy

AD, PD, ALS, and Huntington’s disease (HD) are among the most common and harmful brain disorders. The mechanisms of onset and progression of neurodegenerative diseases still remain an unresolved issue, and disease-modifying treatments are currently lacking.86,87 Immune activation within the CNS and neuroinflammation is associated with neurodegenerative diseases.88 In addition, oxidative stress and endoplasmic reticulum stress induced by aggregated proteins are the most common denominators of neuronal cell loss.89,90 Some genetic risk factors can also contribute to the development and progression of neurodegenerative diseases. For example, Familial Alzheimer’s disease is caused by inherited mutations in presenilin 1 (the most common), presenilin 2 and amyloid precursor protein genes.91 Familial AD accounts only for 5% of all AD cases but leads to early onset of symptoms.92 Similarly, mutations in PARKIN, PINK1, FBXO7, PARK7, GBA1 or leucine-rich repeat kinase 2 genes are the most common causes of familial PD.93 Mutations in alpha-synuclein (αSyn) gene, which encodes αSyn protein, have been found to be linked to familial forms of PD.94

Effective treatments for brain diseases are limited due to difficulties in drug delivery. AD, PD, ALS, and HD are complex, multifactorial diseases which have prompted researchers to design multitarget ligands to address the complementary pathways involved in these disorders. In recent years, nanomaterials have found widespread use in several biomedical fields, including neurodegenerative diseases.29 Pathological misfolding of proteins into amyloid fibrils is a hallmark of several neurodegenerative diseases, including amyloid-beta (Aβ) peptide in AD and αSyn protein in PD. The amyloid hypothesis suggests that an abnormal accumulation of misfolded proteins in the brain causes nerve cell death. In AD, the accumulation of extracellular Aβ in senile plaques and intracellular tau in neurofibrillary tangles over time correlated with memory loss and cognitive decline.95 Similarly, misfolding and aggregation of αSyn in Lewy bodies, the hallmark of PD, mediate disruption of cellular homeostasis and can cause degradation and death of dopamine neurons in PD.96,97 Inhibition of amyloid protein aggregation has become one of the popular approaches for the potential treatment of amyloid disease. For example, the US Food and Drug Administration recently approved human monoclonal antibodies aducanumab98 and lecanemab99 which target various forms of aggregated Aβ. These antibodies were able to reduce amyloid markers in the early stages of AD but showed modest effect on cognitive functions and were associated with side effects such as brain shrinkage and blood clotting.100 This suggests that there is still a lot of space for the development of new therapies against AD.101

The use of nanotechnology in drug delivery has opened various opportunities to develop better therapeutic agents. For instance, dual functionalization of nanocarriers has been actively explored in past years, demonstrating promising dual-targeting applications. In a recent study, NPs coated with red blood cell membranes and functionalized with a blood-brain barrier-crossing peptide TGNYKALHPHN were used for brain delivery of curcumin, a therapeutic agent for AD.102 The incorporation of red blood cell membrane coating not only prolonged the circulation half-life of NPs but also resulted in lower immunogenicity compared to nanocarriers relying on synthetic materials. Compared to untargeted nanoformulation, the BBB-targeting resulted in an ~8-fold increase in curcumin delivery to the brain and resulted in enhanced spatial learning and memory in a mouse model of AD.

PEGylated superparamagnetic iron oxide NPs (SPION) conjugated with Aβ oligomer‑specific single-chain variable antibody W20 and class A scavenger receptor activator XD4 (inhibit Aβ aggregation) decreased Aβ aggregation and significantly reversed cognitive deficits as well as ameliorated neuropathology in AD mice (Figure 2A).103 Furthermore, multifunctional theranostic platform utilizing polymer-coated SPION, coupled with curcumin and decorated with BBB-crossing (CRTIGPSVC) and Aβ targeting (QSHYRHISPAQV) peptides not only successfully traversed the BBB, inducing neuroprotection, but also detected Aβ deposits in vivo through magnetic resonance imaging and reduced plaque burden (Figure 2B).104 Liu et al synthesized zwitterionic poly(carboxybetaine)-based NPs targeted by mannose analog 4-aminophenyl α-d-mannopyranoside to guide the NPs to penetrate through the BBB and target to microglia, and functionalized by positively charged reactive oxygen species (ROS) responsive polymer poly[(2-acryloyl)ethyl(p-boronic acid benzyl)dimethylammonium bromide].105 The NPs normalized dysfunctional microglia via two synergistic approaches: 1) fingolimod (immunomodulating medication) and signal transducer and activator of transcription 3 siRNA based inflammatory modulation, and 2) poly(carboxybetaine)-based Aβ microglial recruitment. The synergistic strategy significantly inhibited crosstalk between microglia and Aβ, which attenuated the AD pathology (Figure 2C).

Figure 2 Application of nanomaterials against Alzheimer’s diseases. (A) W20/XD4-SPION attenuated Aβ pathology in the brains of AD mice. Adapted from Liu X-G, Zhang L, Lu S, et al. Superparamagnetic iron oxide nanoparticles conjugated with Aβ oligomer-specific scFv antibody and class A scavenger receptor activator show therapeutic potentials for Alzheimer’s Disease. J Nanobiotechnology. 2020;18(1):160. Creative Commons.103 Schematic illustration and morphology (TEM, scale bar 50 nm) of W20/XD4-SPION (left). 6E10 immunostaining for plaques in the brains of AD mice treated with SPION reduced Aβ burden (right). Scale bars 400 μm. (B) Significantly reduced Aβ deposition after treatment by curcumin-loaded and decorated with BBB-crossing (CRTIGPSVC) and Aβ targeting (QSHYRHISPAQV) peptides SPION in double transgenic mice (APP/PS1 mice). Adapted from Ruan Y, Xiong Y, Fang W, et al. Highly sensitive Curcumin-conjugated nanotheranostic platform for detecting amyloid-beta plaques by magnetic resonance imaging and reversing cognitive deficits of Alzheimer’s disease via NLRP3-inhibition. J Nanobiotechnol. 2022;20(1):322. Creative Commons.104 (C) Schematic illustration and TEM image of the MCPZFS NPs; scale bar: 200 nm (left). MCPZFS NP treatment could significantly attenuate cognitive and memory impairment of AD transgenic mice as demonstrated by Morris water maze test (middle and right). Adapted from Liu R, Yang J, Liu L, et al. An “Amyloid-β Cleaner” for the Treatment of Alzheimer’s Disease by Normalizing Microglial Dysfunction. Adv Sci (Weinh). 2019;7(2):1901555. Creative Commons.105 Data are presented as the mean ± SD. *P < 0.05, ***P < 0.001.

Recently, Lei and colleagues presented an oral brain-targeting delivery system designed for multitarget treatment of AD, coupled with a glucose control strategy (Figure 3A).106 These PEGylated polymeric NPs effectively traversed the intestinal epithelial barrier and the functionalization with mannose allowed for glucose transporter 1-dependent crossing of the BBB (Figure 3B). In the brain, the released drug load (fingolimod) transformed the microglia’s polarization from pro-inflammatory M1 to anti-inflammatory M2, normalized activated astrocytes, contributed to the clearance of toxic Aβ protein, and mitigated oxidative stress and neuroinflammation. In another study, polymeric NPs were dual-targeted with a BBB-penetrating peptide derived from the rabies virus and a neuronal-targeting Tet1 peptide.107 This nanoformulation demonstrated effective β-site amyloid precursor protein cleaving enzyme-1 (BACE1) or glycogen synthase kinase 3β (GSK3β) siRNA delivery, leading to the suppression of the expression of the Aβ precursor protein cleaving enzyme. This innovative approach significantly ameliorated behavioural deficits in a mouse model of AD. Similarly, nanomedicines based on rabies virus glycoprotein derived peptide and angiopep-2 decorated hydroxide NPs were employed for the delivery of a siRNA and anti-inflammatory drugs. This novel approach successfully inhibited the aggregation of Aβ and mitigated neuroinflammation, contributing synergistically to restore memory and cognitive deficits in AD mice.108

Figure 3 Application of nanomaterials against Alzheimer’s diseases. (A) Schematic diagram of mannose-guided nanoparticles (NPs) reaching the brain via oral administration and (B) organ biodistribution of (a) only PEGylated NPs, (b) mannose functionalized NPs and (c) mannose functionalized in combination with glucose control strategy by administering glucose 1 h after NP administration. Reprinted from Lei T, Yang Z, Jiang C, et al. Mannose-Integrated Nanoparticle Hitchhike Glucose Transporter 1 Recycling to Overcome Various Barriers of Oral Delivery for Alzheimer’s Disease Therapy. ACS Nano. 2024;18(4):3234–3250. Copyright © 2024 American Chemical Society.106 By implementing a glucose control strategy, NP accumulation at AD lesions was enhanced, resulting in highly efficient oral brain-targeting. (C-D) Delivery of the nerve growth factor (NGF) gene through the blood–brain barrier resulted in a notable reduction in Aβ accumulation in mice with Alzheimer’s disease (AD) carrying the APP/PS1 mutation. Liposomes decorated with penetratin and transferrin (PenTf), exhibited effective transfection capabilities. This formulation demonstrated a remarkable capacity to significantly decrease Aβ levels in the brains of APP/PS1 mice. Reprinted from Rodrigues BDS, Kanekiyo T, Singh J. Nerve growth factor gene delivery across the blood–brain barrier to reduce beta amyloid accumulation in AD mice. Mol Pharm. 2020;17(6):2054–2063. Copyright © 2020 American Chemical Society.109 Data are expressed as mean ± SEM (n = 14/group). Statistically significant differences (p < 0.05) are shown as (*). (E) Liposomes decorated with rabies virus glycoprotein peptide (CCP) and mannose showcased exceptional permeability across the blood-brain barrier (BBB) for gene delivery. Following a single tail vein administration in C57BL/6 mice, these specialized liposomes demonstrated a remarkable twofold increase in Apolipoprotein E expression in the brain. Reprinted from Arora S, Layek B, Singh J. Design and validation of liposomal ApoE2 gene delivery system to evade blood–brain barrier for effective treatment of Alzheimer’s disease. Mol Pharm. 2021;18(2):714–725. American Chemical Society.114

Furthermore, Rodrigues et al designed dual-modified liposomes (transferrin and CPP functionalized) for nerve growth factor gene delivery (Figure 3C) and reported successful brain delivery in vivo, cell transfection, and a significant decrease in toxic Aβ species (Figure 3D).109 Nerve growth factor is important for the treatment of AD because it can reverse atrophy, prevent degeneration, and stimulate the function of cholinergic neurons in the basal forebrain.111,112 In addition, nerve growth factor levels in the basal forebrain are reduced in patients with AD.109 Recently, Andrade et al found that caffeic acid-loaded transferrin-functionalized liposomes could prevent Aβ fibrillization and disaggregate mature fibrils.113 While transferrin has been extensively explored as a brain-targeting ligand for nanomedicines, several factors limit its effectiveness. One prominent issue that limits the capacity of the delivery is the potential saturation of transferrin receptors on the BBB and brain cells. Furthermore, widespread expression of transferrin receptors in different tissues results in accumulation of transferrin receptor targeting ligands in tissues other than brain and triggers potential side effects. Ongoing research aims to refine and optimize transferrin-based strategies, exploring modifications and combinations with other targeting ligands to enhance delivery capacity, specificity, improve transport across the BBB, and mitigate potential immunogenic responses.

Lei et al developed multifunctional self-assembled peptide micelles decorated with ROS scavenger thioketal-glutathione, inhibitor for Aβ aggregation LPFFD, and autophagy activator for degrading Aβ derived from autophagy protein Beclin 1.110 The resultant nanoformulation showed transport across the BBB, ability to decrease the Aβ plaques and eliminate ROS for AD prevention and treatment. In another study, liposomes were loaded with apolipoprotein E plasmid and functionalized with CPP and glucose transporter 1 targeting ligand mannose to enhance brain-targeting and cellular internalization (apolipoprotein E reduces AD risk, delays the onset of disease, and attenuate the clearance of toxic Aβ proteins from the brain) (Figure 3E).114 Approximately 2-fold protein expression was observed in the neurons treated with the liposomal formulation. Notably, the inclusion of apolipoprotein E on the outer layer of nanomedicines enhances drug delivery to the brain and uptake by nerve cells. For example, Yu et al developed apolipoprotein E-coated nano micelles delivering Oridonin and Phillyrin to the brains of AD mice, resulting in improved cognitive abilities, alleviated Aβ deposition, and reduced neuroinflammation.115 Despite being a widely studied brain-targeting ligand, apolipoprotein E poses certain challenges for nanomedicine applications. One key limitation is the polymorphic nature of the apolipoprotein E gene, resulting in different isoforms (ApoE2, ApoE3, ApoE4) with distinct affinities for receptors. This genetic diversity may influence the binding efficiency of apolipoprotein E-conjugated nanocarriers to receptors on the BBB, potentially affecting the ligand’s reliability and consistency in brain targeting. Moreover, analogous to transferrin, apolipoprotein E receptors are not exclusive to the BBB; they are expressed in various tissues throughout the body. This non-specific distribution may lead to off-target effects, reducing the selectivity of apolipoprotein E mediated nanomedicines for brain tissues. In overcoming these hurdles, ongoing research seeks to refine apolipoprotein E-based strategies, exploring isoform-specific modifications and combination approaches with other ligands to enhance specificity, improve transport efficiency across the BBB, and mitigate potential off-target effects and immunogenic responses.

Several other approaches have been explored in the brain-targeted nanomedicines again neurogenerative diseases. For example, pH-responsive glucose-functionalized polymer nanomicelles enhanced the delivery of anti-Aβ aggregation 3D6 antibody fragments to the brain by up to 41-fold compared with free antibodies.116 Likewise, Zhou et al used glycosylated siRNA nanoformulations to target BACE1 (β-site amyloid precursor protein cleavage enzyme 1) in amyloid precursor protein/PS1 mice and observed effective delivery of the therapeutics to the brain, ameliorating AD-like pathology.117

Nanomedicines have also been developed to target phosphorylated tau and αSyn deposition in PD. In a recent study, phosphatidylserine liposomes encapsulated with astragaloside IV (decrease αSyn expression) and nesfatin-1 (anti-apoptotic characteristics by suppressing rotenone-induced αSyn aggregation), and grafted with wheat germ agglutinin (BBB transport via receptor-mediated endocytosis) and leptin (BBB transport by either direct penetration or transcytosis via interaction with leptin receptor) demonstrated excellent BBB permeability, protection of dopamine neurons from apoptosis, reduced expression of αSyn and phosphorylated tau in vivo.118 Gao et al applied surface-modified MgO NPs for targeted treatment of PD.119 The NPs were coated with polydopamine and plasmid DNA, and functionalized by PEG, lactoferrin (to improve solubility and target the BBB) and puerarin (to inhibit oxidative stress and cell apoptosis). This multi-targeting nanomedicine inhibited cell apoptosis by suppressing the expression of αSyn and reducing ROS levels. In addition, NPs showed neuroprotection and recovery of the motor functions in (1-methyl-4-phenyl-1,2,3,6- tetra hydropyridine induced PD mouse model. Liposomes decorated with transferrin were employed for the targeted delivery of an anti-αSyn antibody to the brain.120 These brain-targeted liposomes achieved a sevenfold increase in antibody concentration within brain cells, leading to reduced αSyn aggregation, alleviated neuroinflammation, and improved behavioural motor function and learning ability in mice. Similarly, liposomes modified with transferrin and rabies virus glycoprotein demonstrated improved BBB-targeting and delivery of epigallocatechin gallate, effectively suppressing αSyn aggregation.121

Notably, L-DOPA (levodopa), a BBB-permeable drug used to increase dopamine levels in the brain for the treatment of PD and alleviate motor symptoms, has also been used as a brain-targeting ligand for nanovehicles.122,123 For example, L-DOPA-functionalized AuNPs can effectively cross the BBB in vitro and be internalized by brain macrophages without causing inflammation, demonstrating potential application as non-inflammatory BBB-penetrating nanocarriers for efficient delivery of therapeutics to the brain.123 Similarly, liposomes guided by L-DOPA and cholesterol were designed for DNA delivery, ensuring targeted delivery to dopamine neurons of αSyn downregulating genes.124 These biomimetic NPs exhibited high BBB permeability, evasion of the cellular reticuloendothelial system, and efficient targeting of dopamine neurons. Despite its potential as a ligand for brain-targeted nanomedicines, L-DOPA presents certain challenges that need consideration. The enzymatic metabolism of L-DOPA poses a challenge for sustained and targeted drug delivery. L-DOPA is a precursor to dopamine, and its rapid conversion can occur systemically, reducing the availability of intact L-DOPA for brain targeting. Finally, L-DOPA treatment alleviates motor symptoms but does not slow down or stop neurodegeneration.

Another potential neuroprotective strategy for supporting and protecting neurons in neurological diseases such as PD involves delivering neurotrophic factors (NTF) or NTF-encoding genes to the brain.29,125,126 Although the clinical potential of NTF has been proven,97,127 a major obstacle remains the inability of NTF to cross the BBB.128 To overcome this limitation, NTF delivery via nanocarriers has been explored in recent years.29 For example, an ultrasound-responsive neurotrophic factor-loaded microbubble-liposome complex was able to improve behavioural impairments and prevent to loss of dopamine neurons in PD mouse model.53 However, despite ongoing research, available reports on nano-based NTF delivery methods are still limited. Combining NPs with DNA or RNA encoding NTF and decorating these NPs with brain-homing peptides would be very promising. In the case of NTF, it is also important that they reach the brain neurons that naturally secrete NTF.

Despite these advances, challenges persist in the field of brain delivery for neurodegenerative diseases. Achieving optimal drug distribution within the intricate neural networks, addressing the heterogeneity of neurodegenerative diseases, and ensuring the long-term safety of nanomedicines are critical hurdles. The complexity of the BBB and its dynamic nature present challenges in sustaining effective drug concentrations over time. Furthermore, concerns about potential immunogenicity and long-term toxicity associated with nanomaterials call for rigorous investigation and safety assessments.

Future perspectives in brain-targeted nanomedicines for neurodegenerative diseases necessitate innovative solutions. Tailoring nanomedicine formulations to target specific molecular signatures of diseases, employing multifunctional nanocarriers, and integrating real-time imaging techniques for precise drug tracking represent promising directions. Given that the key targets of modern nanomedicines are amyloid proteins (tau and Aβ) and neuroinflammation, there is an urgent need for new drugs with multi-therapeutic capabilities that could prevent early disease progression, rather than just treat symptoms. Collaborations between interdisciplinary research teams, combining expertise in nanotechnology, neuroscience, and pharmacology, can propel the development of transformative therapies. As nanomedicine continues to evolve, it holds immense potential in reshaping the landscape of neurodegenerative disease treatment, providing hope for improved patient outcomes and quality of life.

Nanomedicine in Brain Cancer

Brain cancers and tumors are another area in which nanomedicines have great therapeutic promise. For example, chemotherapeutic drugs for the treatment of brain tumors suffer from poor bioavailability, insufficient BBB permeability, and a lack of selective targeting, which inevitably leads to low efficacy and systemic toxicity of chemotherapy. Notably, the average life expectancy for patients with glioblastoma, the most common form of brain cancer, is less than 2 years.129 Despite the widespread observation of brain-impenetrant contrast material accumulation in high grade gliomas, the clinical data consistently reveals a significant tumor burden in brain tumors with an intact BBB.130 Consequently, drugs with poor BBB permeability fail to achieve therapeutically effective exposures in these tumor regions and new drug delivery strategies are needed to achieve more efficient therapeutic effects against brain tumors.

To target both the BBB and blood-brain tumor barrier, nanostructured dual-targeting lipid carriers were developed to deliver Bortezomib, a proteasome inhibitor anti-cancer agent. Specifically, the surface of targeted NPs modified with BBB-targeting 8-residue D-peptide and RI-VAP ligand of glucose-regulated protein 78 (GRP78), with superior glioma-homing property, could simultaneously cross the BBB and the blood-brain tumor barrier with high efficiency and target the glioma cells with excellent selectivity in vitro and in vivo.131 Another dual-modified liposomes for anti-cancer drug delivery (doxorubicin) were developed via surface-functionalization by glucose and mitochondria-targeting ligand triphenylphosphonium.132 Designed NPs demonstrated excellent targeting and internalization ability for tumor cells, endo/lysosomal escape and superior mitochondria targeting capability.

Angiopep-2 is being actively explored as a brain-targeting ligand for anti-glioma nanomedicines.133–139 For example, polymeric NPs, featuring dual surface modification with Angiopep-2 and L-histidine to facilitate BBB-to-glioblastoma transport and endosomal escape, successfully delivered the docetaxel drug to glioblastoma, resulting in ~12-fold increase in tumor cell uptake (Figure 4A).140 Furthermore, a biomimetic nanomedicine was created using Angiopep-2 peptide-decorated red blood cell membranes and pH-sensitive dextran NPs for the targeted delivery of Navitoclax (anti-cancer drug) and the inducer of myeloid leukemia cell differentiation protein-specific inhibitor A-1210477 to the brain.141 This nanomedicine demonstrated effective BBB penetration and successfully inhibited tumor growth, leading to prolonged survival in mice (Figure 4B). Similarly, gold surface-coated iron titanium-shell NPs conjugated with Angiopep-2 exhibited outstanding brain-targeting capabilities and applicability for magnetic resonance imaging.142 In another study, Angiopep-2 modified liposome-silica hybrid nanovehicles functionalized with polyacrylic acid (pH-sensitive release) and loaded with arsenic trioxide (ATO, glioma drug) achieved targeted delivery and on-demand release of ATO for brain glioma therapy (Figure 4C).137 A similar but ROS-responsive and Angiopep-2-targeted siRNA delivery system was developed by Zheng et al.135 This BBB-permeable polymeric siRNA nanomedicine stabilized by triple interactions (electrostatic, hydrogen bond, and hydrophobic), showed efficient release of siRNA resulting from tumor-derived ROS triggered sequential destabilization of the nanocarrier. In addition, siRNA against epidermal growth factor receptor and programmed cell death ligand-1 for combined targeted and immunotherapy against glioblastoma was delivered using radiation-induced targeted NPs.143 A short burst of radiation therapy enhanced the uptake of NPs by glioblastoma cells, resulting in activation of immune response, inhibition of tumor growth, and increased survival of mice.

Figure 4 Application of brain-targeted nanomedicines for brain cancer. (A) L-histidine (His) functionalized and pH-responsive acid-cleavable angiopep-2 (ANG2) NPs showed superiors BBB-permeability and brain accumulation in comparison to non-functionalized or non-cleavable NPs. Adapted from Martins C, Araújo M, Malfanti A, et al. Stimuli-Responsive Multifunctional Nanomedicine for Enhanced Glioblastoma Chemotherapy Augments Multistage Blood-to-Brain Trafficking and Tumor Targeting. Small. 2023;19(22):2300029. © 2023 The Authors. Small published by Wiley-VCH GmbH.140 (B) Schematic illustration of the biomimetic nanomedicine penetrating the BBB and targeting the tumor site via the specific recognition of apolipoprotein E peptide and the multiple low-density lipoprotein receptor (eg, LDLR, LRP1, and LRP2) overexpressed in the BBB endothelial cells and glioblastoma (GBM) cells (upper). Evaluation of the tumor inhibition rate after five successive injections of NPs (bottom). Data are presented as the mean ± SD (*p < 0.05 and ***p < 0.001). Adapted from He W, Li X, Morsch M, et al. Brain-Targeted Codelivery of Bcl-2/Bcl-xl and Mcl-1 Inhibitors by Biomimetic Nanoparticles for Orthotopic Glioblastoma Therapy. ACS Nano. 2022;16(4):6293–6308. Copyright © 2022 American Chemical Society.141 (C) Angiopep-2-conjugated hybrid nanovehicles targeted glioma cells and delivered arsenic trioxide reducing necrosis of tumor tissue in the brain. Reprinted from Tao J, Fei W, Tang H, et al. Angiopep-2-conjugated “core–shell” hybrid nanovehicles for targeted and pH-triggered delivery of arsenic trioxide into glioma. Mol Pharm. 2019;16(2):786–797. Copyright © 2019 American Chemical Society.137

Transferrin and affinity ligands targeting its receptor have also been investigated as targeting ligand in anticancer nanomedicines. For instance, PEGylated NPs functionalized with transferrin and loaded with temozolomide revealed enhanced BBB penetration and glioblastoma targeting ability in vivo.83 Similarly, transferrin-modified and asiatic acid-loaded poly(lactic-co-glycolic acid) NPs showed increased selectivity of NPs toward glioblastoma cells by enhancing their uptake through the transferrin-mediated endocytosis mechanism.144 transferrin. Earlier, Kim et al demonstrated that temozolomide-loaded liposomes modified with anti-transferrin single-chain variable fragments can deliver tumor suppressor p53 gene to limit chemoresistance and prolong survival in a mouse model of glioblastoma.145 p53 inhibits DNA-repair protein O6-methylguanine-DNA methyltransferase activity which can repair DNA damage caused by temozolomide, reversing the therapeutic effects.146 Notably, this is the only anti-glioblastoma drug delivery system using transferrin to reach clinical stage (ClinicalTrials.gov Identifier: NCT02340156).

Conjugation with tumor-homing peptides can also increase tumor tropism and antitumor therapeutic efficacy of NPs. For example, clinical-stage tumor penetrating peptide iRGD that accumulates in vascular endothelial cells and tumor cells positive for expression of angiogenic integrins and Neuropilin-1,68 has been demonstrated to increase glioma accumulation and preclinical therapeutic efficacy of several nanosystems including temozolomide-loaded polyhedral oligomeric silsesquioxane NPs,147 tirapazamine and zinc phthalocyanine NPs,148 and NIR-II phototheranostics.149 Whereas iRGD is known as a generic solid tumor homing peptide, in brain tumors it has the added value of facilitating the NP penetration of the BBB.150 Beyond iRGD, several other tumor homing peptides have demonstrated effectiveness in NP delivery to glioma lesions. Examples include the LinTT1 peptide, which targets Neuropilin-1, and p32, enhancing glioma homing of iron oxide nanoworms and albumin-paclitaxel NPs across a spectrum of glioblastoma models, ranging from infiltratively-disseminating to angiogenic phenotypes.151 Recently, silica NPs functionalized with the cell penetrating TAT peptide demonstrated promising outcomes in delivering the chemotherapeutic agent methotrexate to the brain.152 Additionally, the extracellular matrix-targeting PL1 peptide has shown engagement with tumor-associated isoforms of extracellular matrix molecules Tenascin C and Fibronectin in glioblastoma models.153 These peptides contribute to the advancement of targeted drug delivery strategies for various glioma subtypes. To enhance tumor specific delivery, cyclo(-Arg-Gly-Asp-D-Phe-Cys) peptide was used as the targeting ligand specific to tumor integrin αvβ3 overexpressed on angiogenic endothelial cells in the tumor vasculature.154 Iron oxide NPs conjugated with this tumor-specific peptide exhibited an impressive tumor delivery 11.5%ID/g of a water-insoluble DNA topoisomerase I inhibitor, resulting in over 40% inhibition of tumor growth.

Effective therapy for brain tumors in general remains a medical challenge and requires concentrated research efforts. The development of multitarget drugs for the treatment of glioblastoma remains a very promising therapeutic approach. However, despite notable progress and advancements, challenges persist in brain cancer nanomedicine. One important contributing factor that applies to affinity targeting ligands may be the limited capacity of receptors in the target tissue, imposing constraints on the number of conjugates that can home to the target.155 Once these receptors are saturated, the specific delivery plateaus, and off target effects ensue due to the presence of remaining conjugates circulating in the system. In tumors, this problem may be further compounded by inadequate drug penetration. iRGD peptide, and potentially other tumor penetrating C-end Rule peptides, may provide a path around this limitation to achieve precision drug delivery at therapeutically relevant concentrations.156 In addition, overcoming the heterogeneity of brain tumors, improving drug release kinetics, and ensuring sustained efficacy remain focal points. Future perspectives involve refining targeting ligands, exploring combination therapies, and leveraging emerging technologies like artificial intelligence for personalized treatments. Collaborative efforts among researchers, clinicians, and industry partners will be pivotal in realizing the full potential of brain-targeted nanomedicines for enhanced brain cancer therapy.

Nanomedicine for Stroke Treatment

A stroke is a severe and acute brain condition that can result in disability and death. Strokes are broadly categorized into two main types: hemorrhagic and ischemic. Hemorrhagic stroke results from bleeding caused by the rupture of a blood vessel in the brain and is associated with severe morbidity and high mortality.157 An ischemic stroke is a critical and sudden brain condition that can result in disability and death. It is marked by a sudden reduction in cerebral blood flow caused by an obstruction in the cerebral blood vessels, leading to damage to nerve tissue.158 Lack of blood flow causes brain cell dysfunctions, oxidative stress, and neurological damage.159 Dead neuronal cells release damage-associated molecular patterns that activate the innate and adaptive immune systems, triggering inflammatory pathways. Although BBB is compromised in stroke, the BBB opening is heterogeneous and stroke treatment has limitations due to poor ability to deliver therapeutic agents.160 Therefore, efforts have been made to develop new brain-targeted drug delivery methods. Recently, nanotechnology emerged as an innovative drug delivery tool to improve the treatment of ischemic stroke. Most research has focused on the treatment of ischemic stroke, as it is the most common form of the disease in Europe and America but not in Asia.161 Treatment of ischemic stroke aims to quickly restore blood flow to the brain and is an important strategy to protect neurological damage.162 Neuroprotective approaches include reducing immune cell adhesion, blocking of proinflammatory cytokines, reducing lipid peroxidation, and decreasing cell apoptosis. NP-drug delivery systems have been developed to overcome the short plasma half-life time and low BBB permeability of anti-stroke drugs.

To tackle this challenge, Wang et al innovatively crafted polymeric micelles with surface modification using cyclo(Arg-Gly-Asp-d-Tyr-Lys) peptide, enabling efficient BBB penetration via transcytosis.163 Additionally, these micelles incorporated triphenylphosphine for mitochondria targeting. This acid-responsive formulation effectively mitigates oxidative stress and inflammation by enhancing the delivery of resveratrol to microglia mitochondria, thereby reversing the microglia phenotype through the scavenging of ROS for the treatment of ischemia-reperfusion injury (Figure 5A). To target ischemic brain, Lv et al designed ROS-responsive boronic red blood cell membrane shell polymeric NPs decorated with stroke homing peptide (SHp) for delivery of a neuroprotective agent NR2B9C.164 These bioengineered NPs improved neuroscores and infarct volume in response to surgical middle cerebral artery occlusion injury, reducing ischemic brain damage (Figure 5B). Angiopep-2 is also studied as a ligand to guide stroke medicines aggress the BBB. For instance, Angiopep-2 conjugated to PEGylated edaravone-loaded ceria NPs, mediated BBB transcytosis and ROS-scavenging effect (Figure 5C). 165 Edaravone-loaded ceria NPs could not only enhance the intracephalic uptake but also prevent the injuries on both brain tissues and BBB in strokes.

Figure 5 Brain-targeted nanomedicines against stroke. (A) Schematic illustration of brain-targeted micelles releasing a long PEG chain to expose triphenylphosphine at pH 5.0 (upper). By T2-weighted magnetic resonance imaging (MRI), the recovery of brain injury was remarkably enhanced after treatment with resveratrol-loaded micelles, reducing the infarct area by 65.7% than in the saline group (bottom). Adapted from Wang Z, Pan J, Yuan R, Chen M, Guo X, Zhou S. Shell-Sheddable Polymeric Micelles Alleviate Oxidative Stress and Inflammation for Enhanced Ischemic Stroke Therapy. Nano Lett. 2023;23(14):6544–6552. Copyright © 2023 American Chemical Society.163 Data are presented as mean ± SD. One-way ANOVA was used to calculate P values (*P < 0.05, **P < 0.01, ***P < 0.001). (B) The nanocarrier composed of a dextran polymer core modified with ROS-responsive boronic ester (PHB) and a red blood cell (RBC) membrane shell with stroke homing peptide (SHp) inserted (scheme design upper panel), enhance targeting of ischemic area and reduce ischemic brain damage (lower panel). Adapted from Lv W, Xu J, Wang X, Li X, Xu Q, Xin H. Bioengineered boronic ester modified dextran polymer nanoparticles as reactive oxygen species responsive nanocarrier for ischemic stroke treatment. ACS nano. 2018;12(6):5417–5426. Copyright © 2018 American Chemical Society.164 Statistical analysis used one-way ANOVA test (**P < 0.01). (C) Ceria NPs, which were loaded with edaravone and modified with Angiopep-2 and PEG on their surface (E-A/P-CeO2), demonstrated great intracephalic uptake, and ability to prevent the injuries on both brain tissues and BBB in strokes. Adapted from Bao Q, Hu P, Xu Y, et al. Simultaneous blood–brain barrier crossing and protection for stroke treatment based on edaravone-loaded ceria nanoparticles. ACS Nano. 2018;12(7):6794–6805. Copyright © 2018 American Chemical Society.165

The search for new therapeutic strategies for the treatment of stroke has increased significantly, and combining these treatments with the effective solutions offered in the field of nanotechnology will help in finding new effective therapeutics to improve the outcome of stroke patients. However, despite the success of improving stroke therapy with various nanomedicines in preclinical studies, none of these systems have yet been used to benefit patients. Looking ahead, a multifaceted approach integrating advances in nanotechnology, targeted ligands, and formulation engineering, alongside a deeper understanding of the complex biology of stroke, holds the key to unlocking the full potential of brain-targeted nanomedicine for stroke treatment.

Conclusion and Perspectives

Targeting drugs to the brain is one of the most challenging problems in pharmaceutical research due to the highly selective permeability of the BBB. Obstacles encountered in drug delivery to the brain have been significantly improved by recent developments in drug targeting techniques. In addition, high therapeutic drug concentrations in the brain must be achieved through an effective and safe carrier. Thus, there is a need for good vehicles that deliver the drug across the BBB at an effective concentration without causing systemic side effects. Nanomedicines have been implicated in solving problems associated with targeted drug delivery for the treatment of neurological disorders. Different classes of nanomaterials have been explored as drug nanocarriers across the BBB, and various targeting ligands have shown great promise for drug delivery to the brain. Notably, targeting ligands, such as peptides and antibodies, have shown substantial promise in enhancing the specificity of drug delivery to the brain. The ongoing exploration and refinement of nanotechnology in conjunction with efficient brain-targeting methods hold immense potential to reshape our therapeutic approach to neurological disorders.

However, it is imperative to critically analyze the existing situation and acknowledge persisting challenges. The immunogenicity and potential toxicity of nanocarriers demand meticulous scrutiny and continuous refinement to ensure the safety of these innovative drug delivery systems. Additionally, the heterogeneity of neurological disorders further complicates drug design and requires a detailed understanding of the unique challenges associated by each condition. Future directions in the field should focus on developing personalized and disease-specific nanomedicines, tailoring formulations to the distinctive characteristics of various neurological disorders.

Translating successful results of brain-targeted nanomedicine from preclinical settings to clinical applications is a challenging task, requiring the collaborative efforts of multidisciplinary teams to overcome the complexities of human physiology. Although some nanomaterials have received FDA approval or entered clinical trials, practical implementation of brain-targeted nano-based drug delivery systems in clinical settings remains very limited. This highlights the complex process of moving from laboratory success to widespread clinical use, requiring innovative strategies to optimize this translational process. The complex nature of the blood-brain barrier, interindividual variability in patient responses, and the need for precision in drug delivery mechanisms present significant obstacles. Ethical considerations, safety concerns, and the exorbitant cost of clinical trials are also contributing to a cautious approach to using brain-targeted nanomedicines on a larger clinical scale.

In addressing these challenges, the integration of advanced imaging techniques and the identification of reliable biomarkers is critical. These methodologies offer valuable information on the pharmacokinetics and biodistribution of nanomedicines in the human brain, providing the basis for informed and effective clinical strategies. Despite the complexity of these obstacles, ongoing research initiatives and collaborative efforts provide optimism that these challenges can be overcome and brain-targeted nanomedicines will be widely implemented in clinical practice in the foreseeable future.

In the years ahead, interdisciplinary collaboration, rigorous safety assessments, and a deepened understanding of the intricacies of neurological disorders will be essential to successfully realize the potential of nanotechnology in brain-targeted therapies. The evolving field of nanomedicine for brain disorders holds the promise of not only overcoming current challenges but also opening new frontiers in precision medicine, ultimately transforming the prognosis and treatment paradigms of a variety of brain-related diseases.

Acknowledgments

We are grateful to Dr. Olesja Bondarenko and Dr. Helen Forgham for fruitful discussions.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Funding

AK was supported by the National Health and Medical Research Council Emerging Leadership Fellowship (APP2009231, Australia), the European Union’s Horizon 2020 Research and Innovation Programme under Marie Skłodowska-Curie grant agreement no. 101027379. MS was supported by the Cure Parkinson’s Trust, UK. TT was funded by the Estonian Research Council (grants PRG230, PRG1788, and EAG79), and EuronanomedII projects ECM-CART and iNanoGun.

Disclosure

Mart Saarma (MS) is the founder and a shareholder in Herantis Pharma Plc. that develops PD drugs. MS is also a shareholder in Nanoform Plc that develops new nanotechnologies. Tambet Teesalu is an inventor on patents on CendR peptides and a shareholder of Lisata Therapeutics, a company that develops tumor-penetrating CendR peptides for cancer therapy. The authors report no other conflicts of interest in this work.

References

1. Ehrlich P. Das Sauerstoff-Bedürfniss des Organismus; Eine farbenanalytische Studie. Berlin: Verlag von August Hirschwald; 1885.

2. Gite VZ, Ghume VK, Kachave RN. Brain Targeted Drug Delivery System. World J Pharm Med Res. 2020;6(11):45–47.

3. Singh SB. Novel approaches for brain drug delivery system-review. Int J Pharm Res Rev. 2013;2(6):36–44.

4. Misra A, Ganesh S, Shahiwala A, Shah SP. Drug delivery to the central nervous system: a review. J Pharm Pharm Sci. 2003;6(2):252–273.

5. Pardridge WM. Blood–brain barrier delivery. Drug Discov Today. 2007;12(1–2):54–61.

6. Sweeney MD, Sagare AP, Zlokovic BV. Blood–brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat Rev Neurol. 2018;14(3):133–150.

7. Desai BS, Monahan AJ, Carvey PM, Hendey B. Blood-brain barrier pathology in Alzheimer’s and Parkinson’s disease: implications for drug therapy. Cell Transplant. 2007;16(3):285–299.

8. Yang C, Hawkins KE, Doré S, Candelario-Jalil E. Neuroinflammatory mechanisms of blood-brain barrier damage in ischemic stroke. Am J Physiol Cell Physiol. 2019;316(2):C135–C153.

9. Arvanitis CD, Ferraro GB, Jain RK. The blood–brain barrier and blood–tumour barrier in brain tumours and metastases. Nat Rev Cancer. 2020;20(1):26–41.

10. Ferrari A, Santomauro D, Herrera A, et al. Global, regional, and national burden of 12 mental disorders in 204 countries and territories, 1990-2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet Psychiatry. 2022;9(2):137–150.

11. Olesen J. The cost of neurological disorders in Europe. J Neurol Sci. 2015;357:e500–e501.

12. Gooch CL, Pracht E, Borenstein AR. The burden of neurological disease in the United States: a summary report and call to action. Ann Neurol. 2017;81(4):479–484.

13. Feigin VL, Nichols E, Alam T, et al. Global, regional, and national burden of neurological disorders, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019;18(5):459–480.

14. Feigin VL, Vos T, Nichols E, et al. The global burden of neurological disorders: translating evidence into policy. Lancet Neurol. 2020;19(3):255–265.

15. Nichols E, Steinmetz JD, Vollset SE, et al. Estimation of the global prevalence of dementia in 2019 and forecasted prevalence in 2050: an analysis for the Global Burden of Disease Study 2019. Lancet Public Health. 2022;7(2):e105–e125.

16. Pardridge WM. Drug targeting to the brain. Pharm Res. 2007;24(9):1733–1744.

17. Chen Y, Liu L. Modern methods for delivery of drugs across the blood-brain barrier. Adv Drug Deliv Rev. 2012;64(7):640–665.

18. Yi X, Manickam DS, Brynskikh A, Kabanov AV. Agile delivery of protein therapeutics to CNS. J Control Release. 2014;190:637–663.

19. Qizilbash FF, Ashhar MU, Zafar A, et al. Thymoquinone-enriched naringenin-loaded nanostructured lipid carrier for brain delivery via nasal route: in vitro prospect and in vivo therapeutic efficacy for the treatment of depression. Pharmaceutics. 2022;14(3):656.

20. Petkova AI, Kubajewska I, Vaideanu A, Schätzlein AG, Uchegbu IF. Gene Targeting to the Cerebral Cortex Following Intranasal Administration of Polyplexes. Pharmaceutics. 2022;14(6):1136.

21. Griggs DJ, Garcia AD, Au WY, et al. Improving the efficacy and accessibility of intracranial viral vector delivery in non-human primates. Pharmaceutics. 2022;14(7):1435.

22. Rechberger JS, Porath KA, Zhang L, et al. IL-13Rα2 status predicts GB-13 (IL13. E13K-PE4E) efficacy in high-grade glioma. Pharmaceutics. 2022;14(5):922.

23. Vert M, Doi Y, Hellwich K-H, et al. Terminology for biorelated polymers and applications (IUPAC Recommendations 2012). Pure Appl Chem. 2012;84(2):377–410.

24. Zhang CY, Dong X, Gao J, Lin W, Liu Z, Wang Z. Nanoparticle-induced neutrophil apoptosis increases survival in sepsis and alleviates neurological damage in stroke. Sci Adv. 2019;5(11):eaax7964.

25. Zhao Z, Ukidve A, Kim J, Mitragotri S. Targeting strategies for tissue-specific drug delivery. Cell. 2020;181(1):151–167.

26. Saeedi M, Eslamifar M, Khezri K, Dizaj SM. Applications of nanotechnology in drug delivery to the central nervous system. Biomed Pharmacother. 2019;111:666–675.

27. Ghorai SM, Deep A, Magoo D, Gupta C, Gupta N. Cell-Penetrating and Targeted Peptides Delivery Systems as Potential Pharmaceutical Carriers for Enhanced Delivery across the Blood–Brain Barrier (BBB). Pharmaceutics. 2023;15(7):1999.

28. Zhou X, Smith QR, Liu X. Brain penetrating peptides and peptide–drug conjugates to overcome the blood–brain barrier and target CNS diseases. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2021;13(4):e1695.

29. Bondarenko O, Saarma M. Neurotrophic Factors in Parkinson’s Disease: clinical Trials, Open Challenges and Nanoparticle-Mediated Delivery to the Brain. Front Cell Neurosci. 2021;15:682597.

30. 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.

31. de Lange EC. The mastermind approach to CNS drug therapy: translational prediction of human brain distribution, target site kinetics, and therapeutic effects. Fluids Barriers CNS. 2013;10(1):12.

32. Kalvass JC, Maurer TS. Influence of nonspecific brain and plasma binding on CNS exposure: implications for rational drug discovery. Biopharm Drug Dispos. 2002;23(8):327–338.

33. Rawal SU, Patel BM, Patel MM. New drug delivery systems developed for brain targeting. Drugs. 2022;82(7):749–792.

34. Wilhelm I, Nyúl-Tóth Á, Suciu M, Hermenean A, Krizbai IA. Heterogeneity of the blood-brain barrier. Tissue Barriers. 2016;4(1):e1143544.

35. Nakatsuji Y. What is the difference between the blood–nerve barrier and blood–brain barrier? Clin Exp Neuroimmunol. 2017;8(1):13–14.

36. Abbott NJ, Rönnbäck L, Hansson E. Astrocyte–endothelial interactions at the blood–brain barrier. Nat Rev Neurosci. 2006;7(1):41–53.

37. Sasson E, Anzi S, Bell B, et al. Nano-scale architecture of blood-brain barrier tight-junctions. Elife. 2021;10:e63253.

38. Allt G, Lawrenson JG. Pericytes: cell biology and pathology. Cells Tissues Organs. 2001;169(1):1–11.

39. Díaz-Flores L, Gutiérrez R, Madrid JF, et al. Pericytes. Morphofunction, interactions and pathology in a quiescent and activated mesenchymal cell niche. Histol Histopathol. 2009;24(7):909–969.

40. Fujimoto K. Pericyte-endothelial gap junctions in developing rat cerebral capillaries: a fine structural study. Anat Rec. 1995;242(4):562–565.

41. Ransohoff RM, Kivisäkk P, Kidd G. Three or more routes for leukocyte migration into the central nervous system. Nat Rev Immunol. 2003;3(7):569–581.

42. Kubotera H, Ikeshima-Kataoka H, Hatashita Y, Allegra Mascaro AL, Pavone FS, Inoue T. Astrocytic endfeet re-cover blood vessels after removal by laser ablation. Sci Rep. 2019;9(1):1263.

43. Schlageter KE, Molnar P, Lapin GD, Groothuis DR. Microvessel Organization and Structure in Experimental Brain Tumors: microvessel Populations with Distinctive Structural and Functional Properties. Microvasc Res. 1999;58(3):312–328.

44. Reichenbach A. Neuroglia-das andere zelluläre Element im Nervensystem: Die Müllersche Gliazelle. SMV, Edition Materia Medica; 1999.

45. Winkler EA, Sengillo JD, Bell RD, Wang J, Zlokovic BV. Blood–Spinal Cord Barrier Pericyte Reductions Contribute to Increased Capillary Permeability. J Cereb Blood Flow Metab. 2012;32(10):1841–1852.

46. Batiuk MY, Martirosyan A, Wahis J, et al. Identification of region-specific astrocyte subtypes at single cell resolution. Nat Commun. 2020;11(1):1220.

47. Macdonald JA, Murugesan N, Pachter JS. Endothelial cell heterogeneity of blood-brain barrier gene expression along the cerebral microvasculature. J Neurosci Res. 2010;88(7):1457–1474.

48. Lawrenson JG, Reid AR, Finn TM, Orte C, Allt G. Cerebral and pial microvessels: differential expression of gamma-glutamyl transpeptidase and alkaline phosphatase. Anat Embryol. 1999;199(1):29–34.

49. Pardridge WM. The blood-brain barrier: bottleneck in brain drug development. NeuroRX. 2005;2(1):3–14.

50. Zlokovic BV. The blood-brain barrier in health and chronic neurodegenerative disorders. Neuron. 2008;57(2):178–201.

51. Pardridge WM. Re-engineering therapeutic antibodies for Alzheimer’s disease as blood-brain barrier penetrating bi-specific antibodies. Expert Opin Biol Ther. 2016;16(12):1455–1468.

52. Lin C-Y, Hsieh H-Y, Chen C-M, et al. Non-invasive, neuron-specific gene therapy by focused ultrasound-induced blood-brain barrier opening in Parkinson’s disease mouse model. J Control Release. 2016;235:72–81.

53. Lin C-Y, Lin Y-C, Huang C-Y, S-R W, Chen C-M, Liu H-L. Ultrasound-responsive neurotrophic factor-loaded microbubble-liposome complex: preclinical investigation for Parkinson’s disease treatment. J Control Release. 2020;321:519–528.

54. Papisov MI, Belov VV, Gannon KS. Physiology of the intrathecal bolus: the leptomeningeal route for macromolecule and particle delivery to CNS. Mol Pharm. 2013;10(5):1522–1532.

55. Bennewitz MF, Saltzman WM. Nanotechnology for delivery of drugs to the brain for epilepsy. Neurotherapeutics. 2009;6(2):323–336.

56. Kozlovskaya L, Abou-Kaoud M, Stepensky D. Quantitative analysis of drug delivery to the brain via nasal route. J Control Release. 2014;189:133–140.

57. Lee H, Pienaar IS. Disruption of the blood-brain barrier in Parkinson’s disease: curse or route to a cure? FBL. 2014;19(2):272–280.

58. Kreuter J. Nanoparticulate systems for brain delivery of drugs. Adv Drug Deliv Rev. 2001;47(1):65–81.

59. Masserini M. Nanoparticles for brain drug delivery. ISRN Biochem. 2013;2013:18.

60. Parton RG, Howes MT. Revisiting caveolin trafficking: the end of the caveosome. J Cell Biol. 2010;191(3):439–441.

61. Nance EA, Woodworth GF, Sailor KA, et al. A dense poly (ethylene glycol) coating improves penetration of large polymeric nanoparticles within brain tissue. Sci Transl Med. 2012;4(149):149ra119.

62. Kreuter J, Shamenkov D, Petrov V, et al. Apolipoprotein-mediated transport of nanoparticle-bound drugs across the blood-brain barrier. J Drug Target. 2002;10(4):317–325.

63. Kreuter J, Petrov VE, Kharkevich DA, Alyautdin RN. Influence of the type of surfactant on the analgesic effects induced by the peptide dalargin after its delivery across the blood–brain barrier using surfactant-coated nanoparticles. J Control Release. 1997;49(1):81–87.

64. Koffie RM, Farrar CT, Saidi L-J, William CM, Hyman BT, Spires-Jones TL. Nanoparticles enhance brain delivery of blood–brain barrier-impermeable probes for in vivo optical and magnetic resonance imaging. Proc Natl Acad Sci U S A. 2011;108(46):18837–18842.

65. Niewoehner J, Bohrmann B, Collin L, et al. Increased brain penetration and potency of a therapeutic antibody using a monovalent molecular shuttle. Neuron. 2014;81(1):49–60.

66. Savier E, Tuffery P, Simon-Gracia L, et al. Bi-functional Peptides as a New Therapeutic Tool for Hepatocellular Carcinoma. HPB. 2023;25:S218.

67. Simón-Gracia L, Scodeller P, Fisher WS, et al. Paclitaxel-Loaded Cationic Fluid Lipid Nanodiscs and Liposomes with Brush-Conformation PEG Chains Penetrate Breast Tumors and Trigger Caspase-3 Activation. ACS Appl Mater Interfaces. 2022;14(51):56613–56622.

68. Sugahara KN, Teesalu T, Karmali PP, et al. Tissue-Penetrating Delivery of Compounds and Nanoparticles into Tumors. Cancer Cell. 2009;16(6):510–520.

69. Josephson L, Tung C-H, Moore A, Weissleder R. High-efficiency intracellular magnetic labeling with novel superparamagnetic-Tat peptide conjugates. Bioconjug Chem. 1999;10(2):186–191.

70. Blasi P, Giovagnoli S, Schoubben A, Ricci M, Rossi C. Solid lipid nanoparticles for targeted brain drug delivery. Adv Drug Deliv Rev. 2007;59(6):454–477.

71. Erazo-Oliveras A, Muthukrishnan N, Baker R, Wang T-Y, Pellois J-P. Improving the endosomal escape of cell-penetrating peptides and their cargos: strategies and challenges. Pharmaceuticals (Basel). 2012;5(11):1177–1209.

72. Smith SA, Selby LI, Johnston APR, Such GK. The endosomal escape of nanoparticles: toward more efficient cellular delivery. Bioconjug Chem. 2019;30(2):263–272.

73. Varkouhi AK, Scholte M, Storm G, Haisma HJ. Endosomal escape pathways for delivery of biologicals. J Control Release. 2011;151(3):220–228.

74. Martens TF, Remaut K, Demeester J, De Smedt SC, Braeckmans K. Intracellular delivery of nanomaterials: how to catch endosomal escape in the act. Nano Today. 2014;9(3):344–364.

75. Dos Santos Rodrigues B, Lakkadwala S, Kanekiyo T, Singh J. Development and screening of brain-targeted lipid-based nanoparticles with enhanced cell penetration and gene delivery properties. J Nanomed. 2019;14:6497–6517.

76. Zhang H, Gerson T, Varney ML, Singh RK, Vinogradov SV. Multifunctional peptide-PEG intercalating conjugates: programmatic of gene delivery to the blood-brain barrier. Pharm Res. 2010;27(12):2528–2543.

77. Sun R, Liu M, Lu J, et al. Bacteria loaded with glucose polymer and photosensitive ICG silicon-nanoparticles for glioblastoma photothermal immunotherapy. Nat Commun. 2022;13(1):5127.

78. L-P W, Ahmadvand D, Su J, et al. Crossing the blood-brain-barrier with nanoligand drug carriers self-assembled from a phage display peptide. Nat Commun. 2019;10(1):4635.

79. Lee J-H, Zhang A, You SS, Lieber CM. Spontaneous internalization of cell penetrating peptide-modified nanowires into primary neurons. Nano Lett. 2016;16(2):1509–1513.

80. Kumar P, Wu H, McBride JL, et al. Transvascular delivery of small interfering RNA to the central nervous system. Nature. 2007;448(7149):39–43.

81. Wu C-H, Liu I-J, Lu R-M, Wu H-C. Advancement and applications of peptide phage display technology in biomedical science. J Biomed Sci. 2016;23:8.

82. Oller‐Salvia B, Sánchez‐Navarro M, Ciudad S, et al. MiniAp-4: a venom-inspired peptidomimetic for brain delivery. Angew Chem Int Ed Engl. 2016;55(2):572–575.

83. Lam FC, Morton SW, Wyckoff J, et al. Enhanced efficacy of combined temozolomide and bromodomain inhibitor therapy for gliomas using targeted nanoparticles. Nat Commun. 2018;9(1):1991.

84. Gandhi M, Bhatt P, Chauhan G, Gupta S, Misra A, Mashru R. IGF-II-Conjugated Nanocarrier for Brain-Targeted Delivery of p11 Gene for Depression. AAPS Pharm Sci Tech. 2019;20(2):50.

85. Song J, Lu C, Leszek J, Zhang J. Design and development of nanomaterial-based drug carriers to overcome the blood–brain barrier by using different transport mechanisms. Int J Mol Sci. 2021;22(18):10118.

86. Spuch C, Saida O, Navarro C. Advances in the treatment of neurodegenerative disorders employing nanoparticles. Recent Pat Drug Deliv Formul. 2012;6(1):2–18.

87. Vieira DB, Gamarra LF. Multifunctional Nanoparticles for Successful Targeted Drug Delivery across the Blood-Brain Barrier. In: Parikesit AA, editor. Mol Insight Drug Des. Rijeka: IntechOpen; 2018.

88. Kwon HS, Koh S-H. Neuroinflammation in neurodegenerative disorders: the roles of microglia and astrocytes. Transl Neurodegener. 2020;9(1):42.

89. Li J, W O, Li W, Jiang Z-G, Ghanbari HA. Oxidative Stress and Neurodegenerative Disorders. Int J Mol Sci. 2013;14(12):24438–24475.

90. Lindholm P, Saarma M. Cerebral dopamine neurotrophic factor protects and repairs dopamine neurons by novel mechanism. Mol Psychiatry. 2022;27(3):1310–1321.

91. Naj AC, Schellenberg GD. Genomic variants, genes, and pathways of Alzheimer’s disease: an overview. Am J Med Genet B Neuropsychiatr Genet. 2017;174(1):5–26.

92. Johns P. Chapter 12 - Dementia. In: Johns P, editor. Clinical Neuroscience. Churchill Livingstone; 2014:145–162.

93. Abeliovich A, Gitler AD. Defects in trafficking bridge Parkinson’s disease pathology and genetics. Nature. 2016;539(7628):207–216.

94. Stefanis L. α-Synuclein in Parkinson’s disease. Cold Spring Harb Perspect Med. 2012;2(2):a009399.

95. Selkoe DJ, Hardy J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol Med. 2016;8(6):595–608.

96. Meade RM, Fairlie DP, Mason JM. Alpha-synuclein structure and Parkinson’s disease – lessons and emerging principles. Mol Neurodegeneration. 2019;14(1):29.

97. Sidorova YA, Saarma M. Can Growth Factors Cure Parkinson’s Disease? Trends Pharmacol Sci. 2020;41(12):909–922.

98. Schneider L. A resurrection of aducanumab for Alzheimer’s disease. Lancet Neurol. 2020;19(2):111–112.

99. van Dyck CH, Swanson CJ, Aisen P, et al. Lecanemab in Early Alzheimer’s Disease. N Engl J Med. 2022;388(1):9–21.

100. Couzin-Frankel J. Promising Alzheimer’s therapy and related drugs shrink brains. Science. 2023;380(19).

101. Andrikopoulos N, Tang H, Wang Y, et al. Exploring Peptido-Nanocomposites in the Context of Amyloid Diseases. Angew Chem. 2023.

102. Gu J, Yan C, Yin S, et al. Erythrocyte membrane-coated nanocarriers modified by TGN for Alzheimer’s disease. J Control Release. 2024;366:448–459.

103. Liu X-G, Zhang L, Lu S, et al. Superparamagnetic iron oxide nanoparticles conjugated with Aβ oligomer-specific scFv antibody and class A scavenger receptor activator show therapeutic potentials for Alzheimer’s Disease. J Nanobiotechnology. 2020;18(1):160.

104. Ruan Y, Xiong Y, Fang W, et al. Highly sensitive Curcumin-conjugated nanotheranostic platform for detecting amyloid-beta plaques by magnetic resonance imaging and reversing cognitive deficits of Alzheimer’s disease via NLRP3-inhibition. J Nanobiotechnol. 2022;20(1):322.

105. Liu R, Yang J, Liu L, et al. An “Amyloid-β Cleaner” for the Treatment of Alzheimer’s Disease by Normalizing Microglial Dysfunction. Adv Sci (Weinh). 2019;7(2):1901555.

106. Lei T, Yang Z, Jiang C, et al. Mannose-Integrated Nanoparticle Hitchhike Glucose Transporter 1 Recycling to Overcome Various Barriers of Oral Delivery for Alzheimer’s Disease Therapy. ACS Nano. 2024;18(4):3234–3250.

107. Jia T, Wang H, Chi W, et al. Knockdown of BACE1 by a multistage brain-targeting polyion complex improved memory and learning behaviors in APP/PS1 transgenic mouse model. Int J Pharm. 2024;650:123727.

108. Zhang L, Hou S, Movahedi F, et al. Amyloid-β/Tau burden and neuroinflammation dual-targeted nanomedicines synergistically restore memory and recognition of Alzheimer’s disease mice. Nano Today. 2023;49:101788.

109. Rodrigues BDS, Kanekiyo T, Singh J. Nerve growth factor gene delivery across the blood–brain barrier to reduce beta amyloid accumulation in AD mice. Mol Pharm. 2020;17(6):2054–2063.

110. Lei L, Zou Z, Liu J, et al. Multifunctional peptide-assembled micelles for simultaneously reducing amyloid-β and reactive oxygen species. Chem Sci. 2021;12(18):6449–6457.

111. Aloe L, Rocco ML, Bianchi P, Manni L. Nerve growth factor: from the early discoveries to the potential clinical use. J Transl Med. 2012;10(1):239.

112. Calissano P, Matrone C, Amadoro G. Nerve growth factor as a paradigm of neurotrophins related to Alzheimer’s disease. Dev Neurobiol. 2010;70(5):372–383.

113. Andrade S, Pereira MC, Loureiro JA. Caffeic acid loaded into engineered lipid nanoparticles for Alzheimer’s disease therapy. Colloids Surf B Biointerfaces. 2023;225:113270.

114. Arora S, Layek B, Singh J. Design and validation of liposomal ApoE2 gene delivery system to evade blood–brain barrier for effective treatment of Alzheimer’s disease. Mol Pharm. 2021;18(2):714–725.

115. Yu Y, S-y H, Kong L, et al. Brain-targeted multifunctional micelles delivering Oridonin and Phillyrin for synergistic therapy of Alzheimer’s disease. J Drug Delivery Sci Technol. 2023;87:104794.

116. Xie J, Gonzalez-Carter D, Tockary TA, et al. Dual-sensitive nanomicelles enhancing systemic delivery of therapeutically active antibodies specifically into the brain. ACS Nano. 2020;14(6):6729–6742.

117. Zhou Y, Zhu F, Liu Y, et al. Blood-brain barrier–penetrating siRNA nanomedicine for Alzheimer’s disease therapy. Sci Adv. 2020;6(41).

118. Kuo Y-C, Chen I-Y, Rajesh R. Astragaloside IV-and nesfatin-1-encapsulated phosphatidylserine liposomes conjugated with wheat germ agglutinin and leptin to activate anti-apoptotic pathway and block phosphorylated tau protein expression for Parkinson’s disease treatment. Mater Sci Eng C Mater Biol Appl. 2021;129:112361.

119. Gao Y, Cheng Y, Chen J, et al. NIR‐assisted MgO‐based polydopamine nanoparticles for targeted treatment of Parkinson’s disease through the blood–brain barrier. Adv Healthc Mater. 2022;11(23):e2201655.

120. Sela M, Poley M, Mora-Raimundo P, et al. Brain-Targeted Liposomes Loaded with Monoclonal Antibodies Reduce Alpha-Synuclein Aggregation and Improve Behavioral Symptoms in Parkinson’s Disease. Adv. Mater. 2023;35(51):2304654.

121. Kuo Y-C, Feng K-C, Rajesh R. Rabies virus glycoprotein- and transferrin-functionalized liposomes to elevate epigallocatechin gallate and FK506 activity and mediate MAPK against neuronal apoptosis in Parkinson’s disease. J Taiwan Inst Chem Eng. 2022;132:104142.

122. Dimiou S, Lopes RM, Kubajewska I, et al. Particulate levodopa nose-to-brain delivery targets dopamine to the brain with no plasma exposure. Int J Pharm. 2022;618:121658.

123. Gonzalez-Carter DA, Ong ZY, McGilvery CM, Dunlop IE, Dexter DT, Porter AE. L-DOPA functionalized, multi-branched gold nanoparticles as brain-targeted nano-vehicles. Nanomedicine. 2019;15(1):1–11.

124. Huang S, Y-J L, J-Y W, et al. Biomimetic nanodecoys deliver cholesterol-modified heteroduplex oligonucleotide to target dopaminergic neurons for the treatment of Parkinson’s disease. Acta Biomater. 2024.

125. Mead BP, Kim N, Miller GW, et al. Novel focused ultrasound gene therapy approach noninvasively restores dopaminergic neuron function in a rat Parkinson’s disease model. Nano Lett. 2017;17(6):3533–3542.

126. Xing Y, Wen C-Y, Li S-T, Xia Z-X. Non-viral liposome-mediated transfer of brain-derived neurotrophic factor across the blood-brain barrier. Neural Regen Res. 2016;11(4):617–622.

127. Airaksinen M, Saarma M. The GDNF family: signalling, biological functions and therapeutic value. Nat Rev Neurosci. 2002;3(5):383–394.

128. Huttunen HJ, Saarma M. CDNF Protein Therapy in Parkinson’s Disease. Cell Transplantation. 2019;28(4):349–366.

129. Tan AC, Ashley DM, López GY, Malinzak M, Friedman HS, Khasraw M. Management of glioblastoma: state of the art and future directions. CA Cancer J Clin. 2020;70(4):299–312.

130. Sarkaria JN, Hu LS, Parney IF, et al. Is the blood-brain barrier really disrupted in all glioblastomas? A critical assessment of existing clinical data. Neuro Oncol. 2018;20(2):184–191.

131. Farshbaf M, Mojarad-Jabali S, Hemmati S, et al. Enhanced BBB and BBTB penetration and improved anti-glioma behavior of Bortezomib through dual-targeting nanostructured lipid carriers. J Control Release. 2022;345:371–384.

132. Zhao Y, Peng Y, Yang Z, et al. pH-redox responsive cascade-targeted liposomes to intelligently deliver doxorubicin prodrugs and lonidamine for glioma. Eur. J. Med. Chem. 2022;235:114281.

133. Wen L, Peng Y, Wang K, et al. Regulation of pathological BBB restoration via nanostructured ROS-responsive glycolipid-like copolymer entrapping siVEGF for glioblastoma targeted therapeutics. Nano Res. 2022;15:1455–1465.

134. Liu Y, Zou Y, Feng C, et al. Charge conversional biomimetic nanocomplexes as a multifunctional platform for boosting orthotopic glioblastoma RNAi therapy. Nano Lett. 2020;20(3):1637–1646.

135. Zheng M, Liu Y, Wang Y, et al. ROS‐responsive polymeric siRNA nanomedicine stabilized by triple interactions for the robust glioblastoma combinational RNAi therapy. Adv Mater. 2019;31(37):e1903277.

136. Heggannavar GB, Vijeth S, Kariduraganavar MY. Development of dual drug loaded PLGA based mesoporous silica nanoparticles and their conjugation with Angiopep-2 to treat glioma. J Drug Deliv Sci Technol. 2019;53:101157.

137. Tao J, Fei W, Tang H, et al. Angiopep-2-conjugated “core–shell” hybrid nanovehicles for targeted and pH-triggered delivery of arsenic trioxide into glioma. Mol Pharm. 2019;16(2):786–797.

138. Ye C, Pan B, Xu H, et al. Co-delivery of GOLPH3 siRNA and gefitinib by cationic lipid-PLGA nanoparticles improves EGFR-targeted therapy for glioma. J Mol Med. 2019;97(11):1575–1588.

139. Wen L, Wen C, Zhang F, Wang K, Yuan H, Hu F. siRNA and chemotherapeutic molecules entrapped into a redox-responsive platform for targeted synergistic combination therapy of glioma. Nanomedicine. 2020;28:102218.

140. Martins C, Araújo M, Malfanti A, et al. Stimuli-Responsive Multifunctional Nanomedicine for Enhanced Glioblastoma Chemotherapy Augments Multistage Blood-to-Brain Trafficking and Tumor Targeting. Small. 2023;19(22):2300029.

141. He W, Li X, Morsch M, et al. Brain-Targeted Codelivery of Bcl-2/Bcl-xl and Mcl-1 Inhibitors by Biomimetic Nanoparticles for Orthotopic Glioblastoma Therapy. ACS Nano. 2022;16(4):6293–6308.

142. Thirumurugan S, Dash P, Liu X, et al. Angiopep-2-conjugated FeTi@Au core-shell nanoparticles for tumor targeted dual-mode magnetic resonance imaging and hyperthermic glioma therapy. Nanomed Nanotechnol Biol Med. 2023;50:102673.

143. Erel-Akbaba G, Carvalho LA, Tian T, et al. Radiation-induced targeted nanoparticle-based gene delivery for brain tumor therapy. ACS nano. 2019;13(4):4028–4040.

144. Ramalho MJ, Bravo M, Loureiro JA, Lima J, Pereira MC. Transferrin-modified nanoparticles for targeted delivery of Asiatic acid to glioblastoma cells. Life Sci. 2022;296:120435.

145. Kim -S-S, Rait A, Kim E, Pirollo KF, Chang EH. A tumor-targeting p53 nanodelivery system limits chemoresistance to temozolomide prolonging survival in a mouse model of glioblastoma multiforme. Nanomedicine. 2015;11(2):301–311.

146. Binabaj MM, Bahrami A, ShahidSales S, et al. The prognostic value of MGMT promoter methylation in glioblastoma: a meta-analysis of clinical trials. J Cell Physiol. 2018;233(1):378–386.

147. Zhong X, Wei G, Liu B, et al. Polyhedral Oligomeric Silsesquioxane-Based Nanoparticles for Efficient Chemotherapy of Glioblastoma. Small. 2023;19(18):e2207248.

148. Zhang H, Shi C, Han F, et al. Precise gliomas therapy: hypoxia-activated prodrugs sensitized by nano-photosensitizers. Biomaterials. 2022;289:121770.

149. Wu Y, Hu D, Gao D, Liu C, Zheng H, Sheng Z. Miniature NIR-II Nanoprobes for Active-Targeted Phototheranostics of Brain Tumors. Adv Healthc Mater. 2022;11(23):e2202379.

150. Dong CY, Hong S, Zheng DW, et al. Multifunctionalized Gold Sub-Nanometer Particles for Sensitizing Radiotherapy against Glioblastoma. Small. 2021;17(5):e2006582.

151. Säälik P, Lingasamy P, Toome K, et al. Peptide-guided nanoparticles for glioblastoma targeting. J Control Release. 2019;308:109–118.

152. Shadmani N, Makvandi P, Parsa M, et al. Enhancing Methotrexate Delivery in the Brain by Mesoporous Silica Nanoparticles Functionalized with Cell-Penetrating Peptide using in Vivo and ex Vivo Monitoring. Mol Pharmaceut. 2023;20(3):1531–1548.

153. Lingasamy P, Tobi A, Haugas M, et al. Bi-specific tenascin-C and fibronectin targeted peptide for solid tumor delivery. Biomaterials. 2019;219:119373.

154. Li Y, Xie M, Jones JB, et al. Targeted Delivery of DNA Topoisomerase Inhibitor SN38 to Intracranial Tumors of Glioblastoma Using Sub-5 Ultrafine Iron Oxide Nanoparticles. Adv Healthcare Mater. 2022;11(14):2102816.

155. Hussain S, Rodriguez-Fernandez M, Braun GB, Doyle FJ, Ruoslahti E. Quantity and accessibility for specific targeting of receptors in tumours. Sci Rep. 2014;4:5232.

156. Kang S, Lee S, Park S. iRGD Peptide as a Tumor-Penetrating Enhancer for Tumor-Targeted Drug Delivery. Polymers. 2020;12(9):1906.

157. Chen S, Zeng L, Hu Z. Progressing haemorrhagic stroke: categories, causes, mechanisms and managements. J Neurol. 2014;261(11):2061–2078.

158. Lo EH, Dalkara T, Moskowitz MA. Mechanisms, challenges and opportunities in stroke. Nat Rev Neurosci. 2003;4(5):399–415.

159. Malik R, Dichgans M. Challenges and opportunities in stroke genetics. Cardiovasc Res. 2018;114(9):1226–1240.

160. Prakash R, Carmichael ST. Blood-brain barrier breakdown and neovascularization processes after stroke and traumatic brain injury. Curr Opin Neurol. 2015;28(6):556–564.

161. Venketasubramanian N, Yoon BW, Pandian J, Navarro JC. Stroke Epidemiology in South, East, and South-East Asia: a Review. J Stroke. 2017;19(3):286–294.

162. Shcharbina N, Shcharbin D, Bryszewska M. Nanomaterials in stroke treatment: perspectives. Stroke. 2013;44(8):2351–2355.

163. Wang Z, Pan J, Yuan R, Chen M, Guo X, Zhou S. Shell-Sheddable Polymeric Micelles Alleviate Oxidative Stress and Inflammation for Enhanced Ischemic Stroke Therapy. Nano Lett. 2023;23(14):6544–6552.

164. Lv W, Xu J, Wang X, Li X, Xu Q, Xin H. Bioengineered boronic ester modified dextran polymer nanoparticles as reactive oxygen species responsive nanocarrier for ischemic stroke treatment. ACS nano. 2018;12(6):5417–5426.

165. Bao Q, Hu P, Xu Y, et al. Simultaneous blood–brain barrier crossing and protection for stroke treatment based on edaravone-loaded ceria nanoparticles. ACS Nano. 2018;12(7):6794–6805.

Creative Commons License © 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.