Next Article in Journal
Revolutionizing Diabetes Management Through Nanotechnology-Driven Smart Systems
Previous Article in Journal
mPEG-PCL Nanoparticles to Improve Oral Bioavailability of Acalabrutinib: Effect of Polymer Lipophilicity and Hydrophilicity on Physicochemical Properties and In Vivo Performance in Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Intranasal Drug Delivery Technology in the Treatment of Central Nervous System Diseases: Challenges, Advances, and Future Research Directions

School of Medicine, Huaqiao University, Quanzhou 362021, China
*
Author to whom correspondence should be addressed.
Pharmaceutics 2025, 17(6), 775; https://doi.org/10.3390/pharmaceutics17060775 (registering DOI)
Submission received: 17 April 2025 / Revised: 3 June 2025 / Accepted: 10 June 2025 / Published: 13 June 2025

Abstract

:
As population aging becomes an increasingly critical global issue, the incidence of central nervous system (CNS) diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), and stroke, has risen sharply. However, the blood–brain barrier (BBB) presents a significant obstacle to the effective treatment of these CNS disorders, limiting the ability of therapeutic agents to reach the brain. In this context, intranasal drug delivery, which bypasses the BBB, has attracted considerable attention in recent years. By utilizing pathways such as the olfactory and trigeminal nerves, intranasal drug delivery facilitates the rapid transport of drugs to the brain, thereby enhancing both the bioavailability and targeting efficiency of the drugs. This review provides an overview of the molecular mechanisms underlying intranasal drug delivery, its advancements in the treatment of CNS diseases, strategies to improve delivery efficiency, and a discussion of the challenges and potential future directions in this field. The aim of this paper is to offer valuable insights and guidance for researchers and clinicians working in the area of CNS disease treatment.

1. Introduction

Crossing the blood–brain barrier (BBB) has long been a significant challenge in the development of novel therapeutics for central nervous system (CNS) diseases [1]. Traditional CNS drug delivery approaches, including oral and intravenous administration, are hindered by several critical limitations. The BBB severely restricts drug penetration into the central nervous system, while metabolic degradation and first-pass hepatic effects reduce systemic bioavailability. Non-specific distribution often leads to off-target toxicity. Additionally, poor CNS permeability, short half-life, narrow therapeutic windows, and frequent dosing requirements further limit clinical efficacy and patient adherence. These challenges underscore the necessity for more efficient and targeted CNS drug delivery systems. Unlike traditional oral or intravenous routes, intranasal drug delivery facilitates drug absorption through the nasal mucosa, allowing direct access to systemic circulation and avoiding the first-pass metabolism in the liver and gastrointestinal tract, and offers a promising solution for bypassing the BBB. This enhances the bioavailability of drugs [2,3].
Consequently, identifying strategies to bypass the BBB has become a key area of scientific inquiry. Intranasal drug delivery represents a promising, non-invasive approach that circumvents the BBB, enabling direct drug delivery to the brain [4,5,6]. The nasal mucosa is rich in blood vessels and microvilli, significantly increasing the surface area for drug absorption and providing an efficient pathway for drugs to enter the bloodstream [7,8,9]. Furthermore, the olfactory epithelium, connected to various brain regions via the olfactory and trigeminal nerves, serves as a crucial route for drug transport to the brain. This connection not only enables targeted delivery to specific brain regions but also allows for precision treatment of various neurological diseases [4,10,11]. The non-invasive nature of intranasal drug delivery alleviates the drawbacks associated with invasive procedures such as injections or surgery, thus improving patient compliance—especially in the case of chronic diseases like neurodegenerative disorders, which require long-term treatment. Additionally, intranasal delivery bypasses the first-pass metabolism in the liver, thereby reducing systemic drug exposure and decreasing the potential for systemic side effects [7,12,13]. This makes intranasal drug delivery particularly advantageous for drugs with low bioavailability or poor stability [14]. In recent years, this method has gained considerable attention in the treatment of CNS disorders and beyond [15,16,17]. Numerous studies have now progressed to clinical trials, rigorously evaluating safety, efficacy, and dosing to bridge preclinical findings with real-world therapeutic applications. For example, in a phase 2/3 clinical trial, intranasal administration of zavegepant for the treatment of migraine has achieved positive results, significantly improving patients’ pain freedom [18]. In addition, the FDA has approved the use of intranasal administration of Foralumab for the first patient with moderate Alzheimer’s disease. Meanwhile, nalmefene nasal spray has been approved for the emergency treatment of known or suspected opioid overdose [19,20]. Another clinical study has explored the esketamine nasal spray combined with a selective serotonin reuptake inhibitor or a serotonin-norepinephrine reuptake inhibitor significantly improved remission and reduced relapse in treatment-resistant depression compared to quetiapine augmentation [21].
This paper provides a comprehensive review of the molecular mechanisms underlying intranasal drug delivery, its advancements in treating CNS diseases, and strategies to enhance its delivery efficiency. It also examines the challenges faced by intranasal delivery, including issues related to unstable drug absorption, local irritation, and the development of drug formulations. Finally, the paper discusses the future directions of intranasal drug delivery systems and proposes strategies for improving drug permeability, stability, and personalized treatment. This review synthesizes the latest research findings and technological innovations in the field, offering valuable insights for researchers and clinicians.

2. Molecular Mechanisms of Intranasal Drug Delivery

Intranasal drug delivery primarily relies on the olfactory and trigeminal nerves, utilizing both intracellular and extracellular transport pathways to enable direct access to the CNS [22]. Drugs administered via the olfactory route cross the cribriform plate to reach the olfactory bulb and other brain regions, while those delivered through the trigeminal nerve are transported to the brainstem via the anterior lacerated foramen [23]. Intracellular transport involves endocytosis by olfactory sensory neurons followed by axonal transport, whereas extracellular transport enables paracellular diffusion across the nasal epithelium into the perineural space, eventually reaching the subarachnoid space via cerebrospinal fluid (CSF) flow. The CSF, driven by arterial pulsations, distributes the drug throughout the brain via the glymphatic system, facilitating brain-wide diffusion [24,25,26] (Figure 1).

2.1. Olfactory Nerve Pathway

The olfactory nerve pathway is the most extensively studied mechanism for intranasal drug delivery to the brain. Drugs absorbed by the olfactory epithelium are transported along olfactory nerve fibers, bypassing the BBB and directly reaching the cerebral cortex, hippocampus, and other brain regions. This provides a crucial foundation for the treatment of CNS diseases such as stroke, epilepsy, and traumatic brain injury [8,9]. The olfactory pathway supports the transport of not only small molecules but also peptides, proteins, and macromolecules that are otherwise restricted by the BBB, achieving rapid absorption and high brain concentrations [4,27] (Figure 2).
Upon entering the olfactory mucosa, drugs follow two primary routes to reach the CNS: (1) Intracellular transport, wherein drugs undergo clathrin-mediated endocytosis by olfactory receptor neurons, followed by microtubule-based axonal transport to the olfactory bulb, where they are released into the cytoplasm. This process is relatively slow, typically requiring hours to days. (2) Extracellular transport, which is faster and involves passive transcellular or paracellular diffusion through epithelial tight junctions or membranes into the extracellular space of olfactory nerve bundles. From there, drugs are carried by bulk flow to the olfactory bulb and diffuse into the CSF, enabling widespread brain distribution [22,23,28]. Nanoparticles exhibit enhanced diffusion along this route due to their affinity for the lipid-rich environment, while hydrophilic compounds may require protein carriers. Nanoparticles with sizes ranging from 50 to 200 nm, particularly those under 150 nm, are most suitable for nose-to-brain delivery. Liposomes with a particle size below 150 nm have demonstrated enhanced brain deposition and prolonged release (up to 96 h), significantly improving drug bioavailability compared to conventional formulations [29,30]. Nanoemulsions typically sized between 20 and 200 nm (e.g., 106.8 nm) have shown a 1.68-fold increase in brain permeation efficiency [31]. Solid lipid nanoparticles, often within the 100–300 nm range, with optimized sizes around 129 nm, allow for sustained drug release and higher brain targeting [32]. Similarly, nanostructured lipid carriers around 190.98 nm have achieved superior brain delivery efficacy, with intranasal administration yielding higher AUC values than intravenous routes. Overall, particles within this nanometric range ensure effective mucosal adhesion, enhanced permeability through olfactory and trigeminal pathways, and minimized systemic side effects, making them ideal for central nervous system targeting via nasal administration [33]. These unique anatomical and molecular features render the olfactory pathway a key route for intranasal CNS drug delivery.

2.2. Trigeminal Nerve Pathway

In addition to the olfactory route, the trigeminal nerve serves as a significant non-invasive conduit for direct drug transport to the brain. The trigeminal nerve regulates facial sensory function and projects to the brainstem and thalamic regions [34,35,36] (Figure 2).
Drugs administered through the nasal mucosa are absorbed by epithelial cells and interact with sensory neurons of the trigeminal nerve, facilitating their delivery to deeper CNS structures. This pathway may involve activation of transient receptor potential (TRP) channels, such as TRPV1, which sensitize nerve endings and mediate receptor-dependent endocytosis, followed by dynein-driven retrograde axonal transport to brainstem nuclei [37,38]. The maxillary and ophthalmic branches of the trigeminal nerve innervate the nasal respiratory mucosa and terminate in the pons and cribriform plate. Drug transport via the trigeminal pathway may occur through both intra-axonal transport and extracellular diffusion, operating independently of the olfactory route [24]. One major distinction lies in the target site: while the olfactory pathway delivers drugs to the olfactory bulb, the trigeminal route carries them to the pons, resulting in slower diffusion into the CSF due to the longer anatomical distance to CNS targets [39,40]. Although both pathways have been validated for brain delivery, the relative contribution of each remains to be elucidated. The trigeminal nerve, as the fifth cranial nerve composed of ophthalmic, maxillary, and mandibular branches converging at the trigeminal ganglion, originates from the pons and provides a viable structural basis for CNS-targeted delivery. Drugs absorbed via the ophthalmic and maxillary branches can reach the brainstem and ultimately the brain parenchyma. Similar to the olfactory route, the trigeminal nerve supports nose-to-brain transport through both intra-axonal and extracellular mechanisms, although intracellular transport along this pathway tends to be slower [28,41,42]. These anatomical and physiological differences underscore the importance of further studies to quantify the relative contributions of each pathway and optimize intranasal formulations accordingly.

2.3. Transduction Mechanisms of Paracellular and Transcellular Pathways in Intranasal Drug Delivery

Beyond the neural pathways, drugs may also traverse the neurovascular spaces surrounding the olfactory and trigeminal nerves. These interstitial fluid-filled compartments enable both paracellular and transcellular diffusion into the CNS, providing an additional mechanism to enhance intranasal drug delivery efficiency [43,44]. This supplementary route facilitates drug penetration into deeper brain structures, complementing neural transport [4,35,45,46] (Figure 2).
Paracellular transport allows passive drug diffusion between epithelial cells, entering the perineural space and subsequently the subarachnoid space via CSF. Within the CSF, arterial pulsations drive distribution through glymphatic (perivascular) pathways, functioning similarly to a brain-specific lymphatic system and ensuring widespread drug delivery [10,47]. Tight junction integrity, maintained by structural proteins such as claudin-4, claudin-5, occludin, and zonula occludens-1 (ZO-1), plays a critical role in regulating this transport route [35,48]. Notably, insulin and interferon-β have demonstrated effective CNS targeting via the paracellular route, diffusing across epithelial junctions into the CSF [49,50].
Transcellular transport encompasses multiple mechanisms, including clathrin-mediated endocytosis, caveolae-mediated endocytosis, macropinocytosis, carrier-mediated active transport, and efflux systems. Small molecules, such as peptides and nanoparticles, commonly enter cells via clathrin-coated vesicles involving clathrin heavy chain and adaptor protein complex 2. Lipophilic agents like paclitaxel in liposomes are predominantly absorbed via caveolae-mediated pathways, while macromolecular drugs, including exosomes and proteins, are mainly internalized through micropinocytosis [49,50].

3. Advances in Intranasal Drug Delivery Applications

Intranasal drug delivery demonstrates broad therapeutic potential, encompassing diverse agents such as chemical drugs, biomacromolecule drugs, and cell-derived drugs. As a non-invasive alternative to injections or oral delivery, it enhances patient compliance while enabling targeted therapeutic effects (Figure 3, Table 1).

3.1. Progress in Intranasal Delivery of Small Molecule Drugs

Small molecule drugs are widely used in clinical practice due to their small molecular weight and good oral absorption. Intranasal delivery of small molecule drugs has been widely explored, particularly in the treatment of CNS diseases. For example, intranasal delivery of galantamine, 9-cis retinoic acid, lacosamide, or isocyanomethane has shown effectiveness in treating AD and epilepsy [51,52,53,54]. Furthermore, intranasal delivery has also achieved significant results in pain management, sedation, and smoking cessation therapy, such as intranasal ketamine, dexmedetomidine, lidocaine, and chloral hydrate, which effectively alleviate postoperative pain and sedation [55,56,57,58,59]. Intranasal delivery offers advantages such as rapid onset, ease of use, and good patient tolerance, especially for advanced patients who have difficulty swallowing oral drugs [60]. In palliative care and community hospice treatment, intranasal delivery of morphine or midazolam can effectively treat pain and agitation, with simple administration and excellent suitability for patients with swallowing difficulties or fear of needles [61]. Additionally, intranasal drug delivery technology can be combined with nanotechnology or biomaterials to further enhance drug penetration and immune response, providing new delivery strategies for early intervention in neurodegenerative diseases such as AD [62,63,64] (Table 2). The above research provides a non-invasive, convenient, and efficient therapeutic approach for the intranasal delivery of small molecular chemical drugs.

3.2. Research Progress of Intranasal Delivery of Biomacromolecule Drugs

Biomacromolecule drugs (such as proteins, antibodies, vaccines, etc.) typically face challenges such as low bioavailability and difficulty penetrating biological barriers due to their large molecular size and complex structures [66]. These challenges often make the delivery of biomacromolecule drugs more complex than that of traditional small-molecule drugs. Therefore, the delivery of biomacromolecule drugs has been a key challenge limiting their clinical application. However, intranasal drug delivery offers significant advantages for the delivery of biologic drugs.

3.2.1. Progress in Intranasal Delivery of Growth Factors and Peptide Drugs

Intranasal drug delivery offers a key advantage by bypassing the gastrointestinal tract and first-pass metabolism, thereby significantly improving the delivery efficiency of growth factors and peptide drugs. However, its effectiveness is still limited by factors such as the molecular characteristics of the growth factor, intranasal absorption capacity, stability, and the local environment. For example, intranasal delivery of nerve growth factor (NGF) has shown good therapeutic potential in treating traumatic brain injury (TBI) and other neurodegenerative diseases such as AD and PD, providing new insights for the clinical application of NGF. However, there are also some side effect risks associated with intranasal delivery, so future research needs to focus on areas such as dose optimization, combination therapy, and improvements in delivery technology to further enhance the efficacy and local drug accessibility of the drugs [67,68,69,70]. Additionally, intranasal delivery of leukemia inhibitory factor (LIF) has provided a new approach for treating neurological function after mild TBI in children [71].
Intranasal delivery is also widely applied in the delivery of peptide drugs. Intranasal delivery of insulin can enter the brain through the olfactory or trigeminal nerves, improving cognitive function with good safety [72]. Using nanotechnology to further enhance insulin delivery efficiency can also improve its therapeutic effect [73,74,75]. Clinical trials and animal studies have shown that intranasal insulin delivery improves memory performance and has significant neuroprotective effects related to diabetes [76,77,78,79,80,81]. Another notable example is the MemAID trial, a Phase II randomized controlled study involving older adults with and without type 2 diabetes. Daily intranasal administration of 40 IU insulin over 24 weeks led to significant improvements in gait speed, prefrontal cerebral blood flow, and insulin sensitivity in diabetic patients, while non-diabetic participants showed enhanced executive function and verbal memory [82]. Furthermore, the intranasal route has provided new opportunities for delivering glucagon-like peptide-1 (GLP-1) to the brain for the treatment of obesity [83]. The novel oxytocin intranasal spray TTA-121 may achieve effective treatment with lower doses and fewer sprays, offering new hope for the treatment of autism spectrum disorder [84,85]. In addition, a randomized controlled trial investigated the efficacy of intranasal oxytocin as an adjunct to exposure therapy for social anxiety disorder (SAD). The study demonstrated that oxytocin administration improved positive evaluations of appearance and speech performance during exposure sessions, indicating its potential to enhance treatment outcomes for SAD [86]. Intranasal delivery of the bicyclic peptide OL-CTOP, which contains two disulfide bonds, can effectively antagonize the analgesic effect of morphine and prevent its respiratory suppression side effects, demonstrating its potential as a novel brain-targeting drug [87] (Table 3).

3.2.2. Research Progress in Intranasal Vaccine Delivery

Intranasal vaccines can deliver antigens directly to CNS-associated lymphoid tissues and neural structures. This allows for neuroimmune modulation, which is particularly valuable in treating or preventing neuroinflammatory and neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, and multiple sclerosis [88,89]. Previous studies have demonstrated that viruses like the hepatitis virus can enter the brain from the nasal cavity through the olfactory nerve, bypassing the BBB [90]. In addition, HHV-6 has been implicated in neurological conditions such as multiple sclerosis and encephalitis through nasal-brain transmission [91]. Experimental studies with adeno-associated virus (AAV) vectors have also shown that gene therapies can reach the brain through intranasal administration and exert therapeutic effects [92,93,94]. These findings underscore the nasal cavity as both a potential route for viral neuroinvasion and a promising pathway for non-invasive therapeutic delivery. Intranasal mucosal vaccines can effectively activate the mucosal immune system and overcome the limitations of traditional vaccines, addressing issues related to immunogenicity, stability, and safety that traditional vaccines face [95,96,97]. This is especially important for the prevention of respiratory diseases [15,87,96]. Additionally, intranasal delivery of subunit vaccines and nanomaterials can trigger both local and systemic immune responses, with breakthroughs particularly in penetrating the intranasal epithelial barrier [95,98,99]. Studies on vaccines targeting various pathogens such as the novel coronavirus (SARS-CoV-2), pneumococcus, and tuberculosis show that intranasal vaccines offer advantages of higher compliance, lower costs, and the ability to induce strong immune responses, making them especially suitable for large-scale immunization campaigns and more easily accepted by children [100,101,102]. Furthermore, intranasal vaccination is more effective than subcutaneous vaccination in alleviating clinical symptoms and pulmonary lesions, indicating that intranasal vaccination has advantages during epidemic outbreaks [103]. Intranasal vaccine delivery strategies provide new research directions for the prevention and control of respiratory viral infections [104,105,106].
Intranasal delivery of receptor-binding domain nanoparticles can effectively activate local immune responses and cellular immune responses. This provides a theoretical basis for the development of COVID-19 vaccines [107,108]. Intranasal immunization with nicotine vaccine candidates can induce both systemic and mucosal antibodies, which specifically neutralize nicotine, offering the potential for developing new smoking cessation therapies [109]. Additionally, adjuvants such as xanthan gum can enhance T cell responses and provide effective protection, while monoclonal antibody immune complexes can induce long-lasting immune responses [110,111]. Overall, these advancements offer new directions and technological support for the development of better intranasal vaccines.

3.2.3. Research Progress in Intranasal Delivery of Nucleic Acid Drugs

Current gene therapy applications in the brain are limited by existing delivery systems. Intranasal delivery of CRISPR-dCas9 system-based nanoparticles has effectively upregulated the expression of the target gene Sirt1, reducing brain edema and improving survival rates after permanent middle cerebral artery occlusion [112]. Intranasal delivery of self-assembled antagomir-21/RAP nanoparticles can enhance glioblastoma treatment effects without using potentially toxic carriers [113]. Intranasal delivery of apolipoprotein A-I nanoparticles carrying antisense oligonucleotide (ASO) can reduce mutated Huntington protein levels in a Huntington’s disease mouse model [114]. Hyaluronidase-coated glycerol chitosan-DNA complexes enhance gene targeting and distribution in the brain, providing a new non-invasive gene therapy approach for neurodegenerative diseases such as AD [115]. Intranasal delivery of nucleic acid drugs is a non-invasive, fast, cell-free, and targeted therapeutic method that can significantly promote functional recovery after complete spinal cord injury (SCI) [116] (Table 4). Preclinical studies have demonstrated the efficacy of this delivery method. For instance, intranasal administration of siRNA-loaded lipid nanoparticles has shown significant reductions in glioma growth in rodent models. Similarly, intranasal delivery of mRNA vaccines has led to antigen expression in olfactory bulb neurons, indicating successful CNS targeting [117,118,119]. Intranasal delivery of nucleic acids offers a promising non-invasive route to the CNS. It effectively bypasses the BBB. However, challenges such as susceptibility to nuclease degradation and limited membrane permeability necessitate the use of protective carriers to ensure stability and facilitate cellular uptake. Once administered intranasally, nucleic acid can access the brain via two primary neuronal pathways: the olfactory and trigeminal nerves. The olfactory pathway allows direct axonal transport from the nasal epithelium to the olfactory bulb, while the trigeminal pathway provides access to the brainstem. Additionally, astrocytes and microglia can internalize these carriers through endocytosis or receptor-mediated mechanisms, including interactions with scavenger receptors [43,44]. These findings underscore the potential of intranasal nucleic acid delivery systems in treating various CNS disorders.

3.3. Research Progress in Intranasal Delivery of Cell-Derived Therapeutic Drugs

3.3.1. Research Progress in Intranasal Delivery of Cell Therapy Drugs

In recent years, this approach has made significant progress in the delivery of biologic drugs, particularly cell-based therapies. This delivery route leverages the direct pathway between the intranasal cavity and the CNS, providing a new strategy for the treatment of CNS diseases. For example, intranasal delivery of human olfactory mucosal progenitor cells (OMPCs) or human neural stem cells (hNSCs) can specifically migrate to the vicinity of damaged neurons and axons, offering a non-invasive stem cell therapy for brain injury with potential clinical application value [120,121]. Intranasal delivery of hNSCs into the brain, where they differentiate into neurons, reduces β-amyloid plaque accumulation, decreases neuroinflammation, and improves cognitive function, providing a new avenue for the treatment of AD [122]. Additionally, intranasal delivery of bone marrow-derived mesenchymal stem cells (BMSCs) or deciduous dental pulp stem cells (DPSCs) significantly improves motor coordination and olfactory function in PD mice, reducing the degeneration of dopaminergic neurons and offering new methods for PD treatment [123,124]. Intranasal delivery of mesenchymal stromal cells (MSCs) can also treat neuronal damage caused by prion diseases [125,126], and intranasal delivery of bone marrow stromal cells (BMSCs) promotes neuronal regeneration and functional repair after stroke [127]. Furthermore, recent studies have shown that intranasal delivery of mitochondria has significant therapeutic effects on various neurological diseases, expanding the application scope of organelles in disease treatment [128,129] (Table 5).

3.3.2. Research Progress in Exosome Intranasal Delivery

Exosomes are small extracellular vesicles (EVs) secreted by various cells that have multiple functions, including anti-apoptotic and anti-inflammatory effects. Compared to stem cells, exosomes are easier to store, have lower immunogenicity, and can be used as drug carriers [9]. Intranasal delivery of stem cell-derived exosomes has shown significant therapeutic effects on various CNS diseases, such as ischemic stroke, TBI, SCI, perinatal brain injury, cognitive impairments, PD, and autism [9,130]. For example, intranasal delivery of exosomes derived from human umbilical mesenchymal stem cells effectively alleviates brain injury, providing a new avenue for stroke treatment [131]. Additionally, intranasal delivery of stem cell-derived exosomes can significantly improve cognitive impairments after subarachnoid hemorrhage, reduce neuronal apoptosis and inflammation, and significantly alleviate symptoms of experimental autoimmune encephalomyelitis, reducing inflammatory cell infiltration and demyelination while enhancing BBB integrity [5]. Intranasal delivery of exosomes also reduces neuronal apoptosis and inhibits microglial inflammation [132].

3.4. Promising Formulations and Translational Potential

A variety of intranasal drug delivery systems demonstrate significant translational potential for CNS disorders, each with distinct advantages and limitations. Nanoparticle-based formulations offer high clinical potential due to their versatility, scalability, and compatibility with diverse drugs. Liposomes are effective in delivering both hydrophilic and lipophilic agents, though limited by short nasal residence time [133]. Polymeric nanoparticles (e.g., PLGA, chitosan) provide controlled release and targeted brain delivery, with some progressing to clinical trials [51,134]. Self-emulsifying drug delivery systems (SEDDS) are ideal for lipophilic CNS drugs, enhancing solubility and bypassing first-pass metabolism. They show reduced systemic toxicity but require particle size control (<200 nm) and more data on long-term nasal safety [135,136]. Hydrogel-based formulations are especially thermosensitive in situ gels; these platforms offer sustained release and improved nasal retention. Applications like berberine and self-healing hydrogels show enhanced therapeutic effects, though mucosal irritation and crosslinker biocompatibility remain challenges [137,138]. Exosome-derived systems leverage their innate CNS targeting and immune tolerance, and exosomes effectively deliver proteins and nucleic acids across the BBB. MSC-derived exosomes show promise in neuroregeneration, but production scalability and formulation stability are key hurdles [139,140,141]. Viral vectors (e.g., AAVs): Clinically validated for gene therapy in neurological diseases, these vectors offer efficient gene transfer and strong neuronal tropism [142,143]. Innovations like FUSIN improve delivery precision, but immunogenicity, payload limits, and regulatory complexity must be addressed [3,144]. Overall, these systems represent a robust and diverse toolkit for advancing non-invasive CNS therapies, with ongoing research focused on overcoming technical and safety-related challenges.

4. Advancements in Strategies to Enhance Intranasal Drug Delivery

4.1. Advances in Drug Delivery Devices for Intranasal Administration

To enhance the delivery of drugs into the brain, researchers explored various strategies, such as mechanical stimulation. New intranasal drug delivery devices play a key role in improving drug delivery efficiency within the intranasal cavity, enhancing drug distribution uniformity, increasing drug retention time on the intranasal mucosa, and improving patient compliance [145]. For example, spray devices that precisely control drug dosage and spray patterns ensure that drugs are accurately delivered to specific regions of the intranasal cavity, particularly the olfactory and respiratory regions, which are important for brain targeting. Furthermore, improvements in dry powder inhalers allow drugs to be more evenly dispersed as dry powders within the intranasal cavity, extending the drug’s action time in the nose [1,146]. New catheter technologies and spray device designs optimize drug delivery to the CNS, enhancing organ-specific drug concentration [147,148]. For example, optimizing the catheter insertion angle and depth with 3D printing ensures that drugs are accurately delivered to the olfactory regions, avoiding accidental inhalation or swallowing, providing a new method for highly accurate, reproducible, region-specific drug delivery [149]. New intranasal implant drug delivery systems have also been developed, using radioactive labeled risperidone, which allows non-invasive monitoring of drug release through MicroSPECT/CT imaging, offering a new path for CNS disease treatment [150].
Ultrasound-mediated intranasal drug delivery (FUSIN) technology enhances drug delivery efficiency through transcranial ultrasound, bypasses the BBB, and enables non-invasive brain drug delivery, demonstrating significant clinical application potential [151,152]. FUSIN technology bypasses the BBB via the nasal route, enhances drug delivery efficiency, and minimizes systemic exposure in major organs such as the heart, lungs, liver, and kidneys, showcasing its potential as a non-invasive gene therapy platform. For example, FUSIN technology can efficiently and safely deliver adeno-associated viruses (AAVs) to specific brain regions, offering advantages of low systemic exposure and non-invasiveness, highlighting its potential in gene therapy [153]. Furthermore, innovative intranasal drug delivery methods, such as magnetic stem cell micro-robots [154,155], spray delivery of neuroactive peptides [156], three types of spray devices for insulin delivery [157], and transcranial magnetic stimulation combined with magnetic nanoparticles [158], have further improved drug delivery efficiency.

4.2. Progress in Intranasal Drug Delivery with Nanotechnology

Nanotechnology has emerged as a promising strategy for intranasal drug delivery, offering enhanced retention time, mucosal penetrability, and uptake efficiency, particularly valuable for treating CNS disorders [11]. Among the various nanocarrier platforms, liposomal systems have significantly improved brain drug absorption [15,159], while polymer-based nanoparticles, nanoemulsions, and other formulations have enhanced the ability of drugs to cross the BBB [12,14,108,160,161]. Recent advances in nano- and microparticle-based delivery systems have shown considerable potential in enhancing the treatment of CNS diseases. Novel nanocomposites have been effective in managing memory deficits and neurological pathologies [162]. Additionally, hyaluronic acid-modified nanocarriers have enhanced brain targeting and cellular uptake, offering innovative treatment strategies for AD [163]. Notably, intranasal delivery of polymer-based nanoparticles has yielded superior therapeutic outcomes compared to traditional administration routes. For example, lamotrigine-loaded PLGA nanoparticles achieved higher brain concentrations and improved efficacy in animal models than oral delivery [164]. Similarly, phenytoin-loaded chitosan nanoparticles provided sustained release and elevated brain-specific drug levels in rats [165], while PEGylated nanoparticles encapsulating miR-132 successfully crossed the nasal-brain barrier and improved cognitive performance in AD mouse models [166]. Beyond small molecules, nanocarrier systems have facilitated the intranasal delivery of neuroactive peptides, oxytocin, insulin, and other biologics, thereby broadening their clinical application [167]. Emerging delivery platforms such as magnetic nanoparticles and plant-derived extracellular vesicles (EVs) have also been applied to treat epilepsy and gliomas, significantly enhancing drug targeting and immune responses [168,169,170]. Furthermore, advanced formulations such as PEG-modified chitosan-lipid nanocapsules and carbon nanotubes have exhibited notable potential in nasal drug delivery, contributing to improved therapeutic outcomes [171,172].
Several disease-specific applications further underscore the versatility of these systems. Zolmitriptan-loaded liposomes have shown effectiveness in migraine treatment [133]. Biodegradable nanoparticles delivering thyrotropin-releasing hormone demonstrated favorable safety profiles in preclinical models [173]. Research has also optimized drug delivery using various nanocarriers, including liposomes containing antioxidants, naringin nanocapsules, and rutin nanolamellar vesicles, significantly increasing brain drug concentrations and therapeutic efficacy, particularly in epilepsy, memory disorders, and neuroinflammation [174,175,176]. Moreover, studies have explored novel intranasal delivery methods using self-assembled nanoparticles, EVs, and miRNA nanoparticles, showing potential for improving neurodegenerative diseases, brain glioblastomas, and ischemic brain injury [177,178,179,180,181,182]. Additionally, spanlastic nanovesicles, prepared using film hydration and modified spray technologies, have enhanced the delivery efficiency of rasagiline mesylate to the brain [183]. Intranasal drug delivery of nanoemulsions has significant advantages in improving the efficiency of CNS drug delivery. Nanoemulsions can enhance the ability of drugs to penetrate the olfactory epithelium, thereby improving the local drug accessibility and brain delivery efficiency of CNS drugs [184]. For example, meloxicam nanoemulsions, zotepine microemulsions, and memantine nanoemulsions exhibit superiority in enhancing drug solubility, permeability, and organ-specific drug concentration, significantly enhancing therapeutic effects [185,186,187]. Moreover, butter and omega-3 fatty acid-rich fish oil, as permeation enhancers, significantly increase the ability of drugs to penetrate the nasal mucosa and enhance brain absorption [188]. Vitamin E-loaded naringin nanoemulsions and lactoferrin-modified huperzine A nanoemulsions demonstrate their potential in increasing drug concentration and reducing side effects [189,190,191].
In conclusion, intranasal drug delivery technology, in combination with nanotechnology and biomaterials, provides a new non-invasive therapeutic approach for CNS diseases, immunotherapy, cancer treatment, and other fields, demonstrating enormous clinical application potential [171,192,193].

4.3. Research Progress in Hydrogels for Intranasal Drug Delivery

Hydrogels have a promising application in intranasal drug delivery, effectively improving drug efficacy and safety and providing new approaches and methods for the development of intranasal drug delivery systems. For example, thermosensitive hydrogels delivering berberine for the treatment of depression not only bypass the BBB but also prolong the drug’s action time, significantly enhancing brain-specific drug concentration and improving antidepressant effects [194]. Self-assembled thermosensitive in situ hydrogels co-delivering berberine and evodiamine significantly improved the drug’s organ-specific drug concentration in intranasal drug delivery [195]. Loading resveratrol into sodium alginate nanogels successfully improved depressive behaviors in chronic stress rats, demonstrating strong antidepressant potential [196]. Hydrogels loaded with EVs for intranasal delivery can also treat myocardial ischemia–reperfusion injury, showing higher absorption efficiency and therapeutic potential, providing a new direction for treating myocardial infarction-related diseases [197,198,199]. Additionally, the AXT-NLC13-G4 system, based on nanolipid carriers and in situ gel technology, significantly improved the drug’s targeting to the brain and cognitive improvement effects [200]. Carboxymethyl chitosan and sodium hyaluronate hydrocolloid systems, with good wettability and rapid release properties, are suitable for insulin intranasal delivery [201]. Intranasal drug delivery technologies targeting specific brain regions, combined with self-healing supramolecular hydrogel systems, successfully increased drug concentrations in the brain while bypassing first-pass liver metabolism, providing effective treatment methods for neurological diseases [202]. Moreover, disulfide nanoparticle emulsified gels effectively treated glioblastoma through intranasal delivery, showing excellent brain targeting and safety [203]. A representative preclinical study utilized a chitosan hydrogel loaded with liposomal donepezil HCl, showing significantly enhanced brain targeting for the treatment of Alzheimer’s disease [204]. Similarly, a thermosensitive hydrogel highlighted the potential of temperature-sensitive hydrogels in facilitating direct drug delivery to the brain via the intranasal route [205]. In the field of peptide delivery, insulin-conjugated poly(N-vinyl pyrrolidone)-based nanogels can significantly enhance insulin concentration and biological activity in the brain [206]. These cases reflect the translational promise of hydrogel-based nasal systems, especially in CNS therapy, chronic disease management, and non-invasive alternatives to injections. Hydrogels offer significant advantages for intranasal drug delivery, including strong mucoadhesion, sustained release, and good biocompatibility, enabling effective brain targeting and bypassing the BBB. They have shown promising results in treating CNS disorders, such as depression and glioblastoma, especially when combined with nanoparticles or self-healing systems. However, limitations remain, including poor permeability for large biomolecules, sensitivity to nasal environmental conditions, and challenges in large-scale production. Compared to nanoparticles and SEDDS, hydrogels provide better safety and retention but moderate CNS delivery efficiency, making them ideal for small-molecule delivery and combinational strategies. These novel systems provide a non-invasive and efficient delivery route for the treatment of CNS diseases.

4.4. Research Progress on Permeation Enhancers in Intranasal Drug Delivery

Although intranasal drug delivery is non-invasive and efficient, many drugs are difficult to apply due to low bioavailability. Permeation enhancers play a crucial role in enhancing drug absorption and delivery efficiency [207]. Recent advancements in intranasal drug delivery have highlighted the pivotal role of permeation enhancers in improving the bioavailability of therapeutics, particularly for CNS disorders. Among these, alkylsaccharides such as dodecyl maltoside and tetradecyl maltoside have been extensively studied. These compounds transiently open tight junctions in the nasal epithelium, facilitating enhanced drug absorption without causing significant mucosal damage. Notably, dodecyl maltoside has been incorporated into several FDA-approved intranasal formulations, including diazepam for seizure clusters, nalmefene for opioid overdose, and sumatriptan for migraines, underscoring its clinical relevance [208]. For example, lauroylcholine chloride as a permeation enhancer significantly improved the drug’s delivery efficiency from the nasal cavity to the brain [209]. L-penetratin enhances direct drug entry into the brain through the olfactory mucosa, especially increasing hippocampal drug accumulation, providing a new therapeutic strategy for neurodegenerative diseases [210]. Additionally, a new platform of lactobacillus lactate was developed as a mucosal vaccine, demonstrating its broad-spectrum protection potential [211]. By improving the intranasal delivery formulations of ketamine, tunicamycin, and insulin, organ-specific drug concentration and brain delivery effects were significantly enhanced [212,213,214,215]. A novel PTD used as an absorption enhancer improved insulin delivery, providing a convenient solution for diabetes treatment [215]. Permeation enhancers offer significant advantages in intranasal drug delivery by improving mucosal absorption and overcoming the low bioavailability. Agents like lauroylcholine chloride and L-penetratin have been shown to enhance brain targeting, particularly increasing hippocampal accumulation, which is beneficial for neurodegenerative disease treatment [216,217]. Novel platforms, such as lactobacillus-based mucosal vaccines and PTD-assisted insulin delivery, further expand nasal delivery applications to immunotherapy and metabolic disorders. However, barriers remain, including potential mucosal irritation, variability in enhancer efficacy across individuals, and limited long-term safety data. Compared to traditional systems, permeation enhancers provide a practical, non-invasive means to boost drug bioavailability and brain targeting, especially when integrated into advanced formulations like nanoparticle or hydrogel-based systems [218,219]. These studies offer new insights for the application of intranasal drug delivery in the treatment of neurological diseases and other conditions (Table 6).

5. Challenges and Future Research Directions in Intranasal Drug Delivery

Intranasal drug delivery offers multiple advantages, including non-invasiveness, ease of administration, high patient compliance, and the ability to bypass the BBB, thus enabling direct drug transport to the CNS [8,14,228,229]. These benefits make it an attractive alternative to invasive routes like intravenous or intrathecal administration, particularly for chronic neurological conditions. However, intranasal drug delivery also faces several physiological and technical challenges. Drug absorption is often limited by inherent anatomical and physiological features such as the nasal epithelium’s barrier function, mucociliary clearance, enzymatic degradation, and variable solubility of drugs, which together affect both the extent and consistency of drug absorption [8]. Furthermore, local metabolic activity and rapid clearance mechanisms in the nasal cavity can significantly reduce drug bioavailability before it reaches the brain.
Formulation development remains a major hurdle, requiring careful optimization of drug stability, controlled release kinetics, mucoadhesive properties, and targeting capability to ensure effective CNS delivery [8]. Although intranasal drug delivery has demonstrated a favorable safety profile in non-human primate studies. It especially excels at increasing brain exposure while reducing systemic side effects. However, repeated administration over extended periods may lead to mucosal irritation and potential olfactory dysfunction, necessitating more rigorous investigation into its long-term safety in humans [230]. Drugs and excipients used in formulations, such as permeation enhancers, surfactants, or cationic polymers like chitosan, may disrupt nasal epithelial integrity, leading to neuronal inflammation, degeneration, or altered signaling. Repeated or high-dose exposure to neuroactive drugs can also result in excitotoxicity or oxidative stress in olfactory neurons and trigeminal fibers. Although many delivery systems demonstrate improved bioavailability, their safety on neuronal structures remains insufficiently characterized [1,231]. Therefore, it is essential for review studies to emphasize the need for preclinical and clinical evaluations of both drug- and excipient-induced neuronal toxicity to ensure safe and effective intranasal therapies.
Beyond physiological and formulation hurdles, intranasal drug delivery also faces regulatory and translational challenges. Currently, there is a lack of standardized FDA or EMA guidelines specifically tailored for CNS-targeted nasal delivery systems, and much of the regulatory evaluation relies on surrogate endpoints such as CSF levels rather than direct brain biodistribution [14,228]. Moreover, significant anatomical and physiological differences between animal models and humans contribute to discrepancies in clinical outcomes, as observed in trials like the modest translation of intranasal insulin efficacy from rodents to Alzheimer’s patients [229].
To overcome these limitations and fully realize the potential of intranasal drug delivery, future research should focus on the development of bioresponsive and smart formulations that adapt to the nasal environment, integration of AI and machine learning for personalized drug delivery optimization, establishment of predictive human-relevant in vitro and in silico models, and the design of scalable, cost-effective manufacturing platforms. In parallel, long-term clinical studies are needed to evaluate mucosal safety and efficacy across diverse populations [228,229].
This review identifies the following future research directions. First, it is essential to develop novel drug delivery systems, such as nanoparticles and gels, that effectively enhance drug absorption and minimize local side effects. Second, research should focus on individual differences in responses to intranasal drug delivery, aiming to develop personalized treatment plans to improve efficacy and reduce adverse effects. Additionally, long-acting, controlled-release systems are an important direction in intranasal drug delivery development. By controlling drug release, the duration of drug action can be extended, and the frequency of administration can be reduced. Furthermore, with the integration of artificial intelligence, in vitro experiments and computer simulations should be used to evaluate the behavior of intranasal drug delivery devices and formulations, combined with high-resolution imaging techniques and computational fluid dynamics simulations, to better understand the drug delivery process. Lastly, driven by single-cell technologies, efforts should be made to enhance the construction of the nasal-brain axis cell atlas, advancing intranasal drug delivery research from traditional pharmacology to the paradigm of precision medicine.

6. Conclusions

Intranasal drug delivery has emerged as a highly promising strategy for overcoming the limitations imposed by the BBB in the treatment of CNS diseases. As this review demonstrates, intranasal drug delivery methods provide direct, non-invasive access to the brain, enabling effective delivery of a wide range of therapeutics, including small molecules, biomacromolecules, nucleic acids, and cell-derived therapies. In addition, this method minimizes systemic exposure and enhances patient compliance. Mechanistically, both intracellular and extracellular transport mechanisms are involved, with formulation strategies such as nanoparticles, hydrogels, and permeation enhancers playing a central role in improving drug absorption, retention, and targeting efficiency. Significant progress has been made in optimizing delivery systems for various CNS disorders, including AD, PD, stroke, and brain tumors. Despite physiological and technical challenges, such as mucociliary clearance, local toxicity, and manufacturing complexities, the accumulating preclinical and clinical evidence supports the feasibility and translational potential of intranasal drug delivery. This review consolidates key advances and provides a comprehensive understanding of the mechanisms, applications, and challenges of this approach, offering a solid scientific foundation for its continued development in CNS therapeutics.

Author Contributions

Writing—original draft preparation, X.W.; writing—review and editing, X.W., R.Z., Y.Q., Y.Z., J.P., Z.C., S.W., M.L., and Y.D.; supervision, X.W. and Y.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Fujian Natural Science Foundation (2022J05062), the Xiamen Natural Science Foundation (3502Z20227041), and the Scientific Research Funds of Huaqiao University (21bs126).

Institutional Review Board Statement

Not applicable.

Acknowledgments

During the preparation of this manuscript, the authors used OpenAI’s ChatGPT-4o (June 2024 release) for the purposes of linguistic polishing. The authors have reviewed and edited the output and take full responsibility for the content of this publication. The authors thank Servier Medical Art for providing freely available medical illustrations (https://smart.servier.com/), accessed on 5 April 2025.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

AAVsAdeno-associated viruses
ADAlzheimer’s disease
ASOAntisense oligonucleotide
BBBBlood–brain barrier
BMSCsBone marrow-derived mesenchymal stem cells
BPDBronchopulmonary dysplasia
CSFCerebrospinal fluid
CNSCentral nervous system
DPSCsDeciduous dental pulp stem cells
EVsExtracellular vesicles
FUSINFocused ultrasound-mediated nasal drug delivery
GLP-1Glucagon-like peptide-1
IGF-1Insulin-like growth factor 1
hNSCsHuman neural stem cells
LIFLeukemia inhibitory factor
MSCMesenchymal stem cell
NGFNerve growth factor
OMPCsOlfactory mucosal progenitor cells
PDParkinson’s disease
PEGPolyethylene glycol
PTDProtein transduction domain
SADSocial anxiety disorder
SCISpinal cord injury
TBITraumatic brain injury
ZO-1Zonula occludens-1

References

  1. Agrawal, M.; Saraf, S.; Saraf, S.; Antimisiaris, S.; Chougule, M.; Shoyele, S.; Alexander, A. Nose-to-brain drug delivery: An update on clinical challenges and progress towards approval of anti-Alzheimer drugs. J. Control. Release 2018, 281, 139–177. [Google Scholar] [CrossRef] [PubMed]
  2. Kou, D.; Gao, Y.; Li, C.; Zhou, D.; Lu, K.; Wang, N.; Zhang, R.; Yang, Z.; Zhou, Y.; Chen, L.; et al. Intranasal Pathway for Nanoparticles to Enter the Central Nervous System. Nano Lett. 2023, 23, 5381–5390. [Google Scholar] [CrossRef] [PubMed]
  3. Chen, H.; Chen, C.C.; Acosta, C.; Wu, S.Y.; Sun, T.; Konofagou, E.E. A new brain drug delivery strategy: Focused ultrasound-enhanced intranasal drug delivery. PLoS ONE 2014, 9, e108880. [Google Scholar] [CrossRef] [PubMed]
  4. Lochhead, J.J.; Davis, T.P. Perivascular and Perineural Pathways Involved in Brain Delivery and Distribution of Drugs after Intranasal Administration. Pharmaceutics 2019, 11, 598. [Google Scholar] [CrossRef]
  5. Wu, J.; Li, A.; Shi, Y.; Wang, Y.; Luo, J.; Zhuang, W.; Ma, X.; Qiao, Z.; Xiu, X.; Lang, X.; et al. Intranasal delivery of mesenchymal stem cell-derived exosomes ameliorates experimental autoimmune encephalomyelitis. Int. Immunopharmacol. 2025, 146, 113853. [Google Scholar] [CrossRef]
  6. Jia, Y.; Xu, L.; Leng, S.; Sun, Y.; Huang, X.; Wang, Y.; Ren, H.; Li, G.; Bai, Y.; Zhang, Z.; et al. Nose-to-Brain Delivery of Circular RNA SCMH1-Loaded Lipid Nanoparticles for Ischemic Stroke Therapy. Adv. Mater. 2025, 37, 2500598. [Google Scholar] [CrossRef]
  7. Snyers, D.; Tribolet, S.; Rigo, V. Intranasal Analgosedation for Infants in the Neonatal Intensive Care Unit: A Systematic Review. Neonatology 2022, 119, 273–284. [Google Scholar] [CrossRef]
  8. Keller, L.-A.; Merkel, O.; Popp, A. Intranasal drug delivery: Opportunities and toxicologic challenges during drug development. Drug Deliv. Transl. Res. 2021, 12, 735–757. [Google Scholar] [CrossRef]
  9. Gotoh, S.; Kawabori, M.; Fujimura, M. Intranasal administration of stem cell-derived exosomes for central nervous system diseases. Neural Regen. Res. 2024, 19, 1249–1255. [Google Scholar] [CrossRef]
  10. Gandhi, S.; Shastri, D.H.; Shah, J.; Nair, A.B.; Jacob, S. Nasal Delivery to the Brain: Harnessing Nanoparticles for Effective Drug Transport. Pharmaceutics 2024, 16, 481. [Google Scholar] [CrossRef]
  11. Zha, S.; Wong, K.L.; All, A.H. Intranasal Delivery of Functionalized Polymeric Nanomaterials to the Brain. Adv. Healthc. Mater. 2022, 11, 2102610. [Google Scholar] [CrossRef] [PubMed]
  12. Awad, R.; Avital, A.; Sosnik, A. Polymeric nanocarriers for nose-to-brain drug delivery in neurodegenerative diseases and neurodevelopmental disorders. Acta Pharm. Sin. B 2023, 13, 1866–1886. [Google Scholar] [CrossRef] [PubMed]
  13. Kosyakovsky, J.; Witthuhn, B.A.; Svitak, A.L.; Frey, W.H.; Hanson, L.R.; Fine, J.M. Quantifying Intranasally Administered Deferoxamine in Rat Brain Tissue with Mass Spectrometry. ACS Chem. Neurosci. 2019, 10, 4571–4578. [Google Scholar] [CrossRef] [PubMed]
  14. Patel, A.A.; Patel, R.J.; Patel, S.R. Nanomedicine for Intranasal Delivery to Improve Brain Uptake. Curr. Drug Deliv. 2018, 15, 461–469. [Google Scholar] [CrossRef]
  15. Duong, V.-A.; Nguyen, T.-T.-L.; Maeng, H.-J. Recent Advances in Intranasal Liposomes for Drug, Gene, and Vaccine Delivery. Pharmaceutics 2023, 15, 207. [Google Scholar] [CrossRef]
  16. Pandya, J.D.; Musyaju, S.; Modi, H.R.; Okada-Rising, S.L.; Bailey, Z.S.; Scultetus, A.H.; Shear, D.A. Intranasal delivery of mitochondria targeted neuroprotective compounds for traumatic brain injury: Screening based on pharmacological and physiological properties. J. Transl. Med. 2024, 22, 167. [Google Scholar] [CrossRef]
  17. Ojo, A.S.; Odipe, O.G.; Owoseni, O. Improving the Emergency Department Management of Sickle Cell Vaso-Occlusive Pain Crisis: The Role and Options of Sublingual and Intranasally Administered Analgesia. J. Clin. Med. Res. 2023, 15, 10–22. [Google Scholar] [CrossRef]
  18. Martirosov, A.; Giuliano, C.; Shupp, M.; Channey, S.; Kale-Pradhan, P.B. Zavegepant Intranasal Spray for Migraines. Ann. Pharmacother. 2024, 58, 827–833. [Google Scholar] [CrossRef]
  19. Mehta, M.U.; Uppoor, R.S.; Roca, R.A.; Sabarinath, S.; Florian, J.; Xu, Y.; Nallani, S.C.; Li, Z.; Brescia-Oddo, T. FDA Approval Summary: Nalmefene Nasal Spray for the Emergency Treatment of Known or Suspected Opioid Overdose. Clin. Pharmacol. Ther. 2024, 117, 620–626. [Google Scholar] [CrossRef]
  20. Sposito, B.; Broggi, A.; Pandolfi, L.; Crotta, S.; Clementi, N.; Ferrarese, R.; Sisti, S.; Criscuolo, E.; Spreafico, R.; Long, J.; et al. The interferon landscape along the respiratory tract impacts the severity of COVID-19. Cell 2021, 184, 4953–4968.e16. [Google Scholar] [CrossRef]
  21. Buyze, J.; Llorca, P.-M.; Messer, T.; Rive, B.; Young, A.H.; Frey, R.; Bitter, I.; Fu, D.-J.; Kambarov, Y.; Reif, A.; et al. Esketamine Nasal Spray versus Quetiapine for Treatment-Resistant Depression. N. Engl. J. Med. 2023, 389, 1298–1309. [Google Scholar] [CrossRef]
  22. Illum, L. Transport of drugs from the nasal cavity to the central nervous system. Eur. J. Pharm. Sci. 2000, 11, 1–18. [Google Scholar] [CrossRef] [PubMed]
  23. Thorne, R.G.; Lochhead, J.J. Intranasal delivery of biologics to the central nervous system. Adv. Drug Deliv. Rev. 2011, 64, 614–628. [Google Scholar] [CrossRef]
  24. Crowe, T.P.; Greenlee, M.H.W.; Kanthasamy, A.G.; Hsu, W.H. Mechanism of intranasal drug delivery directly to the brain. Life Sci. 2018, 195, 44–52. [Google Scholar] [CrossRef]
  25. Mestre, H.; Rasmussen, M.K.; Nedergaard, M. The glymphatic pathway in neurological disorders. Lancet Neurol. 2018, 17, 1016–1024. [Google Scholar] [CrossRef]
  26. Illum, L. pharmacology. Is nose-to-brain transport of drugs in man a reality? J. Pharm. Pharmacol. 2004, 56, 3–17. [Google Scholar] [CrossRef]
  27. Bagley, J.H.; Geltzeiler, M.; Liu, J.J.; Sanusi, O.R.; Piantino, J.; Dogan, A.; Yamamoto, E.A. The perivascular space is a conduit for cerebrospinal fluid flow in humans: A proof-of-principle report. Proc. Natl. Acad. Sci. USA 2024, 121, e2407246121. [Google Scholar] [CrossRef]
  28. Wu, H.; Zhou, Y.; Wang, Y.; Tong, L.; Wang, F.; Song, S.; Xu, L.; Liu, B.; Yan, H.; Sun, Z. Current State and Future Directions of Intranasal Delivery Route for Central Nervous System Disorders: A Scientometric and Visualization Analysis. Front. Pharmacol. 2021, 12, 717192. [Google Scholar] [CrossRef]
  29. Saka, R.; Chella, N.; Khan, W. Development of Imatinib Mesylate-Loaded Liposomes for Nose to Brain Delivery: In Vitro and In Vivo Evaluation. AAPS PharmSciTech 2021, 22, 192. [Google Scholar] [CrossRef]
  30. Katona, G.; Csóka, I.; Sipos, B. Risk-Assessment-Based Optimization Favours the Development of Albumin Nanoparticles with Proper Characteristics Prior to Drug Loading. Pharmaceutics 2022, 14, 2036. [Google Scholar] [CrossRef]
  31. Baboota, S.; Kumar, S.; Arora, A.; Ali, J. Intranasal delivery of tetrabenazine nanoemulsion via olfactory region for better treatment of hyperkinetic movement associated with Huntington’s disease: Pharmacokinetic and brain delivery study. Chem. Phys. Lipids 2020, 230, 104917. [Google Scholar] [CrossRef]
  32. Agrawal, M.; Ajazuddin; Murty, U.S.; Ravichandiran, V.; Dubey, S.K.; Saraf, S.; Saraf, S.; Alexander, A.; Patel, R.J.; Puri, A. Recent strategies and advances in the fabrication of nano lipid carriers and their application towards brain targeting. J. Control. Release 2020, 321, 372–415. [Google Scholar] [CrossRef]
  33. Jing, T.; Xinchen, Y.; Jiaoqiong, G. Lipid-based nanoparticles via nose-to-brain delivery: A mini review. Front. Cell Dev. Biol. 2023, 11, 1214450. [Google Scholar] [CrossRef]
  34. Préat, V.; Beloqui, A.; Rieux, A.D. Mechanisms of transport of polymeric and lipidic nanoparticles across the intestinal barrier. Adv. Drug Deliv. Rev. 2016, 106, 242–255. [Google Scholar] [CrossRef]
  35. Dhuria, S.V.; Hanson, L.R.; Frey, W.H. Intranasal delivery to the central nervous system: Mechanisms and experimental considerations. J. Pharm. Sci. 2010, 99, 1654–1673. [Google Scholar] [CrossRef]
  36. Hanson, L.R.; Frey, W.H. Intranasal delivery bypasses the blood-brain barrier to target therapeutic agents to the central nervous system and treat neurodegenerative disease. BMC Neurosci. 2008, 9, S5. [Google Scholar] [CrossRef]
  37. Andresen, M.J.F. Understanding diverse TRPV1 signaling—An update. F1000Research 2019, 8, 1978. [Google Scholar] [CrossRef] [PubMed]
  38. Hsu, W.H.; Crowe, T.P. Evaluation of Recent Intranasal Drug Delivery Systems to the Central Nervous System. Pharmaceutics 2022, 14, 629. [Google Scholar] [CrossRef]
  39. Dighe, S.; Jog, S.; Momin, M.; Sawarkar, S.; Omri, A. Intranasal Drug Delivery by Nanotechnology: Advances in and Challenges for Alzheimer’s Disease Management. Pharmaceutics 2024, 16, 58. [Google Scholar] [CrossRef]
  40. de Lange, E.C.M.; Ruigrok, M.J.R. Emerging Insights for Translational Pharmacokinetic and Pharmacokinetic-Pharmacodynamic Studies: Towards Prediction of Nose-to-Brain Transport in Humans. AAPS J. 2015, 17, 493–505. [Google Scholar] [CrossRef]
  41. Padmanabhan, V.; Frey, W.; Thorne, R.; Pronk, G. Delivery of insulin-like growth factor-I to the rat brain and spinal cord along olfactory and trigeminal pathways following intranasal administration. Neuroscience 2004, 127, 481–496. [Google Scholar] [CrossRef]
  42. Gänger, S.; Schindowski, K. Tailoring Formulations for Intranasal Nose-to-Brain Delivery: A Review on Architecture, Physico-Chemical Characteristics and Mucociliary Clearance of the Nasal Olfactory Mucosa. Pharmaceutics 2018, 10, 116. [Google Scholar] [CrossRef] [PubMed]
  43. Huang, Q.; Yu, S.; Gong, G.; Chen, X.; Shu, H. Research progress in brain-targeted nasal drug delivery. Front. Aging Neurosci. 2023, 15, 1341295. [Google Scholar] [CrossRef]
  44. Chiono, V.; Marcello, E. Biomaterials-Enhanced Intranasal Delivery of Drugs as a Direct Route for Brain Targeting. Int. J. Mol. Sci. 2023, 24, 3390. [Google Scholar] [CrossRef]
  45. Shen, H.; Aggarwal, N.; Cui, B.; Foo, G.; He, Y.; Srivastava, S.; Li, S.; Seah, M.; Wun, K.; Ling, H.; et al. Engineered commensals for targeted nose-to-brain drug delivery. Cell 2025, 188, 1545–1562.e16. [Google Scholar] [CrossRef]
  46. Ladel, S.; Flamm, J.; Schlossbauer, P.; Mizaikoff, B.; Luksch, H.; Schindowski, K. Improved In Vitro Model for Intranasal Mucosal Drug Delivery: Primary Olfactory and Respiratory Epithelial Cells Compared with the Permanent Nasal Cell Line RPMI 2650. Pharmaceutics 2019, 11, 367. [Google Scholar] [CrossRef] [PubMed]
  47. Lundgaard, I.; Munk, A.S.F.; Nedergaard, M.; Jessen, N.A. The Glymphatic System: A Beginner’s Guide. Neurochem. Res. 2015, 40, 2583–2599. [Google Scholar] [CrossRef]
  48. Illum, L.; Exposito-Harris, R.; Heras, A.; Vllasaliu, D.; Garnett, M.; Stolnik, S.; Casettari, L. Tight junction modulation by chitosan nanoparticles: Comparison with chitosan solution. Int. J. Pharm. 2010, 400, 183–193. [Google Scholar] [CrossRef]
  49. Craft, S.; Raman, R.; Chow, T.W.; Rafii, M.S.; Sun, C.K.; Rissman, R.A.; Donohue, M.C.; Brewer, J.B.; Jenkins, C.; Harless, K.; et al. Safety, Efficacy, and Feasibility of Intranasal Insulin for the Treatment of Mild Cognitive Impairment and Alzheimer Disease Dementia: A Randomized Clinical Trial. JAMA Neurol. 2020, 77, 1099–1109. [Google Scholar] [CrossRef]
  50. Sotomayor, P.; Morales, P.; Acuña, E.; González, L.F.; Arellano, G.; Oyarzun-Ampuero, F.; Naves, R. Intranasal delivery of interferon-β-loaded nanoparticles induces control of neuroinflammation in a preclinical model of multiple sclerosis: A promising simple, effective, non-invasive, and low-cost therapy. J. Control. Release 2021, 331, 443–459. [Google Scholar] [CrossRef]
  51. Nanaki, S.G.; Spyrou, K.; Papadopoulos, G.C.; Bekiari, C.; Veneti, P.; Karouta, N.; Bikiaris, D.N.; Baroud, T.N.; Gournis, D.; Grivas, I. Hierarchical Porous Carbon-PLLA and PLGA Hybrid Nanoparticles for Intranasal Delivery of Galantamine for Alzheimer’s Disease Therapy. Pharmaceutics 2020, 12, 227. [Google Scholar] [CrossRef] [PubMed]
  52. Gonçalves, J.; Alves, G.; Fonseca, C.; Carona, A.; Bicker, J.; Falcão, A.; Fortuna, A. Is intranasal administration an opportunity for direct brain delivery of lacosamide? Eur. J. Pharm. Sci. 2021, 157, 105632. [Google Scholar] [CrossRef] [PubMed]
  53. Zolkowska, D.; Wu, C.-Y.; Rogawski, M.A. Intranasal Allopregnanolone Confers Rapid Seizure Protection: Evidence for Direct Nose-to-Brain Delivery. Neurotherapeutics 2021, 18, 544–555. [Google Scholar] [CrossRef]
  54. Hong, Z.; Shuo, L.; Zhuo, L.; Shuo, Y.; Dandan, L.; Jiaolin, Z.; Hongmei, G.; Ling, Y.; YingLi, G.; Longxuan, L.; et al. Intranasal delivery of 9-cis retinoic acid reduces beta-amyloid deposition via inhibiting astrocyte-mediated inflammation. Aging 2020, 12, 5469–5478. [Google Scholar] [CrossRef]
  55. Bhargavi, M.; Sai Sarath, G.; Surana, P.; Dhull, K.S.; Shaikh, M.; Rajan, M. Efficacy of Intranasal Atomized Dexmedetomidine for Sedation in Surgical Removal of Impacted Mandibular Third Molars: A Prospective Study. Cureus 2023, 15, e36721. [Google Scholar] [CrossRef]
  56. Korczeniewska, O.A.; Tatineni, K.; Faheem, S.; Fresin, W.; Bonitto, J.; Khan, J.; Eliav, E.; Benoliel, R. Effects of intra-nasal melanocortin-4 receptor antagonist on trigeminal neuropathic pain in male and female rats. Neurosci. Lett. 2023, 796, 137054. [Google Scholar] [CrossRef] [PubMed]
  57. Abolfazl, F.; Nafiseh, F.-M.; Zeinab, N.; Mouna Faghani, A. Effect of Intranasal Ketamine on Pain Intensity after Cesarean Section: A Single-Center, Double Blind, Randomized Controlled Trial. Ethiop. J. Health Sci. 2023, 33, 55–64. [Google Scholar] [CrossRef]
  58. Siegler, B.H.; Gruß, M.; Oehler, B.; Keßler, J.; Fluhr, H.; Weis, C.; Schulz, F.; Weigand, M.A. Intranasale Lidocainvernebelung als neue und nichtinvasive Therapieoption des Postpunktionskopfschmerzes. Der Anaesthesist 2020, 70, 392–397. [Google Scholar] [CrossRef]
  59. Micheli, L.; Di Cesare Mannelli, L.; Lucarini, E.; Parisio, C.; Toti, A.; Fiorentino, B.; Rigamonti, M.A.; Calosi, L.; Ghelardini, C. Intranasal Low-Dose Naltrexone Against Opioid Side Effects: A Preclinical Study. Front. Pharmacol. 2020, 11, 576624. [Google Scholar] [CrossRef]
  60. Ingielewicz, A.; Szymczak, R.K. Intranasal Therapy in Palliative Care. Pharmaceutics 2024, 16, 519. [Google Scholar] [CrossRef]
  61. Nicholas, T.G.; Husbands, E.L. Benefits of Intranasal Administration of Diamorphine and Midazolam in the Management of Patients Receiving Palliative Care in the Community: A Case Series. J. Pain Palliat. Care Pharmacother. 2022, 36, 34–39. [Google Scholar] [CrossRef] [PubMed]
  62. Pérez-Osorio, I.N.; Espinosa, A.; Giraldo Velázquez, M.; Padilla, P.; Bárcena, B.; Fragoso, G.; Jung-Cook, H.; Besedovsky, H.; Meneses, G.; Sciutto Conde, E.L. Nose-to-Brain Delivery of Dexamethasone: Biodistribution Studies in Mice. J. Pharmacol. Exp. Ther. 2021, 378, 244–250. [Google Scholar] [CrossRef]
  63. Xu, D.; Lu, Y.-R.; Kou, N.; Hu, M.-J.; Wang, Q.-S.; Cui, Y.-L. Intranasal delivery of icariin via a nanogel-thermoresponsive hydrogel compound system to improve its antidepressant-like activity. Int. J. Pharm. 2020, 586, 119550. [Google Scholar] [CrossRef] [PubMed]
  64. Tiozzo Fasiolo, L.; Manniello, M.D.; Banella, S.; Napoli, L.; Bortolotti, F.; Quarta, E.; Colombo, P.; Balafas, E.; Kostomitsopoulos, N.; Rekkas, D.M.; et al. Flurbiprofen sodium microparticles and soft pellets for nose-to-brain delivery: Serum and brain levels in rats after nasal insufflation. Int. J. Pharm. 2021, 605, 120827. [Google Scholar] [CrossRef]
  65. Harris, H.M.; Boyet, K.L.; Liu, H.; Dwivedi, R.; Ashpole, N.M.; Tandon, R.; Bidwell, G.L.; Cheng, Z.; Fassero, L.A.; Yu, C.S.; et al. Safety and Pharmacokinetics of Intranasally Administered Heparin. Pharm. Res. 2022, 39, 541–551. [Google Scholar] [CrossRef]
  66. Gallegos, C.E.; Bartos, M.; Gumilar, F.; Minetti, A.; Baier, C.J. Behavioral and neurochemical impairments after intranasal administration of chlorpyrifos formulation in mice. Pestic. Biochem. Physiol. 2023, 189, 105315. [Google Scholar] [CrossRef] [PubMed]
  67. Chiaretti, A.; Eftimiadi, G.; Soligo, M.; Manni, L.; Di Giuda, D.; Calcagni, M. Topical delivery of nerve growth factor for treatment of ocular and brain disorders. Neural Regen. Res. 2021, 16, 1740–1750. [Google Scholar] [CrossRef]
  68. Manni, L.; Conti, G.; Chiaretti, A.; Soligo, M. Intranasal Delivery of Nerve Growth Factor in Neurodegenerative Diseases and Neurotrauma. Front. Pharmacol. 2021, 12, 754502. [Google Scholar] [CrossRef]
  69. Soligo, M.; Manni, L.; Conti, G.; Chiaretti, A. Intranasal nerve growth factor for prevention and recovery of the outcomes of traumatic brain injury. Neural Regen. Res. 2023, 18, 773–778. [Google Scholar] [CrossRef]
  70. Antonio, G.; Lavinia, C.; Giorgio, C.; Gemma, E.; Serena, F.; Luigi, M.; Antonietta, C.; Benedetta, G.; Lorenzo, D.S.; Maria Lucia, C.; et al. Intranasal human-recombinant NGF administration improves outcome in children with post-traumatic unresponsive wakefulness syndrome. Biol. Direct 2023, 18, 61. [Google Scholar] [CrossRef]
  71. D’Mello, V.; Subramaniam, M.; Bhalla, A.P.; Saavedra, S.; Leiba, O.; Levison, S.W. Intranasal Leukemia Inhibitory Factor Attenuates Gliosis and Axonal Injury and Improves Sensorimotor Function After a Mild Pediatric Traumatic Brain Injury. Neurotrauma Rep. 2023, 4, 236–250. [Google Scholar] [CrossRef] [PubMed]
  72. Zorina, I.I.; Avrova, N.F.; Zakharova, I.O.; Shpakov, A.O. Prospects for the Use of Intranasally Administered Insulin and Insulin-Like Growth Factor-1 in Cerebral Ischemia. Biochemistry 2023, 88, 374–391. [Google Scholar] [CrossRef] [PubMed]
  73. Tashima, T. Shortcut Approaches to Substance Delivery into the Brain Based on Intranasal Administration Using Nanodelivery Strategies for Insulin. Molecules 2020, 25, 5188. [Google Scholar] [CrossRef]
  74. Attama, A.A.; Ofokansi, K.C.; Mumuni, M.A.; Díaz, D.D.; Kenechukwu, F.C. Insulin-loaded mucoadhesive nanoparticles based on mucin-chitosan complexes for oral delivery and diabetes treatment. Carbohydr. Polym. 2019, 229, 115506. [Google Scholar] [CrossRef]
  75. Shen, X.; Mao, S.; Gao, M.; Kou, Y.; Huo, Y.; Sun, Z.; Liu, C.; Sun, Y.; Zhang, X. Effect of Glyceryl Monocaprylate-Modified Chitosan on the Intranasal Absorption of Insulin in Rats. J. Pharm. Sci. 2019, 108, 3623–3629. [Google Scholar] [CrossRef]
  76. Wong, C.Y.J.; Baldelli, A.; Hoyos, C.M.; Tietz, O.; Ong, H.X.; Traini, D. Insulin Delivery to the Brain via the Nasal Route: Unraveling the Potential for Alzheimer’s Disease Therapy. Drug Deliv. Transl. Res. 2024, 14, 1776–1793. [Google Scholar] [CrossRef] [PubMed]
  77. Hallschmid, M. Intranasal Insulin for Alzheimer’s Disease. CNS Drugs 2021, 35, 21–37. [Google Scholar] [CrossRef]
  78. Fine, J.M.; Stroebel, B.M.; Faltesek, K.A.; Terai, K.; Haase, L.; Knutzen, K.E.; Kosyakovsky, J.; Bowe, T.J.; Fuller, A.K.; Frey, W.H.; et al. Intranasal delivery of low-dose insulin ameliorates motor dysfunction and dopaminergic cell death in a 6-OHDA rat model of Parkinson’s Disease. Neurosci. Lett. 2020, 714, 134567. [Google Scholar] [CrossRef]
  79. Bae, H.-D.; Lee, J.-S.; Pyun, H.; Kim, M.; Lee, K. Optimization of formulation for enhanced intranasal delivery of insulin with translationally controlled tumor protein-derived protein transduction domain. Drug Deliv. 2019, 26, 622–628. [Google Scholar] [CrossRef]
  80. Salah, M.; Mahmoud, S.; Hammam, O.A.; Kandeel, W.; Attia, Y.M.; Maher, M.A. Histopathological evaluation of insulin-DMSO formula designed for direct nose-to-brain delivery. Histol. Histopathol. 2022, 37, 431–439. [Google Scholar] [CrossRef]
  81. Zhang, T.-T.; Cai, Q.-L.; Gu, Z.-Y.; Song, T.-Y. Novel ACE-inhibiting peptides from soybean protein hydrolysates by peptidomics combined with in silico analysis and their inhibitory effects on proliferation and migration of Ang II-induced VSMCs. Food Med. Homol. 2024, 1, 9420023. [Google Scholar] [CrossRef]
  82. Novak, V.; Mantzoros, C.; Novak, P.; McGlinchey, R.; Dai, W.; Lioutas, V.; Buss, S.; Fortier, C.; Khan, F.; Aponte Becerra, L.; et al. MemAID: Memory advancement with intranasal insulin vs. placebo in type 2 diabetes and control participants: A randomized clinical trial. J. Neurol. 2022, 269, 4817–4835. [Google Scholar] [CrossRef] [PubMed]
  83. Khan, T.T.S.; Sheikh, Z.; Maleknia, S.; Oveissi, F.; Fathi, A.; Abrams, T.; Ong, H.X.; Traini, D. Intranasal delivery of glucagon-like peptide-1 to the brain for obesity treatment: Opportunities and challenges. Expert Opin. Drug Deliv. 2024, 21, 1081–1101. [Google Scholar] [CrossRef]
  84. De Luca, V.; Ishii, D.; Kageyama, M.; Umeda, S. Cerebral and extracerebral distribution of radioactivity associated with oxytocin in rabbits after intranasal administration: Comparison of TTA-121, a newly developed oxytocin formulation, with Syntocinon. PLoS ONE 2021, 16, e0261451. [Google Scholar] [CrossRef]
  85. DuBois, M.; Tseng, A.; Francis, S.M.; Haynos, A.F.; Peterson, C.B.; Jacob, S. Utility of Downstream Biomarkers to Assess and Optimize Intranasal Delivery of Oxytocin. Pharmaceutics 2022, 14, 1178. [Google Scholar] [CrossRef] [PubMed]
  86. Guastella, A.J.; Howard, A.L.; Mitchell, P.; Carson, D.S.; Dadds, M. A randomized controlled trial of intranasal oxytocin as an adjunct to exposure therapy for social anxiety disorder. Psychoneuroendocrinology 2009, 34, 917–923. [Google Scholar] [CrossRef]
  87. Rayala, R.; Tiller, A.; Majumder, S.A.; Stacy, H.M.; Eans, S.O.; Nedovic, A.; McLaughlin, J.P.; Cudic, P. Solid-Phase Synthesis of the Bicyclic Peptide OL-CTOP Containing Two Disulfide Bridges, and an Assessment of Its In Vivo μ-Opioid Receptor Antagonism after Nasal Administration. Molecules 2023, 28, 1822. [Google Scholar] [CrossRef]
  88. Hiroki, T.; Keiko, I.; Bin, J.; Kayoko, T.; Takako, E.; Keizo, T.; Tsuyoshi, M.; Masato, H.; Naruhiko, S.; Nobuhisa, I.; et al. Nasal vaccine delivery attenuates brain pathology and cognitive impairment in tauopathy model mice. npj Vaccines 2020, 5, 28. [Google Scholar] [CrossRef]
  89. Tonelli, L.H.; Postolache, T.T. Airborne inflammatory factors: “From the nose to the brain”. Front. Biosci. 2010, 2, 135–152. [Google Scholar] [CrossRef]
  90. Barnett, E.M.; Perlman, S. The olfactory nerve and not the trigeminal nerve is the major site of CNS entry for mouse hepatitis virus, strain JHM. Virology 1993, 194, 185–191. [Google Scholar] [CrossRef]
  91. Kristensson, K. Microbes’ roadmap to neurons. Nat. Rev. Neurosci. 2011, 12, 345–357. [Google Scholar] [CrossRef] [PubMed]
  92. Belur, L.; Temme, A.; Podetz-Pedersen, K.; Riedl, M.; Vulchanova, L.; Robinson, N.; Hanson, L.; Kozarsky, K.; Orchard, P.; Frey, W.; et al. Intranasal Adeno-Associated Virus Mediated Gene Delivery and Expression of Human Iduronidase in the Central Nervous System: A Noninvasive and Effective Approach for Prevention of Neurologic Disease in Mucopolysaccharidosis Type I. Hum. Gene Ther. 2017, 28, 576–587. [Google Scholar] [CrossRef] [PubMed]
  93. Ye, D.; Chukwu, C.; Yang, Y.; Hu, Z.; Chen, H. Adeno-associated virus vector delivery to the brain: Technology advancements and clinical applications. Adv. Drug Deliv. Rev. 2024, 211, 115363. [Google Scholar] [CrossRef]
  94. Maguire, C.A.; Peters, C.W.; Hanlon, K.S. Delivering AAV to the Central Nervous and Sensory Systems. Trends Pharmacol. Sci. 2021, 42, 461–474. [Google Scholar] [CrossRef]
  95. Cossette, B.; Kelly, S.H.; Collier, J.H. Intranasal Subunit Vaccination Strategies Employing Nanomaterials and Biomaterials. ACS Biomater. Sci. Eng. 2020, 7, 1765–1779. [Google Scholar] [CrossRef]
  96. Calzas, C.; Chevalier, C. Innovative Mucosal Vaccine Formulations Against Influenza A Virus Infections. Front. Immunol. 2019, 10, 1605. [Google Scholar] [CrossRef] [PubMed]
  97. Yu, Y.-S.; Xu, H.; AboulFotouh, K.; Williams, G.; Suman, J.; Sahakijpijarn, S.; Cano, C.; Warnken, Z.N.; Wu, K.C.W.; Williams, R.O.; et al. Intranasal delivery of thin-film freeze-dried monoclonal antibodies using a powder nasal spray system. Int. J. Pharm. 2024, 653, 123892. [Google Scholar] [CrossRef]
  98. Smith, C.A.; Kulkarni, U.; Chen, J.; Goldstein, D.R. Influenza virus inoculum volume is critical to elucidate age—Dependent mortality in mice. Aging Cell 2019, 18, e12893. [Google Scholar] [CrossRef]
  99. Xu, Y.; Yan, X.; Wei, T.; Chen, M.; Zhu, J.; Gao, J.; Liu, B.; Zhu, W.; Liu, Z. Transmucosal Delivery of Nasal Nanovaccines Enhancing Mucosal and Systemic Immunity. Nano Lett. 2023, 23, 10522–10531. [Google Scholar] [CrossRef]
  100. Xu, H.; Cai, L.; Hufnagel, S.; Cui, Z. Intranasal vaccine: Factors to consider in research and development. Int. J. Pharm. 2021, 609, 121180. [Google Scholar] [CrossRef]
  101. Seefeld, M.L.; Templeton, E.L.; Lehtinen, J.M.; Sinclair, N.; Yadav, D.; Hartwell, B.L. Harnessing the potential of the NALT and BALT as targets for immunomodulation using engineering strategies to enhance mucosal uptake. Front. Immunol. 2024, 15, 1419527. [Google Scholar] [CrossRef]
  102. Meinhardt, J.; Radke, J.; Dittmayer, C.; Franz, J.; Thomas, C.; Mothes, R.; Laue, M.; Schneider, J.; Brünink, S.; Greuel, S.; et al. Olfactory transmucosal SARS-CoV-2 invasion as a port of central nervous system entry in individuals with COVID-19. Nat. Neurosci. 2021, 24, 168–175. [Google Scholar] [CrossRef] [PubMed]
  103. Ezeasor, C.; Shoyinka, S.; Emikpe, B.; Bodjo, C. Intranasal Peste des petits ruminants virus vaccination of goats using Irvingia gabonensis gum as delivery system: Hematological and humoral immune responses. J. Immunoass. Immunochem. 2020, 42, 82–94. [Google Scholar] [CrossRef] [PubMed]
  104. Yang, Y.L.; Kim, J.; Jeong, Y.; Jang, Y.-S. Intranasal immunization with a Middle East respiratory syndrome-coronavirus antigen conjugated to the M-cell targeting ligand Co4B enhances antigen-specific mucosal and systemic immunity and protects against infection. Vaccine 2022, 40, 714–725. [Google Scholar] [CrossRef]
  105. Kraus, A.; Huertas, M.; Ellis, L.; Boudinot, P.; Levraud, J.-P.; Salinas, I. Intranasal delivery of SARS-CoV-2 spike protein is sufficient to cause olfactory damage, inflammation and olfactory dysfunction in zebrafish. Brain Behav. Immun. 2022, 102, 341–359. [Google Scholar] [CrossRef]
  106. Neary, M.; Sharp, J.; Gallardo-Toledo, E.; Herriott, J.; Kijak, E.; Bramwell, C.; Cox, H.; Tatham, L.; Box, H.; Curley, P.; et al. Evaluation of Nafamostat as Chemoprophylaxis for SARS-CoV-2 Infection in Hamsters. Viruses 2023, 15, 1744. [Google Scholar] [CrossRef] [PubMed]
  107. Jearanaiwitayakul, T.; Seesen, M.; Chawengkirttikul, R.; Limthongkul, J.; Apichirapokey, S.; Sapsutthipas, S.; Phumiamorn, S.; Sunintaboon, P.; Ubol, S. Intranasal Administration of RBD Nanoparticles Confers Induction of Mucosal and Systemic Immunity against SARS-CoV-2. Vaccines 2021, 9, 768. [Google Scholar] [CrossRef]
  108. Teng, Z.; Meng, L.-Y.; Yang, J.-K.; He, Z.; Chen, X.-G.; Liu, Y. Bridging nanoplatform and vaccine delivery, a landscape of strategy to enhance nasal immunity. J. Control. Release 2022, 351, 456–475. [Google Scholar] [CrossRef]
  109. Alzhrani, R.F.; Xu, H.; Valdes, S.A.; Cui, Z. Intranasal delivery of a nicotine vaccine candidate induces antibodies in mouse blood and lung mucosal secretions that specifically neutralize nicotine. Drug Dev. Ind. Pharm. 2020, 46, 1656–1664. [Google Scholar] [CrossRef]
  110. Yi, E.-J.; Kim, Y.-I.; Song, J.-H.; Ko, H.-J.; Chang, S.-Y. Intranasal immunization with curdlan induce Th17 responses and enhance protection against enterovirus 71. Vaccine 2023, 41, 2243–2252. [Google Scholar] [CrossRef]
  111. Tolman, L.E.; Yates, J.L.; Rong, Y.; Reynolds-Peterson, C.; Ehrbar, D.; Torres-Velez, F.J.; Mantis, N.J. Durable Immunity to Ricin Toxin Elicited by Intranasally Administered Monoclonal Antibody–Based Immune Complexes. ImmunoHorizons 2022, 6, 324–333. [Google Scholar] [CrossRef]
  112. Ryu, J.-Y.; Cerecedo-Lopez, C.; Yang, H.; Ryu, I.; Du, R. Brain-targeted intranasal delivery of protein-based gene therapy for treatment of ischemic stroke. Theranostics 2024, 14, 4773–4786. [Google Scholar] [CrossRef] [PubMed]
  113. Ha, J.; Kim, M.; Lee, Y.; Lee, M. Intranasal delivery of self-assembled nanoparticles of therapeutic peptides and antagomirs elicits anti-tumor effects in an intracranial glioblastoma model. Nanoscale 2021, 13, 14745–14759. [Google Scholar] [CrossRef] [PubMed]
  114. Aly, A.E.E.; Caron, N.S.; Black, H.F.; Schmidt, M.E.; Anderson, C.; Ko, S.; Baddeley, H.J.E.; Anderson, L.; Casal, L.L.; Rahavi, R.S.M.; et al. Delivery of mutant huntingtin-lowering antisense oligonucleotides to the brain by intranasally administered apolipoprotein A-I nanodisks. J. Control. Release 2023, 360, 913–927. [Google Scholar] [CrossRef]
  115. Petkova, A.I.; Kubajewska, I.; Vaideanu, A.; Schätzlein, A.G.; Uchegbu, I.F. Gene Targeting to the Cerebral Cortex Following Intranasal Administration of Polyplexes. Pharmaceutics 2022, 14, 1136. [Google Scholar] [CrossRef] [PubMed]
  116. Guo, S.; Perets, N.; Betzer, O.; Ben-Shaul, S.; Sheinin, A.; Michaelevski, I.; Popovtzer, R.; Offen, D.; Levenberg, S. Intranasal Delivery of Mesenchymal Stem Cell Derived Exosomes Loaded with Phosphatase and Tensin Homolog siRNA Repairs Complete Spinal Cord Injury. ACS Nano 2019, 13, 10015–10028. [Google Scholar] [CrossRef]
  117. Yang, Y.; Zhang, X.; Wu, S.; Zhang, R.; Zhou, B.; Zhang, X.; Tang, L.; Tian, Y.; Men, K.; Yang, L. Enhanced nose-to-brain delivery of siRNA using hyaluronan-enveloped nanomicelles for glioma therapy. J Control. Release 2022, 342, 66–80. [Google Scholar] [CrossRef]
  118. Dhaliwal, H.K.; Fan, Y.; Kim, J.; Amiji, M.M. Intranasal Delivery and Transfection of mRNA Therapeutics in the Brain Using Cationic Liposomes. Mol. Pharm. 2020, 17, 1996–2005. [Google Scholar] [CrossRef]
  119. Lee, D.; Minko, T. Nanotherapeutics for Nose-to-Brain Drug Delivery: An Approach to Bypass the Blood Brain Barrier. Pharmaceutics 2021, 13, 2049. [Google Scholar] [CrossRef]
  120. Kronner, J.M.; Folbe, A.; Meythaler, J.; Nelson, J.O.; Borisov, A.; Peduzzi, J.D. Intranasally Applied Human Olfactory Mucosa Neural Progenitor Cells Migrate to Damaged Brain Regions. Future Sci. OA 2022, 8, FSO806. [Google Scholar] [CrossRef]
  121. Lu, S.; Li, K.; Yang, Y.; Wang, Q.; Yu, Y.; Wang, Z.; Luan, Z. Optimization of an Intranasal Route for the Delivery of Human Neural Stem Cells to Treat a Neonatal Hypoxic-Ischemic Brain Injury Rat Model. Neuropsychiatr. Dis. Treat. 2022, 18, 413–426. [Google Scholar] [CrossRef] [PubMed]
  122. Lu, M.-H.; Ji, W.-L.; Chen, H.; Sun, Y.-Y.; Zhao, X.-Y.; Wang, F.; Shi, Y.; Hu, Y.-N.; Liu, B.-X.; Wu, J.-W.; et al. Intranasal Transplantation of Human Neural Stem Cells Ameliorates Alzheimer’s Disease-Like Pathology in a Mouse Model. Front. Aging Neurosci. 2021, 13, 650103. [Google Scholar] [CrossRef]
  123. Tang, Y.; Han, L.; Bai, X.; Liang, X.; Zhao, J.; Huang, F.; Wang, J. Intranasal Delivery of Bone Marrow Stromal Cells Preconditioned with Fasudil to Treat a Mouse Model of Parkinson’s Disease. Neuropsychiatr. Dis. Treat. 2020, 16, 249–262. [Google Scholar] [CrossRef]
  124. Simon, C.; Gan, Q.F.; Kathivaloo, P.; Mohamad, N.A.; Dhamodharan, J.; Krishnan, A.; Sengodan, B.; Palanimuthu, V.R.; Marimuthu, K.; Rajandas, H.; et al. Deciduous DPSCs Ameliorate MPTP-Mediated Neurotoxicity, Sensorimotor Coordination and Olfactory Function in Parkinsonian Mice. Int. J. Mol. Sci. 2019, 20, 568. [Google Scholar] [CrossRef]
  125. Moreira, A.; Winter, C.; Joy, J.; Winter, L.; Jones, M.; Noronha, M.; Porter, M.; Quim, K.; Corral, A.; Alayli, Y.; et al. Intranasal delivery of human umbilical cord Wharton’s jelly mesenchymal stromal cells restores lung alveolarization and vascularization in experimental bronchopulmonary dysplasia. Stem Cells Transl. Med. 2020, 9, 221–234. [Google Scholar] [CrossRef] [PubMed]
  126. Hay, A.J.D.; Latham, A.S.; Mumford, G.; Hines, A.D.; Risen, S.; Gordon, E.; Siebenaler, C.; Gilberto, V.S.; Zabel, M.D.; Moreno, J.A. Intranasally delivered mesenchymal stromal cells decrease glial inflammation early in prion disease. Front. Neurosci. 2023, 17, 1158408. [Google Scholar] [CrossRef]
  127. Yu, S.P.; Gu, X.; Chau, M.J.; Deveau, T.C.; Kim, Y.S.; Wei, L.; Xu, Y. Delayed and repeated intranasal delivery of bone marrow stromal cells increases regeneration and functional recovery after ischemic stroke in mice. BMC Neurosci. 2018, 19, 20. [Google Scholar] [CrossRef]
  128. Parvez, S.; A Puchowicz, M.; Parveen, K.; Stayton, A.S.; Bajwa, A.; Salman, M.; Parveen, A.; Ishrat, T. Intranasal Delivery of Mitochondria Attenuates Brain Injury by AMPK and SIRT1/PGC-1α Pathways in a Murine Model of Photothrombotic Stroke. Mol. Neurobiol. 2023, 61, 2822–2838. [Google Scholar] [CrossRef]
  129. Price, T.J.; Alexander, J.F.; Heijnen, C.J.; Wangzhou, A.; Ray, P.R.; Arroyo, L.D.; Seua, A.V.; Heiβ-Lückemann, L.; Schedlowski, M.; Kavelaars, A. Nasal administration of mitochondria reverses chemotherapy-induced cognitive deficits. Theranostics 2021, 11, 3109–3130. [Google Scholar] [CrossRef]
  130. Sánchez, S.V.; Otavalo, G.N.; Gazeau, F.; Silva, A.K.A.; Morales, J.O. Intranasal delivery of extracellular vesicles: A promising new approach for treating neurological and respiratory disorders. J. Control. Release 2025, 379, 489–523. [Google Scholar] [CrossRef]
  131. Jiang, Y.; Wang, N.; Liu, J.; Ren, H.; Jiang, W.; Lei, Y.; Fu, X.; Hao, M.; Lang, X.; Liu, Y.; et al. Intranasal delivery of hMSC-derived supernatant for treatment of ischemic stroke by inhibiting the pro-inflammatory polarization of neutrophils. Stem Cell Res. Ther. 2025, 16, 43. [Google Scholar] [CrossRef]
  132. Ikeda, T.; Kawabori, M.; Zheng, Y.; Yamaguchi, S.; Gotoh, S.; Nakahara, Y.; Yoshie, E.; Fujimura, M. Intranasal Administration of Mesenchymal Stem Cell-Derived Exosome Alleviates Hypoxic-Ischemic Brain Injury. Pharmaceutics 2024, 16, 446. [Google Scholar] [CrossRef] [PubMed]
  133. El-Nabarawy, N.A.; Teaima, M.H.; Helal, D.A. Assessment of Spanlastic Vesicles of Zolmitriptan for Treating Migraine In Rats. Drug Des. Dev. Ther. 2019, 13, 3929–3937. [Google Scholar] [CrossRef] [PubMed]
  134. Kouhjani, M.; Jaafari, M.R.; Saberi, A.; Gholami, L.; Tafaghodi, M. Nose to brain delivery of insulin loaded in PLGA and chitosan-coated PLGA nanoparticles: A promising approach for Alzheimer’s disease therapy. J. Drug Deliv. Sci. Technol. 2025, 108, 106857. [Google Scholar] [CrossRef]
  135. Alves, G.; Falcão, A.; Meirinho, S.; Rodrigues, M.; Santos, A.O. Self-Emulsifying Drug Delivery Systems: An Alternative Approach to Improve Brain Bioavailability of Poorly Water-Soluble Drugs through Intranasal Administration. Pharmaceutics 2022, 14, 1487. [Google Scholar] [CrossRef]
  136. Lim, C.; Koo, J.; Oh, K.T. Recent Advances in Intranasal Administration for Brain-Targeting Delivery: A Comprehensive Review of Lipid-Based Nanoparticles and Stimuli-Responsive Gel Formulations. Int. J. Nanomed. 2024, 19, 1767–1807. [Google Scholar] [CrossRef]
  137. Shah, P.; Lalan, M.; Barve, K. Intranasal delivery: An attractive route for the administration of nucleic acid based therapeutics for CNS disorders. Front Pharmacol. 2022, 13, 974666. [Google Scholar] [CrossRef]
  138. Xu, J.; Hsu, S.-H. Self-healing hydrogel as an injectable implant: Translation in brain diseases. J. Biomed. Sci. 2023, 30, 43. [Google Scholar] [CrossRef]
  139. Zhao, W.; Yu, F.; Sun, K.; Zheng, X.; Jin, H. Exosomes as CNS Drug Delivery Tools and Their Applications. Pharmaceutics 2022, 14, 2252. [Google Scholar] [CrossRef]
  140. Shi, B.; Liu, Y.; Zheng, M.; Rehman, F.U. Exosomes based strategies for brain drug delivery. Biomaterials 2022, 293, 121949. [Google Scholar] [CrossRef]
  141. Huang, Y.; Qin, S.; Liu, J.; Wen, Y.; Li, C.; Zhao, W. Overcoming the blood-brain barrier: Exosomes as theranostic nanocarriers for precision neuroimaging. Release 2022, 349, 902–916. [Google Scholar] [CrossRef]
  142. Weinberg, M.S.; Samulski, R.J.; McCown, T.J. Adeno-associated virus (AAV) gene therapy for neurological disease. Neuropharmacology 2013, 69, 82–88. [Google Scholar] [CrossRef]
  143. Grimm, D.; Gadenstaetter, A.J.; Schmutzler, L.; Landegger, L.D. Intranasal application of adeno-associated viruses: A systematic review. Transl. Res. 2022, 248, 87–110. [Google Scholar] [CrossRef]
  144. Price, R.J.; Mead, B.P.; Timbie, K.F. Drug and gene delivery across the blood-brain barrier with focused ultrasound. J. Control. Release 2015, 219, 61–75. [Google Scholar] [CrossRef]
  145. Hafner, A. Advances in Development, Characterisation and Application of Nasal Drug Delivery Systems. Pharmaceutics 2022, 14, 1562. [Google Scholar] [CrossRef]
  146. Kashyap, K.; Shukla, R. Drug Delivery and Targeting to the Brain Through Nasal Route: Mechanisms, Applications and Challenges. Curr. Drug Deliv. 2019, 16, 887–901. [Google Scholar] [CrossRef] [PubMed]
  147. Herzog, H.; Glöckler, S.; Flamm, J.; Ladel, S.; Maigler, F.; Pitzer, C.; Schindowski, K. Intranasal Nose-to-Brain Drug Delivery via the Olfactory Region in Mice: Two In-Depth Protocols for Region-Specific Intranasal Application of Antibodies and for Expression Analysis of Fc Receptors via In Situ Hybridization in the Nasal Mucosa. In Tau Protein; Methods in Molecular Biology; Springer: New York, NY, USA, 2024; pp. 387–410. [Google Scholar]
  148. Chen, J.; Finlay, W.H.; Vehring, R.; Martin, A.R. Characterizing regional drug delivery within the nasal airways. Expert Opin. Drug Deliv. 2024, 21, 537–551. [Google Scholar] [CrossRef]
  149. Flamm, J.; Hartung, S.; Gänger, S.; Maigler, F.; Pitzer, C.; Schindowski, K. Establishment of an Olfactory Region-specific Intranasal Delivery Technique in Mice to Target the Central Nervous System. Front. Pharmacol. 2022, 12, 789780. [Google Scholar] [CrossRef]
  150. Simón, J.A.; Utomo, E.; Pareja, F.; Collantes, M.; Quincoces, G.; Otero, A.; Ecay, M.; Domínguez-Robles, J.; Larrañeta, E.; Peñuelas, I. Radiolabeled Risperidone microSPECT/CT Imaging for Intranasal Implant Studies Development. Pharmaceutics 2023, 15, 843. [Google Scholar] [CrossRef]
  151. Ye, D.; Chen, H. Focused ultrasound-mediated intranasal brain drug delivery technique (FUSIN). MethodsX 2021, 8, 101266. [Google Scholar] [CrossRef]
  152. Ye, D.; Luan, J.; Pang, H.; Yang, Y.; Nazeri, A.; Rubin, J.B.; Chen, H. Characterization of focused ultrasound-mediated brainstem delivery of intranasally administered agents. J. Control. Release 2020, 328, 276–285. [Google Scholar] [CrossRef] [PubMed]
  153. Ye, D.; Yuan, J.; Yang, Y.; Yue, Y.; Hu, Z.; Fadera, S.; Chen, H. Incisionless targeted adeno-associated viral vector delivery to the brain by focused ultrasound-mediated intranasal administration. eBioMedicine 2022, 84, 104277. [Google Scholar] [CrossRef] [PubMed]
  154. Jeon, S.; Park, S.H.; Kim, E.; Kim, J.y.; Kim, S.W.; Choi, H. A Magnetically Powered Stem Cell—Based Microrobot for Minimally Invasive Stem Cell Delivery via the Intranasal Pathway in a Mouse Brain. Adv. Healthc. Mater. 2021, 10, 2100801. [Google Scholar] [CrossRef]
  155. Wang, J.; Liao, Z.-X. Research progress of microrobots in tumor drug delivery. Food Med. Homol. 2024, 1, 9420025. [Google Scholar] [CrossRef]
  156. Stenslik, M.; Evans, A.; Pomerleau, F.; Weeks, R.; Huettl, P.; Foreman, E.; Turchan-Cholewo, J.; Andersen, A.; Cass, W.; Zhang, Z.; et al. Methodology and effects of repeated intranasal delivery of DNSP-11 in awake Rhesus macaques. J. Neurosci. Methods 2018, 303, 30–40. [Google Scholar] [CrossRef] [PubMed]
  157. Wingrove, J.; Swedrowska, M.; Scherließ, R.; Parry, M.; Ramjeeawon, M.; Taylor, D.; Gauthier, G.; Brown, L.; Amiel, S.; Zelaya, F.; et al. Characterisation of nasal devices for delivery of insulin to the brain and evaluation in humans using functional magnetic resonance imaging. J. Control. Release 2019, 302, 140–147. [Google Scholar] [CrossRef]
  158. Ye, E.; Park, E.; Kim, E.; Lee, J.E.; Yang, S.H.; Park, S.-M. Transcranial application of magnetic pulses for improving brain drug delivery efficiency via intranasal injection of magnetic nanoparticles. Biomed. Eng. Lett. 2023, 13, 417–427. [Google Scholar] [CrossRef]
  159. Hong, S.-S.; Oh, K.T.; Choi, H.-G.; Lim, S.-J. Liposomal Formulations for Nose-to-Brain Delivery: Recent Advances and Future Perspectives. Pharmaceutics 2019, 11, 540. [Google Scholar] [CrossRef]
  160. Sawarkar, S.P.; Tanna, V.; Ravikumar, P. Exploring Nose to Brain Nano Delivery for Effective Management of Migraine. Curr. Drug Deliv. 2023, 20, 144–157. [Google Scholar] [CrossRef]
  161. Bai, J.; Wei, Y.-S.; He, Y.; Zhang, H.; Ma, X. Preparation and biological activity of polysaccharide metal ion complex and feasibility analysis of Tremella fuciformis polysaccharide with metal ions. Food Med. Homol. 2025, 2, 9420031. [Google Scholar] [CrossRef]
  162. Nasr, M.; Wahdan, S.A. Neuroprotective effects of novel nanosystems simultaneously loaded with vinpocetine and piracetam after intranasal administration. Life Sci. 2019, 226, 117–129. [Google Scholar] [CrossRef] [PubMed]
  163. Salem, H.F.; Aboud, H.M.; Abdellatif, M.M.; Abou-Taleb, H.A. Nose-to-Brain Targeted Delivery of Donepezil Hydrochloride via Novel Hyaluronic Acid-Doped Nanotransfersomes for Alzheimer’s Disease Mitigation. J. Pharm. Sci. 2024, 113, 1934–1945. [Google Scholar] [CrossRef] [PubMed]
  164. Tyagi, A.; Gabrani, R.; Nigam, K.; Kaur, A.; Nematullah; Khan, F.; Dang, S. Nose-to-brain delivery of lamotrigine-loaded PLGA nanoparticles. Drug Deliv. Transl. Res. 2019, 9, 879–890. [Google Scholar] [CrossRef]
  165. Wasan, E.K.; Cuddihy, G.; Rai, R.; Aibani, N.; Patel, P. Chitosan Nanoparticles at the Biological Interface: Implications for Drug Delivery. Pharmaceutics 2021, 13, 1686. [Google Scholar] [CrossRef]
  166. Walgrave, H.; Zhou, L.; De Strooper, B.; Salta, E. The promise of microRNA-based therapies in Alzheimer’s disease: Challenges and perspectives. Mol. Neurodegener. 2021, 16, 76. [Google Scholar] [CrossRef]
  167. Oppong-Damoah, A.; Zaman, R.U.; D’Souza, M.J.; Murnane, K.S. Nanoparticle encapsulation increases the brain penetrance and duration of action of intranasal oxytocin. Horm. Behav. 2019, 108, 20–29. [Google Scholar] [CrossRef]
  168. Pomatto, M.A.C.; Gai, C.; Negro, F.; Massari, L.; Deregibus, M.C.; Grange, C.; De Rosa, F.G.; Camussi, G. Plant-Derived Extracellular Vesicles as a Delivery Platform for RNA-Based Vaccine: Feasibility Study of an Oral and Intranasal SARS-CoV-2 Vaccine. Pharmaceutics 2023, 15, 974. [Google Scholar] [CrossRef] [PubMed]
  169. Gupta, I.; Adin, S.N.; Aqil, M.; Mujeeb, M. Nose to brain delivery of naringin loaded transniosomes for epilepsy: Formulation, characterisation, blood-brain distribution and invivo pharmacodynamic evaluation. J. Liposome Res. 2023, 34, 60–76. [Google Scholar] [CrossRef]
  170. Tang, L.; Zhang, R.; Wang, Y.; Zhang, X.; Yang, Y.; Zhao, B.; Yang, L. A simple self-assembly nanomicelle based on brain tumor-targeting peptide-mediated siRNA delivery for glioma immunotherapy via intranasal administration. Acta Biomater. 2023, 155, 521–537. [Google Scholar] [CrossRef]
  171. Xu, Y.; Jiang, X.; Zhou, Z.; Ferguson, T.; Obliosca, J.; Luo, C.C.; Chan, K.W.; Kong, X.P.; Tison, C.K. Mucosal Delivery of HIV-1 Glycoprotein Vaccine Candidate Enabled by Short Carbon Nanotubes. Part. Part. Syst. Charact. 2022, 39, 2200011. [Google Scholar] [CrossRef]
  172. Hua, T.; Li, S.; Han, B. Nanomedicines for intranasal delivery: Understanding the nano-bio interactions at the nasal mucus-mucosal barrier. Expert Opin. Drug Deliv. 2024, 21, 553–572. [Google Scholar] [CrossRef] [PubMed]
  173. Ramot, Y.; Rottenberg, Y.; Domb, A.J.; Kubek, M.J.; Williams, K.D.; Nyska, A. Preclinical In-Vivo Safety of a Novel Thyrotropin-Releasing Hormone-Loaded Biodegradable Nanoparticles After Intranasal Administration in Rats and Primates. Int. J. Toxicol. 2023, 42, 334–344. [Google Scholar] [CrossRef] [PubMed]
  174. Allegritti, E.; Battista, S.; Maggi, M.A.; Marconi, C.; Galantini, L.; Giansanti, L. Novel liposomal formulations for protection and delivery of levodopa: Structure-properties correlation. Int. J. Pharm. 2023, 643, 123230. [Google Scholar] [CrossRef]
  175. Elsheikh, M.A.; El-Feky, Y.A.; Al-Sawahli, M.M.; Ali, M.E.; Fayez, A.M.; Abbas, H. A Brain-Targeted Approach to Ameliorate Memory Disorders in a Sporadic Alzheimer’s Disease Mouse Model via Intranasal Luteolin-Loaded Nanobilosomes. Pharmaceutics 2022, 14, 576. [Google Scholar] [CrossRef]
  176. Soligo, M.; Felsani, F.M.; Da Ros, T.; Bosi, S.; Pellizzoni, E.; Bruni, S.; Isopi, J.; Marcaccio, M.; Manni, L.; Fiorito, S. Distribution in the brain and possible neuroprotective effects of intranasally delivered multi-walled carbon nanotubes. Nanoscale Adv. 2021, 3, 418–431. [Google Scholar] [CrossRef]
  177. Hayes, S.H.; Liu, Q.; Selvakumaran, S.; Haney, M.J.; Batrakova, E.V.; Allman, B.L.; Walton, P.A.; Kiser, P.; Whitehead, S.N. Brain Targeting and Toxicological Assessment of the Extracellular Vesicle-Packaged Antioxidant Catalase-SKL Following Intranasal Administration in Mice. Neurotox. Res. 2021, 39, 1418–1429. [Google Scholar] [CrossRef]
  178. Hao, R.; Sun, B.; Yang, L.; Ma, C.; Li, S. RVG29-modified microRNA-loaded nanoparticles improve ischemic brain injury by nasal delivery. Drug Deliv. 2020, 27, 772–781. [Google Scholar] [CrossRef]
  179. Liao, K.; Niu, F.; Dagur, R.S.; He, M.; Tian, C.; Hu, G. Intranasal Delivery of lincRNA-Cox2 siRNA Loaded Extracellular Vesicles Decreases Lipopolysaccharide-Induced Microglial Proliferation in Mice. J. Neuroimmune Pharmacol. 2019, 15, 390–399. [Google Scholar] [CrossRef] [PubMed]
  180. Su, Y.; Sun, B.; Gao, X.; Dong, X.; Fu, L.; Zhang, Y.; Li, Z.; Wang, Y.; Jiang, H.; Han, B. Intranasal Delivery of Targeted Nanoparticles Loaded With miR-132 to Brain for the Treatment of Neurodegenerative Diseases. Front. Pharmacol. 2020, 11, 1165. [Google Scholar] [CrossRef]
  181. Diao, Y.; Wu, X.; Zang, R.; Liu, M.; Wei, S.; Yang, N.; Qiu, Y.; Xu, X. Self-Assembly of Rhein and Matrine Nanoparticles for Enhanced Wound Healing. Molecules 2024, 29, 3326. [Google Scholar] [CrossRef]
  182. Miao, Y.-H.; Wang, X.; Zhao, X.-M.; Hu, Y.-W.; Liu, X.; Deng, D.-W. Co-assembly strategies of natural plant compounds for improving their bioavailability. Food Med. Homol. 2025, 2, 9420022. [Google Scholar] [CrossRef]
  183. Ali, M.M.; Shoukri, R.A.; Yousry, C. Thin film hydration versus modified spraying technique to fabricate intranasal spanlastic nanovesicles for rasagiline mesylate brain delivery: Characterization, statistical optimization, and in vivo pharmacokinetic evaluation. Drug Deliv. Transl. Res. 2022, 13, 1153–1168. [Google Scholar] [CrossRef] [PubMed]
  184. Kadakia, E.; Bottino, D.; Amiji, M. Mathematical Modeling and Simulation to Investigate the CNS Transport Characteristics of Nanoemulsion-Based Drug Delivery Following Intranasal Administration. Pharm. Res. 2019, 36, 75. [Google Scholar] [CrossRef]
  185. Sipos, B.; Csóka, I.; Szivacski, N.; Budai-Szűcs, M.; Schelcz, Z.; Zupkó, I.; Szabó-Révész, P.; Volk, B.; Katona, G. Mucoadhesive meloxicam-loaded nanoemulsions: Development, characterization and nasal applicability studies. Eur. J. Pharm. Sci. 2022, 175, 106229. [Google Scholar] [CrossRef]
  186. Pailla, S.; Sampathi, S.; Junnuthula, V.; Maddukuri, S.; Dodoala, S.; Dyawanapelly, S. Brain-Targeted Intranasal Delivery of Zotepine Microemulsion: Pharmacokinetics and Pharmacodynamics. Pharmaceutics 2022, 14, 978. [Google Scholar] [CrossRef] [PubMed]
  187. Kaur, A.; Nigam, K.; Srivastava, S.; Tyagi, A.; Dang, S. Memantine nanoemulsion: A new approach to treat Alzheimer’s disease. J. Microencapsul. 2020, 37, 355–365. [Google Scholar] [CrossRef]
  188. Khunt, D.; Shrivas, M.; Polaka, S.; Gondaliya, P.; Misra, M. Role of Omega-3 Fatty Acids and Butter Oil in Targeting Delivery of Donepezil Hydrochloride Microemulsion to Brain via the Intranasal Route: A Comparative Study. AAPS PharmSciTech 2020, 21, 45. [Google Scholar] [CrossRef]
  189. Gaba, B.; Khan, T.; Haider, M.F.; Alam, T.; Baboota, S.; Parvez, S.; Ali, J. Vitamin E Loaded Naringenin Nanoemulsion via Intranasal Delivery for the Management of Oxidative Stress in a 6-OHDA Parkinson’s Disease Model. BioMed Res. Int. 2019, 2019, 2382563. [Google Scholar] [CrossRef]
  190. Jiang, Y.; Liu, C.; Zhai, W.; Zhuang, N.; Han, T.; Ding, Z. The Optimization Design Of Lactoferrin Loaded HupA Nanoemulsion For Targeted Drug Transport Via Intranasal Route. Int. J. Nanomed. 2019, 14, 9217–9234. [Google Scholar] [CrossRef]
  191. Pragya; Bisht, S.; Parashar, P. Nanotechnology-driven Microemulsion Based Intranasal Delivery to Neurotechnology-driven Neuralink: Strategies to Improve Management of Neurodegenerative Disorders. AAPS PharmSciTech 2024, 25, 215. [Google Scholar] [CrossRef]
  192. Liu, N.; Yang, C.; Liang, X.; Cao, K.; Xie, J.; Luo, Q.; Luo, H. Mesoporous silica nanoparticle-encapsulated Bifidobacterium attenuates brain Aβ burden and improves olfactory dysfunction of APP/PS1 mice by nasal delivery. J. Nanobiotechnology 2022, 20, 439. [Google Scholar] [CrossRef] [PubMed]
  193. Latif, R.; Makar, R.R.; Hosni, E.A.; El Gazayerly, O.N. The potential of intranasal delivery of nanocrystals in powder form on the improvement of zaleplon performance: In-vitro, in-vivo assessment. Drug Dev. Ind. Pharm. 2021, 47, 268–279. [Google Scholar] [CrossRef] [PubMed]
  194. Wang, Q.-S.; Li, K.; Gao, L.-N.; Zhang, Y.; Lin, K.-M.; Cui, Y.-L. Intranasal delivery of berberine via in situ thermoresponsive hydrogels with non-invasive therapy exhibits better antidepressant-like effects. Biomater. Sci. 2020, 8, 2853–2865. [Google Scholar] [CrossRef]
  195. Salem, H.F.; Ali, A.A.; Hegazy, A.M.; Sadek, A.-R.A.; Aboud, H.M. Harnessing of Doxylamine Succinate/Pyridoxine Hydrochloride-Dual Laden Bilosomes as a Novel Combinatorial Nanoparadigm for Intranasal Delivery: In Vitro Optimization and In Vivo Pharmacokinetic Appraisal. J. Pharm. Sci. 2022, 111, 794–809. [Google Scholar] [CrossRef]
  196. Xu, D.; Qiao, T.; Wang, Y.; Wang, Q.-S.; Cui, Y.-L. Alginate nanogels-based thermosensitive hydrogel to improve antidepressant-like effects of albiflorin via intranasal delivery. Drug Deliv. 2021, 28, 2137–2149. [Google Scholar] [CrossRef] [PubMed]
  197. Wang, J.; Tan, Y.; Dai, Y.; Hu, K.; Tan, X.; Jiang, S.; Li, G.; Zhang, X.; Kang, L.; Wang, X.; et al. Intranasal Delivery of Endothelial Cell-Derived Extracellular Vesicles with Supramolecular Gel Attenuates Myocardial Ischemia-Reperfusion Injury. Int. J. Nanomed. 2023, 18, 5495–5510. [Google Scholar] [CrossRef]
  198. Liu, L.; Hao, H.; Wu, X.; Li, P.; Ye, H.; Zheng, Q.; Yang, H. Protocatechuic aldehyde protects cardiomycoytes against ischemic injury via regulation of nuclear pyruvate kinase M2. Acta Pharm. Sin. B 2021, 11, 3553–3566. [Google Scholar] [CrossRef]
  199. Liu, L.; Hao, H.; Ye, H.; Wu, X.; Yang, H.; Li, P.; Zheng, Q. Dihydrotanshinone I preconditions myocardium against ischemic injury via PKM2 glutathionylation sensitive to ROS. Acta Pharm. Sin. B 2023, 13, 113–127. [Google Scholar] [CrossRef]
  200. Mohanty, D.; Alsaidan, O.A.; Zafar, A.; Dodle, T.; Gupta, J.K.; Yasir, M.; Mohanty, A.; Khalid, M. Development of Atomoxetine-Loaded NLC In Situ Gel for Nose-to-Brain Delivery: Optimization, In Vitro, and Preclinical Evaluation. Pharmaceutics 2023, 15, 1985. [Google Scholar] [CrossRef]
  201. Popescu, R.; Dinu-Pîrvu, C.-E.; Ghica, M.V.; Anuța, V.; Popa, L. Physico-Chemical Characterization and Initial Evaluation of Carboxymethyl Chitosan–Hyaluronan Hydrocolloid Systems with Insulin Intended for Intranasal Administration. Int. J. Mol. Sci. 2024, 25, 10452. [Google Scholar] [CrossRef]
  202. Wang, J.T.W.; Rodrigo, A.C.; Patterson, A.K.; Hawkins, K.; Aly, M.M.S.; Sun, J.; Al Jamal, K.T.; Smith, D.K. Enhanced Delivery of Neuroactive Drugs via Nasal Delivery with a Self-Healing Supramolecular Gel. Adv. Sci. 2021, 8, 2101058. [Google Scholar] [CrossRef] [PubMed]
  203. Qu, Y.; Li, A.; Ma, L.; Iqbal, S.; Sun, X.; Ma, W.; Li, C.; Zheng, D.; Xu, Z.; Zhao, Z.; et al. Nose-to-brain delivery of disulfiram nanoemulsion in situ gel formulation for glioblastoma targeting therapy. Int. J. Pharm. 2021, 597, 120250. [Google Scholar] [CrossRef] [PubMed]
  204. Radwan, M.A.; El Khatib, M.M.; Alavi, S.E.; Al Harthi, S.; AlSarra, I.A. Nasal delivery of donepezil HCl-loaded hydrogels for the treatment of Alzheimer’s disease. Sci. Rep. 2019, 9, 9563. [Google Scholar] [CrossRef]
  205. Tan, Y.; Liu, Y.; Liu, Y.; Ma, R.; Luo, J.; Hong, H.; Chen, X.; Wang, S.; Liu, C.; Zhang, Y.; et al. Rational Design of Thermosensitive Hydrogel to Deliver Nanocrystals with Intranasal Administration for Brain Targeting in Parkinson’s Disease. Research 2021, 2021, 9812523. [Google Scholar] [CrossRef]
  206. Mulè, F.; Picone, P.; Biagio, P.L.S.; Dispenza, C.; Amato, A.; Ditta, L.A.; Sabatino, M.A.; Di Carlo, M.; Giacomazza, D. Nose-to-brain delivery of insulin enhanced by a nanogel carrier. J. Control. Release 2018, 270, 23–36. [Google Scholar] [CrossRef]
  207. Ulusoy, S.; Konstantinidis, I.; Passali, D.; Cingi, C.; Negm, H.; Milkov, M.; Muluk, N.B.; Karpischenko, S.; Passali, G.C.; Dilber, M.; et al. Mechanisms and solutions for nasal drug delivery—A narrative review. Eur. Rev. Med. Pharmacol. Sci. 2022, 26, 72–81. [Google Scholar] [CrossRef]
  208. Carrazana, E.; Rabinowicz, A.L.; Madden, S. Optimizing Absorption for Intranasal Delivery of Drugs Targeting the Central Nervous System Using Alkylsaccharide Permeation Enhancers. Pharmaceutics 2023, 15, 2119. [Google Scholar] [CrossRef]
  209. Almahmoud, A.; Parekh, H.S.; Paterson, B.M.; Tupally, K.R.; Vegh, V. Intranasal delivery of imaging agents to the brain. Theranostics 2024, 14, 5022–5101. [Google Scholar] [CrossRef]
  210. Kamei, N.; Suwabe, S.; Arime, K.; Bando, H.; Murata, K.; Yamaguchi, M.; Yokoyama, N.; Tanaka, E.; Hashimoto, A.; Kanazawa, T.; et al. Investigation of the Transport Pathways Associated with Enhanced Brain Delivery of Peptide Drugs by Intranasal Coadministration with Penetratin. Pharmaceutics 2021, 13, 1745. [Google Scholar] [CrossRef]
  211. Clow, F.; Peterken, K.; Pearson, V.; Proft, T.; Radcliff, F.J. PilVax, a novel Lactococcus lactis—Based mucosal vaccine platform, stimulates systemic and mucosal immune responses to Staphylococcus aureus. Immunol. Cell Biol. 2020, 98, 369–381. [Google Scholar] [CrossRef]
  212. Idoudi, S.; Saleh, A.; Akkbik, M.; Amine, L.; Alansari, K.; Rachid, O.; Alkilany, A.M. Investigating Strategies to Enhance the Aqueous Solubility of Ketamine HCl for Intranasal Delivery. Pharmaceutics 2024, 16, 1502. [Google Scholar] [CrossRef]
  213. Prakapenka, A.V.; Peña, V.L.; Strouse, I.; Northup-Smith, S.; Schrier, A.; Ahmed, K.; Bimonte-Nelson, H.A.; Sirianni, R.W. Intranasal 17β-Estradiol Modulates Spatial Learning and Memory in a Rat Model of Surgical Menopause. Pharmaceutics 2020, 12, 1225. [Google Scholar] [CrossRef]
  214. Gerard Lee, S.; Florencio Jr, A.; Sabrina, D.; Akie, O.; Muhammad Fikri Bin Mohd, F.; Ichiro, H.; Shoko, I.; Masanori, K.; Hiroaki, T.; Kenji, S. Enhanced nose-to-brain delivery of tranilast using liquid crystal formulations. J. Control. Release 2020, 325, 1–9. [Google Scholar] [CrossRef]
  215. Kim, N.A.; Thapa, R.; Jeong, S.H.; Bae, H.-d.; Maeng, J.; Lee, K.; Park, K. Enhanced intranasal insulin delivery by formulations and tumor protein-derived protein transduction domain as an absorption enhancer. J. Control. Release 2019, 294, 226–236. [Google Scholar] [CrossRef]
  216. Fukakusa, S.; Suzuki, C.; Sasaki, K.; Sonoda, Y.; Hatano, Y.; Haruta, S.; Magata, Y. Brain drug delivery from the nasal olfactory region is enhanced using lauroylcholine chloride: An estimation using in vivo PET imaging. Nucl. Med. Biol. 2024, 138–139, 108968. [Google Scholar] [CrossRef] [PubMed]
  217. Maeng, J.; Lee, K. Systemic and brain delivery of antidiabetic peptides through nasal administration using cell-penetrating peptides. Front Pharmacol. 2022, 13, 1068495. [Google Scholar] [CrossRef] [PubMed]
  218. Wells, J.M.; Mercenier, A. Mucosal delivery of therapeutic and prophylactic molecules using lactic acid bacteria. Nat. Rev. Microbiol. 2008, 6, 349–362. [Google Scholar] [CrossRef]
  219. Zhang, H.; Sheng, S.; Li, C.; Bao, X.; Zhao, L.; Chen, J.; Guan, P.; Li, X.; Pan, N.; Liang, Y.; et al. Mucosal immunization with the lung Lactobacillus-derived amphiphilic exopolysaccharide adjuvanted recombinant vaccine improved protection against P. aeruginosa infection. PLoS Pathog. 2024, 20, e1012696. [Google Scholar] [CrossRef]
  220. Marchetti, J.M.; Barcellos, J.P.A.; Lee, R.J.; de Souza, M.C.; Petrilli, R.; Eloy, J.O. Liposomes as carriers of hydrophilic small molecule drugs: Strategies to enhance encapsulation and delivery. Colloids Surf. B Biointerfaces 2014, 123, 345–363. [Google Scholar] [CrossRef]
  221. Shalaby, K.; Abdelgawad, M.A.; Alsaidan, O.A.; Elkomy, M.H.; Gomaa, H.A.M.; Elmowafy, M.; Mostafa, E.M. Polymeric Nanoparticles for Delivery of Natural Bioactive Agents: Recent Advances and Challenges. Polymers 2023, 15, 1123. [Google Scholar] [CrossRef]
  222. Hatton, T.A.; Gupta, A.; Eral, H.B.; Doyle, P.S. Nanoemulsions: Formation, properties and applications. Soft Matter 2016, 12, 2826–2841. [Google Scholar] [CrossRef]
  223. Kurczewska, J.; Dobosz, B. Recent Progress and Challenges Regarding Magnetite-Based Nanoparticles for Targeted Drug Delivery. Appl. Sci. 2024, 14, 1132. [Google Scholar] [CrossRef]
  224. Veronese, F.M.; Pasut, G. PEGylation, successful approach to drug delivery. Drug Discov. Today 2005, 10, 1451–1458. [Google Scholar] [CrossRef] [PubMed]
  225. Shah, B.; Jain, A.; Charbe, N.; Thalla, M.; Immanuel, S.; Kapre, S.; Palakurthi, S.S.; Pasham, S. A comprehensive review of challenges and advances in exosome-based drug delivery systems. Nanoscale Adv. 2024, 6, 5803–5826. [Google Scholar] [CrossRef]
  226. Zelkó, R.; Kazsoki, A.; Kürtösi, B. A Systematic Review on Plant-Derived Extracellular Vesicles as Drug Delivery Systems. Int. J. Mol. Sci. 2024, 25, 7559. [Google Scholar] [CrossRef]
  227. Choi, W.; Kohane, D.S. Hybrid Nanoparticle-Hydrogel Systems for Drug Delivery Depots and Other Biomedical Applications. ACS Nano 2024, 18, 22780–22792. [Google Scholar] [CrossRef]
  228. Frank-Ito, D.O. Olfaction and drug delivery to the human olfactory airspace: Current challenges and recent advances. Expert Opin. Drug Deliv. 2025, 22, 511–524. [Google Scholar] [CrossRef]
  229. Chen, J.; Martin, A.R.; Finlay, W.H. Recent In Vitro and In Silico Advances in the Understanding of Intranasal Drug Delivery. Curr. Pharm. Des. 2021, 27, 1482–1497. [Google Scholar] [CrossRef]
  230. League-Pascual, J.C.; Lester-McCully, C.M.; Shandilya, S.; Ronner, L.; Rodgers, L.; Cruz, R.; Peer, C.J.; Figg, W.D.; Warren, K.E. Plasma and cerebrospinal fluid pharmacokinetics of select chemotherapeutic agents following intranasal delivery in a non-human primate model. J. Neuro-Oncol. 2017, 132, 401–407. [Google Scholar] [CrossRef]
  231. Aderibigbe, B.A.; Naki, T. Chitosan-Based Nanocarriers for Nose to Brain Delivery. Appl. Sci. 2019, 9, 2219. [Google Scholar] [CrossRef]
Figure 1. This illustration shows various central nervous system diseases that can be treated via intranasal drug delivery, including Alzheimer’s disease, Parkinson’s disease, meningitis, ischemic stroke, hemorrhagic stroke, and other unspecified diseases. The areas marked by red stars or indicated by arrows represent the sites of tissue damage or functional loss. (all or parts of the figures were created using Servier Medical Art (https://smart.servier.com/), accessed on 5 April 2025, licensed under CC BY 4.0.).
Figure 1. This illustration shows various central nervous system diseases that can be treated via intranasal drug delivery, including Alzheimer’s disease, Parkinson’s disease, meningitis, ischemic stroke, hemorrhagic stroke, and other unspecified diseases. The areas marked by red stars or indicated by arrows represent the sites of tissue damage or functional loss. (all or parts of the figures were created using Servier Medical Art (https://smart.servier.com/), accessed on 5 April 2025, licensed under CC BY 4.0.).
Pharmaceutics 17 00775 g001
Figure 2. This illustration depicts the mechanisms of intranasal drug delivery. The therapeutics can be transported from the nose to the brain through the olfactory and trigeminal nerves and vascular transport across the BBB. The primary routes for nose-to-brain transport are olfactory and trigeminal nerve pathways (all or parts of the figures were created using Servier Medical Art (https://smart.servier.com/), accessed on 5 April 2025, licensed under CC BY 4.0.).
Figure 2. This illustration depicts the mechanisms of intranasal drug delivery. The therapeutics can be transported from the nose to the brain through the olfactory and trigeminal nerves and vascular transport across the BBB. The primary routes for nose-to-brain transport are olfactory and trigeminal nerve pathways (all or parts of the figures were created using Servier Medical Art (https://smart.servier.com/), accessed on 5 April 2025, licensed under CC BY 4.0.).
Pharmaceutics 17 00775 g002
Figure 3. The diagram illustrates different types of drugs that can be delivered via the intranasal route, including exosome-derived drugs, cell-derived drugs, drug delivery systems, chemical drugs (in the form of solutions or dispersions), biological drugs, and nucleic acid drugs (all or parts of the figures were created using Servier Medical Art (https://smart.servier.com/), accessed on 5 April 2025, licensed under CC BY 4.0.).
Figure 3. The diagram illustrates different types of drugs that can be delivered via the intranasal route, including exosome-derived drugs, cell-derived drugs, drug delivery systems, chemical drugs (in the form of solutions or dispersions), biological drugs, and nucleic acid drugs (all or parts of the figures were created using Servier Medical Art (https://smart.servier.com/), accessed on 5 April 2025, licensed under CC BY 4.0.).
Pharmaceutics 17 00775 g003
Table 1. Types of drugs for intranasal drug delivery.
Table 1. Types of drugs for intranasal drug delivery.
Drug CategorySpecific TypeRelevant Examples
Chemical drugsSolution dispersionSolutions and suspensions
Nanocarriers/polymersLiposomes, nanoparticles, micelles, and hydrogels
Biomacromolecule drugsProtein drugsRecombinant proteins, antibodies, hormones, cytokines, and vaccines
Nucleic acid drugsDNA, RNA, and viruses
Cell-derived drugsCell-derived drugsStem cells, immune cells, and mitochondria
Exosome-derived drugsExosomes from different sources
Table 2. Research progress of intranasal delivery of small molecule drugs.
Table 2. Research progress of intranasal delivery of small molecule drugs.
Drug/
System Name
Application/DiseaseResearch ModelKey FindingsRef.
Mixed Nanoparticle SystemADMouse ModelRapid absorption, effective drug delivery to the brain, and high organ-specific drug concentration.[51]
9-cis Retinoic AcidADAD Transgenic MouseReduces Aβ deposition and improves neuroinflammation and synaptic function.[54]
Fluorobiprofene Microspheres/Soft ParticlesADRat ModelIntranasal powder delivery superior to solution, significant olfactory bulb concentration, and early intervention potential.[64]
LacosamideEpilepsyMouse ModelBetter pharmacokinetics compared to intravenous injection.[52]
Opioids/SedativesPalliative CareClinical StudyRapid onset, good patient tolerance, suitable for late-stage patients unable to take oral drugs.[60]
Dihydrocodeine/MidazolamCommunity Palliative Care (Pain/Agitation)Clinical PracticeEasy to administer, improves patient comfort, and reduces medical delay.[61]
HeparinCOVID-19 PreventionMouse and Human TrialsNo significant toxicity and maintains effective concentration for 12 h.[65]
DexmedetomidineSedation for ExtractionsAnxiety Patient StudyOnset in 30–45 min, lasts 60–75 min, no respiratory suppression, suitable for day surgeries.[55]
MC4R Antagonist HS014Trigeminal NeuralgiaRat ModelSignificant relief of hyperalgesia, upregulation of MC4R protein levels.[56]
KetaminePost-Cesarean Pain ReliefMaternal RCT StudySignificantly reduces postoperative pain and morphine demand, and good tolerance.[57]
IsocyanomethaneEpilepsyMouse ModelRapidly increases seizure threshold and no motor/sedation side effects.[53]
Lidocaine SprayPost-Epidural HeadacheClinical CaseNon-invasive treatment, rapid symptom relief, replaces epidural blood patch.[58]
DexamethasoneNeuroinflammation (e.g., Stroke)Mouse ModelHigher brain concentration, faster onset, and suitable for acute treatment.[62]
Icariin-NGSTH SystemDepressionChronic Stress Rat ModelFaster antidepressant effect and organ-specific drug concentration significantly better than oral administration.[63]
NaltrexoneOpioid Side EffectsRodent ModelAlleviates gastrointestinal and central side effects without affecting analgesic effect.[59]
ChlorpyrifosNeurotoxicityAdult Male MouseHigh doses cause memory impairment, anxiety, and brain oxidative stress.[65]
Table 3. Research progress in intranasal delivery of biologic drugs.
Table 3. Research progress in intranasal delivery of biologic drugs.
Drug/
System Name
Application/
Disease
Research ModelKey FindingsRef.
NGFTBIClinical TrialBypasses the BBB to directly affect brain tissue, reducing systemic side effects.[67,70]
Leukemia Inhibitory Factor (LIF)Mild TBI (mTBI) in ChildrenCD1 MiceAlleviates glial proliferation and axonal damage, improves sensory-motor function, and has no side effects.[71]
InsulinADRat ModelIntranasal insulin rapidly distributes to the brain, improves cognitive function, and optimized formulation reduces systemic side effects.[80]
InsulinADMulticenter Clinical TrialImproves memory performance in patients with mild cognitive impairment or AD.[77]
Insulin (Nanocarrier Technology)CNS Diseases Rat ModelEnhances delivery efficiency with nanotechnology, bypasses the BBB through olfactory or trigeminal nerves, and significantly improves cognitive function.[74,75]
InsulinPDRat ModelDirectly targets the brain and minimizes systemic side effects.[78]
IGF-1Brain IschemiaRat ModelReduces neural damage and inflammation and bypasses the BBB to directly affect the brain.[72]
Oxytocin Intranasal Spray TTA-121ASDRabbit ModelSignificantly higher brain-specific drug concentration compared to Syntocinon, higher concentrations in the prefrontal cortex and cuneus.[84]
Insulin (PTD-Modified Formulation)DiabetesRat ExperimentEnhances absorption with Protein Transduction Domain (PTD) and optimizes intranasal delivery formulation to improve efficacy.[79]
Bicyclic Peptide OL-CTOPMorphine Side Effect AntagonismMouseIntranasal delivery effectively antagonizes morphine’s analgesic and respiratory suppression side effects, demonstrating potential for brain-targeted delivery.[87]
Table 4. Research progress in intranasal delivery of nucleic acid drugs.
Table 4. Research progress in intranasal delivery of nucleic acid drugs.
Drug/System NameApplication/DiseaseResearch ModelKey FindingsReferences
Self-Assembled Antagomir-21/RAP NanoparticlesGlioblastomaMouse ModelEnhanced efficacy with a non-toxic carrier, effectively inhibiting tumor growth.[113]
ApoA-I Nanodisk-Loaded ASO (Antisense Oligonucleotide)Huntington’s DiseaseHD Mouse ModelSingle intranasal delivery significantly reduces mutated Huntington protein (mHTT) levels in the striatum and cortex.[114]
Glycerol Chitosan-DNA Complex (GCP/GCPH)Neurological Diseases (e.g., AD)Mouse ModelGCP targets gene delivery to the cerebral cortex; GCPH (hyaluronidase-coated) enhances brain distribution.[115]
MSC-Exo-loaded PTEN siRNA (Mesenchymal Stem Cell Exosomes)Complete SCIAnimal ModelNon-invasive intranasal delivery promotes functional recovery after SCI.[116]
Table 5. Research progress in intranasal delivery of cell therapy drugs.
Table 5. Research progress in intranasal delivery of cell therapy drugs.
Drug/System NameApplication/
Disease
Research ModelKey FindingsRef.
MitochondriaChemotherapy-induced Cognitive DeficitsMouse ModelIntranasal delivery of mitochondrial-targeted compounds provides neuroprotective effects.[128,129]
Human Olfactory Mucosal Progenitor Cells (OMPCs)Brain InjuryRat Diffuse Axonal Injury ModelOMPCs migrate to the vicinity of damaged neurons and axons via intranasal delivery, supporting non-invasive stem cell therapy.[120]
Human Neural Stem Cells (hNSCs)ADAD Mouse ModelhNSCs survive and differentiate into neurons, reducing β-amyloid plaque accumulation and synapse loss, improving cognitive function.[122]
Bone Marrow-Derived Mesenchymal Stem Cells (BMSCs)PDPD Mouse ModelPre-treated BMSCs enhance efficacy, improving motor function and reducing dopaminergic neuron loss.[123]
Human Umbilical Cord-Derived Mesenchymal Stromal Cells (MSCs)Bronchopulmonary Dysplasia (BPD)Experimental BPD ModelIntranasal delivery of MSCs repairs lung damage caused by BPD with simple methods and clinical potential.[125]
Deciduous Dental Pulp Stem Cells (DPSCs)PDMPTP-induced PD MouseDPSCs improve motor coordination and olfactory function, reducing dopaminergic neuron degeneration.[124]
Delayed Repeated Intranasal Delivery of Bone Marrow Stromal CellsIschemic StrokeMouse Stroke ModelDelayed repeated intranasal delivery promotes regeneration and functional recovery after stroke.[127]
Table 6. Advantages and limitations of nanocarrier types in intranasal drug delivery systems.
Table 6. Advantages and limitations of nanocarrier types in intranasal drug delivery systems.
Nanocarrier TypeAdvantagesLimitationsRef.
LiposomesHigh biocompatibility; good drug loading for hydrophilic drugs.Rapid clearance; low stability.[220]
Polymeric Nanoparticles (PLGA, Chitosan)High drug loading; sustained release; improved stability; versatile surface modification.Potential toxicity; difficulty in large-scale production.[221]
NanoemulsionsEnhanced solubility and permeability of lipophilic drugs.Thermodynamic instability, need stabilizers.[222]
Magnetic NanoparticlesMagnetic targeting, imaging compatibility.Potential safety concerns, complex formulation.[223]
PEGylated NanoparticlesExtended circulation time, improved CNS penetration.Expensive, potential immune response.[224]
ExosomesEndogenous origin; high biocompatibility; excellent penetration across biological barriers.Low production yield; difficulty in drug loading; high cost.[225]
Plant-derived Extracellular Vesicles (EVs)Natural origin; low toxicity; immune modulation potential.Scalability; batch variability.[226]
Hydrogel-nanoparticle hybridsMucoadhesion; biocompatibility; prolonged residence time.Formulation optimization challenges; drug release control.[227]
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

Wu, X.; Zang, R.; Qiu, Y.; Zhang, Y.; Peng, J.; Cheng, Z.; Wei, S.; Liu, M.; Diao, Y. Intranasal Drug Delivery Technology in the Treatment of Central Nervous System Diseases: Challenges, Advances, and Future Research Directions. Pharmaceutics 2025, 17, 775. https://doi.org/10.3390/pharmaceutics17060775

AMA Style

Wu X, Zang R, Qiu Y, Zhang Y, Peng J, Cheng Z, Wei S, Liu M, Diao Y. Intranasal Drug Delivery Technology in the Treatment of Central Nervous System Diseases: Challenges, Advances, and Future Research Directions. Pharmaceutics. 2025; 17(6):775. https://doi.org/10.3390/pharmaceutics17060775

Chicago/Turabian Style

Wu, Xunxun, Ranqing Zang, Yiting Qiu, Yufang Zhang, Junbin Peng, Zhiyun Cheng, Site Wei, Meiyan Liu, and Yong Diao. 2025. "Intranasal Drug Delivery Technology in the Treatment of Central Nervous System Diseases: Challenges, Advances, and Future Research Directions" Pharmaceutics 17, no. 6: 775. https://doi.org/10.3390/pharmaceutics17060775

APA Style

Wu, X., Zang, R., Qiu, Y., Zhang, Y., Peng, J., Cheng, Z., Wei, S., Liu, M., & Diao, Y. (2025). Intranasal Drug Delivery Technology in the Treatment of Central Nervous System Diseases: Challenges, Advances, and Future Research Directions. Pharmaceutics, 17(6), 775. https://doi.org/10.3390/pharmaceutics17060775

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

Article Metrics

Back to TopTop