View Article

Abstract

Alzheimer’s disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and behavioral changes. One of the primary challenges in developing effective treatments for AD is the blood–brain barrier (BBB), a selective, semipermeable membrane that restricts drug entry into the central nervous system. Nanotechnology has emerged as a groundbreaking approach to bypass this barrier, providing innovative solutions for the targeted, efficient, and minimally invasive delivery of therapeutic agents into the brain. This review examines recent advancements in nanoparticle-based drug delivery systems designed to treat AD. Different types of nanocarriers, including metallic nanoparticles, polymeric systems, solid lipid nanoparticles, liposomes, and dendrimers, have shown significant promise in transporting drugs across the BBB. These systems enhance drug bioavailability, allow for controlled release, and help minimize systemic toxicity. By functionalizing nanoparticles with targeting ligands and modifying their surface properties, their ability to penetrate the brain and improve therapeutic outcomes is further optimized. Additionally, green-synthesized nanoparticles and the encapsulation of natural compounds offer eco-friendly, biocompatible alternatives, broadening the possibilities for AD treatment. This review also discusses key mechanisms, such as receptor-mediated and adsorptive-mediated transcytosis, employed by nanoparticles to cross the BBB. Finally, the challenges, potential toxicity issues, and future directions for the clinical application of these nano-therapeutics are also addressed. The integration of nanotechnology into AD treatment strategies presents a promising frontier for managing the disease and potentially developing disease-modifying therapies.

Keywords

Alzheimer’s disease, nanoparticles, blood–brain barrier, targeted drug delivery, nanomedicine, amyloid-beta, tau protein, neuroinflammation, oxidative stress, green synthesis, polymeric nanoparticles, solid lipid nanoparticles, dendrimers, liposomes, natural compounds, nanotheranostics, receptor-mediated transcytosis, CNS drug delivery, brain-targeting nanocarriers, nanotechnology

Introduction

Alzheimer's disease (AD) is the most common form of dementia, responsible for approximately 60–70% of all dementia cases globally. Characterized by progressive cognitive decline, memory loss, and changes in personality, AD poses a significant public health challenge, particularly among aging populations. The World Health Organization reports that more than 55 million people are currently living with dementia, and this number is expected to surge to 152 million by 2050 due to increased life expectancy worldwide. The pathophysiology of AD is complex and involves several key features, including the accumulation of amyloid-beta (Aβ) plaques in the extracellular space, the formation of neurofibrillary tangles from hyperphosphorylated tau protein inside neurons, oxidative stress, neuroinflammation, and the degeneration of cholinergic neurons. Despite years of extensive research, existing treatments—primarily acetylcholinesterase inhibitors and NMDA receptor antagonists—only provide temporary symptom relief without addressing the underlying disease progression. One major challenge in developing effective AD treatments is the blood-brain barrier (BBB), a highly selective interface composed of endothelial cells, pericytes, astrocytic end-feet, and a basement membrane. This barrier restricts the entry of most therapeutic agents, including peptides, antibodies, and small-molecule drugs, with over 98% of potential neurotherapeutics unable to pass through. As a result, delivering sufficient drug concentrations to the brain remains a critical hurdle in AD management. Nanotechnology has opened up promising possibilities for drug delivery to the central nervous system. Nanoparticles (NPs) are particularly effective due to their small size, customizable surfaces, and potential for targeted delivery, making them an excellent solution for overcoming the BBB. Various nanocarriers, such as liposomes, polymeric nanoparticles, metallic nanoparticles, solid lipid nanoparticles (SLNs), micelles, and dendrimers, have been developed to enhance drug transport across the BBB, improving drug solubility, stability, bioavailability, and pharmacokinetics. Furthermore, functionalizing nanoparticles with ligands that target specific receptors on the BBB (e.g., transferrin, insulin, LDL receptors) enables more precise delivery via active targeting and transcytosis. Recent advancements also focus on green-synthesized nanoparticles and carriers loaded with natural compounds, which present environmentally friendly, biocompatible, and less toxic alternatives to traditional methods. This review explores recent progress in nanoparticle-based approaches for crossing the BBB and treating Alzheimer’s disease. It examines different types of nanoparticles, mechanisms for crossing the BBB, drug delivery methods, safety considerations, and future directions, with the goal of demonstrating how nanomedicine could transform AD treatment.

2. Pathophysiology of Alzheimer’s Disease

Alzheimer's disease (AD) is a complex neurodegenerative condition, primarily affecting older adults. Its progression is characterized by gradual cognitive decline, memory loss, changes in behavior, and ultimately, the loss of independence. At the molecular level, several interconnected pathological processes contribute to the onset and advancement of the disease, with the most prominent being:

2.1 Amyloid-β (Aβ) Plaques

One of the earliest and most extensively studied features of AD is the accumulation of amyloid-β (Aβ) peptides, which result from the abnormal cleavage of amyloid precursor protein (APP) by β- and γ-secretases. Particularly, Aβ42 peptides are prone to aggregation, forming insoluble extracellular plaques that interfere with neuronal communication and induce neuroinflammatory responses.

• Oligomeric forms of Aβ are considered highly neurotoxic and are associated with:
• Synaptic dysfunction

• Impaired neurotransmission

• Oxidative stress induction

• Activation of microglia and chronic neuroinflammation

2.2 Neurofibrillary Tangles (NFTs)

The second major pathological feature of AD is the formation of neurofibrillary tangles inside neurons, composed of hyperphosphorylated tau protein. Tau, a protein that stabilizes microtubules in neurons, becomes abnormally phosphorylated in AD, causing it to detach from microtubules, aggregate into twisted filaments, and form NFTs. This leads to neuronal damage and cell death.

2.3 Oxidative Stress and Mitochondrial Dysfunction

Oxidative stress and mitochondrial dysfunction are critical contributors to AD progression. The accumulation of Aβ and tau hyperphosphorylation intensify oxidative damage, resulting in lipid peroxidation, DNA damage, and protein oxidation. This oxidative imbalance disrupts cellular functions and accelerates neurodegeneration.

2.4 Neuroinflammation

Microglia, the brain's resident immune cells, become persistently activated in AD, releasing inflammatory cytokines such as TNF-α, IL-1β, and IL-6. This ongoing neuroinflammatory response exacerbates Aβ accumulation and tau pathology, creating a feedback loop that accelerates disease progression.

2.5 Cholinergic Deficits

AD is also marked by the loss of cholinergic neurons in the basal forebrain, leading to a significant reduction in acetylcholine (ACh) levels. This reduction is closely linked to impaired learning and memory, which is why cholinesterase inhibitors (e.g., donepezil, rivastigmine) are commonly used for symptomatic management.

2.6 Blood–Brain Barrier (BBB) Dysfunction

Recent studies suggest that the blood-brain barrier (BBB) is compromised in AD. A dysfunctional BBB impedes the clearance of Aβ, allows immune cells to infiltrate the brain, and contributes to neuroinflammation, further damaging neurons and complicating the delivery of drugs to the central nervous system (CNS). These overlapping pathological mechanisms highlight the need for therapeutic strategies that target multiple pathways and are capable of penetrating the brain. Nanoparticles, which can bypass or interact with the BBB to deliver drugs directly to the affected areas, present a promising approach to effectively addressing these pathological features.

Table 1. Key Pathological Mechanisms in Alzheimer’s Disease

Pathological Feature

Description

Consequences

Amyloid-β (Aβ) Plaques

Extracellular accumulation of aggregated Aβ peptides (primarily Aβ42) derived from APP cleavage.

Synaptic dysfunction, neurotoxicity, microglial activation, and chronic inflammation.

Neurofibrillary Tangles

Intracellular aggregates of hyperphosphorylated tau protein forming paired helical filaments.

Cytoskeletal disruption, impaired axonal transport, neuronal dysfunction, and cell death.

Oxidative Stress

Imbalance between reactive oxygen species (ROS) and antioxidant defenses.

Mitochondrial damage, DNA/protein/lipid oxidation, neuronal apoptosis.

Neuroinflammation

Chronic activation of microglia and astrocytes.

Release of pro-inflammatory cytokines (e.g., IL-1β, TNF-α), further promoting Aβ and tau damage.

Cholinergic Deficits

Loss of acetylcholine-producing neurons in the basal forebrain.

Impaired memory, attention, and cognitive function.

BBB Dysfunction

Disruption of tight junctions and endothelial cell integrity.

Reduced Aβ clearance, increased neurotoxicity, and impaired drug delivery.

3. The Blood–Brain Barrier and Its Role in Drug Delivery Challenges

The blood-brain barrier (BBB) is a dynamic, highly selective structure that safeguards the brain from potentially harmful substances in the bloodstream while preserving the homeostasis required for proper neural function. Despite its crucial protective role, the BBB presents a significant challenge to the delivery of therapeutic agents aimed at treating central nervous system (CNS) disorders, including Alzheimer’s disease (AD).

3.1 Structure and Function of the BBB

The BBB consists mainly of:

• Endothelial cells connected by tight junctions, which restrict the movement of molecules between cells.
• Pericytes, which help regulate vascular stability and permeability.

• Astrocytic end-feet, forming part of the neurovascular unit.

• A basement membrane, providing structural integrity. Together, these elements create a tightly controlled interface that selectively allows the passage of essential nutrients (e.g., glucose, amino acids) while blocking the entry of harmful substances such as pathogens, toxins, and most large or water-soluble molecules.

3.2 Drug Delivery Limitations Imposed by the BBB

More than 98% of small-molecule drugs and nearly all larger therapeutic agents (e.g., peptides, proteins, antibodies, nucleic acids) cannot cross the BBB at therapeutic concentrations. Key limitations include:

• Efflux transporters (e.g., P-glycoprotein) that actively pump drugs out of the brain.
• Tight junctions that prevent the passive diffusion of molecules.

• Enzymatic breakdown of drugs before or during their transit across the BBB.

• The absence of receptor-mediated uptake for many therapeutic agents. These challenges significantly reduce the effectiveness of traditional AD treatments and impede the development of new drug candidates that are unable to reach their intended targets in the brain.

3.3 BBB Dysfunction in Alzheimer’s Disease

Emerging research indicates that the BBB is often compromised in AD due to:

• Chronic neuroinflammation

• Vascular damage induced by Aβ accumulation
• Age-related changes in endothelial function
This dysfunction worsens disease progression by impairing the clearance of Aβ, allowing neurotoxic substances to enter the brain, and amplifying inflammatory responses.

3.4 The Need for Novel Delivery Strategies

Given these challenges, there is an urgent need for new drug delivery methods capable of bypassing or penetrating the BBB. Nanotechnology presents several promising solutions, including:
• Enhanced targeting of the brain through receptor-ligand interactions.

• Protection of drugs from degradation during transit.

• Controlled, sustained release of therapeutic agents.
• The potential for dual diagnostic and therapeutic functions (theranostics).

4. Nanoparticles as Vehicles to Cross the Blood–Brain Barrier

Nanoparticles (NPs) have transformed drug delivery systems by allowing therapeutic agents to cross the blood-brain barrier (BBB) through targeted and controlled approaches. Due to their distinctive physicochemical properties—such as their small size, adjustable surface charge, and capacity to carry both hydrophilic and hydrophobic drugs—NPs are well-suited for treating neurological disorders like Alzheimer’s disease (AD). This section discusses the various types of nanoparticles used, their mechanisms for penetrating the BBB, and their potential applications in AD treatment.

4.1 Advantages of Nanoparticles in Brain Drug Delivery

Nanoparticles offer several advantages for targeting the central nervous system (CNS):

Small size (typically <200 nm): Enables NPs to diffuse through or undergo transcytosis across tight junctions in the BBB.

Surface functionalization: Allows the attachment of targeting ligands (e.g., transferrin, lactoferrin, peptides) to enhance brain-specific delivery.

Drug protection: Encapsulates drugs, shielding them from enzymatic degradation during transit.

Controlled release: Provides sustained and localized drug delivery at the target site.

Multifunctionality: Facilitates both diagnostic imaging and therapeutic applications within a single platform ("theranostics").

4.2 Mechanisms of BBB Penetration by Nanoparticles

Nanoparticles can cross the BBB via several pathways:

Mechanism

Description

Receptor-Mediated Transcytosis

NPs functionalized with ligands (e.g., transferrin, insulin) bind receptors on BBB endothelial cells.

Adsorptive-Mediated Transcytosis

Cationic NPs interact with negatively charged cell membranes to facilitate uptake.

Carrier-Mediated Transport

NPs mimic endogenous substrates (e.g., glucose, amino acids) to utilize transporters.

Cell-Mediated Transport

NPs are loaded into monocytes/macrophages that naturally cross the BBB.

Disruption Techniques

Temporary BBB opening by focused ultrasound or hyperosmotic agents to allow NP entry.

4.3 Types of Nanoparticles Used in Alzheimer’s Therapy

Nanoparticle Type

Composition

Applications in AD

Polymeric NPs

PLGA, PEG, chitosan

Controlled release; biodegradable; good BBB penetration with surface modifications.

Metallic NPs

Gold, selenium, silver

Antioxidant effects; imaging; functionalized for Aβ targeting.

Solid Lipid Nanoparticles (SLNs)

Lipid-based core/shell systems

High stability; good brain permeability; encapsulate hydrophobic drugs.

Liposomes

Phospholipid bilayers

Biocompatible; can carry both hydrophilic and hydrophobic agents; easy to functionalize.

Dendrimers

Branched polymers (e.g., PAMAM)

High drug-loading capacity; used in gene and peptide delivery.

Green-synthesized NPs

Plant extract-based metallic NPs

Eco-friendly, lower toxicity; antioxidant and anti-inflammatory effects.

4.4 Functionalization for Targeted Delivery

Surface modification plays a crucial role in improving the ability of nanoparticles to cross the BBB and target specific areas:

Ligand targeting: This involves attaching antibodies, peptides (e.g., RVG, TGN), or vitamins (e.g., folate) to nanoparticles for enhanced specificity.

PEGylation: This modification extends the nanoparticles' circulation time in the body and helps prevent rapid clearance.

Dual-functional systems: These systems combine targeting agents with imaging agents, enabling theranostic applications (both therapy and diagnostics in one platform). Nanoparticles have shown great potential in preclinical studies for improving brain drug delivery. Their ongoing development and refinement may help overcome many of the current challenges faced by existing AD treatments.

5. Specific Applications of Nanoparticles in Alzheimer’s Disease Treatment

Nanoparticles are designed not only to cross the blood-brain barrier (BBB) but also to interact with Alzheimer’s disease (AD) pathology at various levels. Their versatility allows them to carry drugs, natural compounds, imaging agents, and even genetic materials, making them powerful tools for both therapeutic and diagnostic (theranostic) applications. This section discusses how nanoparticles have been specifically utilized to target key AD-related mechanisms, such as amyloid-beta aggregation, tau pathology, oxidative stress, inflammation, and cholinergic dysfunction.

5.1 Targeting Amyloid-β Aggregation
The amyloid cascade hypothesis plays a central role in the pathogenesis of AD. Several nanoparticle systems have been developed to either reduce amyloid-beta (Aβ) accumulation or promote its clearance:

• Gold nanoparticles functionalized with peptides can bind to Aβ and prevent its aggregation.

• Selenium nanoparticles (SeNPs) aid in clearing Aβ and provide antioxidant effects.

• Polymeric nanoparticles loaded with Aβ-targeting antibodies or β-secretase inhibitors (BACE1 inhibitors) enhance targeted delivery of drugs to amyloid plaques.

5.2 Inhibiting Tau Hyperphosphorylation

Tau pathology, although less focused on than Aβ, is also a critical target for therapy:

• Dendrimers and polymeric nanoparticles have been used to deliver kinase inhibitors that help reduce tau hyperphosphorylation.

• PLGA-based nanoparticles deliver small molecules that prevent tau aggregation and promote microtubule stability.

5.3 Combating Oxidative Stress

Oxidative stress is both a trigger and a result of AD pathology. Nanoparticles loaded with antioxidants offer neuroprotective benefits:

• Curcumin-loaded liposomes and polymeric nanoparticles scavenge reactive oxygen species (ROS).
• Green-synthesized metal nanoparticles (e.g., silver, zinc oxide) exhibit both antioxidant and anti-amyloidogenic properties.

• Selenium and cerium oxide nanoparticles neutralize ROS and enhance mitochondrial function.

5.4 Modulating Neuroinflammation

Chronic activation of microglia leads to persistent neuroinflammation:

• Chitosan and PEGylated nanoparticles have been developed to deliver anti-inflammatory agents like NSAIDs, siRNA, or dexamethasone across the BBB.
• Natural anti-inflammatory compounds, such as resveratrol encapsulated in lipid-based nanoparticles, help reduce microglial activation and cytokine release.

5.5 Enhancing Cholinergic Function

Approved AD drugs, such as donepezil, rivastigmine, and galantamine, have limited ability to cross the BBB:

• Solid lipid nanoparticles (SLNs) and liposomes have been used to encapsulate donepezil, improving its delivery to cholinergic neurons in the cortex and hippocampus.

• SLNs modified with ApoE or transferrin ligands enhance the brain uptake of acetylcholinesterase inhibitors (AChEIs).

5.6 Natural Compound Delivery

Natural phytochemicals (e.g., curcumin, resveratrol, quercetin) possess neuroprotective effects but have poor solubility and BBB permeability:

• Nanocarriers significantly improve the bioavailability and targeting of these compounds.
• These systems have shown promise in reducing Aβ and tau toxicity, as well as oxidative and inflammatory responses.

5.7 Theranostic Applications

Nanoparticles can be engineered to serve dual diagnostic and therapeutic functions:

• Superparamagnetic iron oxide nanoparticles (SPIONs) conjugated with Aβ antibodies are used for MRI-based detection.

• Quantum dots and fluorescent-labeled nanoparticles enable early-stage AD imaging while simultaneously delivering therapeutic agents.

Nanoparticles provide a multifaceted approach to treating AD by targeting various pathological mechanisms. Their continued development holds the potential to offer more effective, targeted, and minimally invasive treatment options.

Table 2. Summary of Nanoparticle Applications in Alzheimer's Disease Therapy

Therapeutic Target

Pathology Addressed

Nanoparticle Strategy

Examples

Amyloid-β Aggregation

Extracellular plaque formation

Inhibit aggregation, promote clearance

Gold NPs with Aβ-targeting peptides, Selenium NPs, BACE1 inhibitor-loaded NPs

Tau Hyperphosphorylation

Neurofibrillary tangle formation

Deliver kinase inhibitors to reduce tau aggregation

PLGA NPs with tau-targeted agents, dendrimers carrying tau kinase blockers

Oxidative Stress

ROS-induced neuronal damage

Deliver antioxidant compounds

Curcumin-loaded liposomes, green-synthesized metallic NPs (Se, Zn, Ag)

Neuroinflammation

Microglial activation and cytokine overexpression

Modulate immune response, deliver anti-inflammatory drugs

PEGylated NPs with NSAIDs, Resveratrol-loaded lipid NPs

Cholinergic Dysfunction

Loss of acetylcholine and impaired cognition

Enhance delivery of acetylcholinesterase inhibitors (AChEIs)

Donepezil-loaded SLNs, liposomes modified with ApoE

Natural Compound Delivery

Multi-target neuroprotection

Improve solubility, stability, and BBB permeability of plant-derived therapeutics

Quercetin, curcumin, and resveratrol encapsulated in polymeric/lipid NPs

Theranostics (Dual Role)

Diagnosis + treatment

Combine imaging and therapeutic agents in multifunctional NPs

SPIONs for MRI + drug delivery, fluorescent quantum dots with Aβ-targeting

6. Toxicity, Safety, and Regulatory Challenges in Nanoparticle-Based Alzheimer’s Therapy

Nanoparticle (NP)-based drug delivery systems hold great promise for Alzheimer’s disease (AD) treatment, but their clinical application is still hindered by concerns regarding toxicity, safety, and regulatory hurdles. It is essential that nanocarriers are biocompatible, non-immunogenic, and safely degradable in the body before they can be widely used in human therapies.

6.1 Toxicity and Biocompatibility Concerns

Nanoparticles interact with biological systems in complex ways that can sometimes result in unintended toxic effects. Major concerns include:

Accumulation and persistence: Non-degradable nanoparticles may accumulate in organs such as the liver, spleen, and brain, leading to long-term toxicity.

Oxidative stress: Some metal-based nanoparticles (e.g., silver, iron oxide) may produce reactive oxygen species (ROS), which can damage cells and organelles.

Immune responses: Nanoparticle surface properties could trigger immune activation or allergic reactions, particularly with repeated use.

Neurotoxicity: Poorly designed nanoparticles might interfere with neuronal function, potentially worsening AD symptoms.

Preclinical toxicity testing is crucial and should focus on:

• Hemocompatibility

• Inflammation markers

• BBB integrity

• Behavioral and neurocognitive effects in animal models

6.2 Green-Synthesized Nanoparticles: A Safer Alternative
Recent research has explored green synthesis methods using plant extracts and biocompatible materials to reduce toxicity:

• These methods avoid the harsh chemicals commonly used in traditional synthesis.

• They show reduced immunogenicity and improved biodegradability.

• They maintain or even enhance therapeutic efficacy in targeting Aβ and alleviating oxidative stress.
• These environmentally friendly techniques offer a more sustainable and safer path to clinical application, particularly for chronic diseases like AD.

6.3 Regulatory Barriers

The regulatory landscape for nanoparticle-based therapies is complex and evolving. Key challenges include:

Standardization issues: There is a lack of consistent methods for characterizing nanoparticle properties, such as size, charge, drug loading, and release profiles.

Reproducibility and scalability: Lab-scale formulations often fail to meet industrial-scale requirements for Good Manufacturing Practices (GMP).

Long-term safety data: There is limited clinical data on the long-term effects of nanoparticles, particularly in elderly patients with comorbid conditions.

Approval pathways: Current drug approval processes are not fully prepared to evaluate hybrid materials (e.g., combinations of drugs, imaging agents, and carriers). Regulatory agencies like the FDA and EMA are beginning to issue guidance for nanomedicines, but a unified global regulatory framework is needed to accelerate development.

6.4 Strategies to Overcome Challenges
To improve safety and regulatory compliance, the following strategies are recommended:

• Use biodegradable polymers (e.g., PLGA, chitosan).

• Optimize surface modifications to reduce immunogenicity.

• Conduct comprehensive in vivo toxicity studies, including cognitive and behavioral assessments.
• Engage with regulatory agencies early to design adaptive trials and identify accelerated approval pathways.

Despite these challenges, ongoing innovation in nanoparticle design, synthesis, and testing protocols is steadily advancing their safe and effective use in Alzheimer’s therapy.

Table 3. Toxicity and Safety Considerations for Nanoparticles in Alzheimer’s Disease Therapy

Aspect

Potential Issue

Mitigation Strategies

Accumulation & Persistence

Long-term buildup in organs (e.g., brain, liver, spleen) may cause toxicity.

Use biodegradable and excretable materials (e.g., PLGA, chitosan).

Oxidative Stress

Some NPs (especially metallic) can generate reactive oxygen species (ROS).

Surface coating, antioxidant co-delivery, or using green-synthesized NPs.

Immune Response

Nanoparticles may activate immune cells or provoke inflammation.

PEGylation, ligand masking, and surface charge optimization.

Neurotoxicity

Poorly designed NPs may interfere with neuronal function or exacerbate pathology.

Conduct neurobehavioral testing and target-specific delivery.

Regulatory Complexity

Lack of standardized guidelines for nanoparticle characterization and approval.

Early collaboration with regulatory bodies and robust physicochemical profiling.

Manufacturing Challenges

Scalability and batch reproducibility of NP synthesis.

Adopt GMP-compliant, scalable synthesis methods and quality control.

7. Future Prospects and Conclusion

The integration of nanotechnology into Alzheimer’s disease (AD) therapy represents a significant breakthrough in overcoming the challenges imposed by the blood–brain barrier (BBB) and the complex nature of the disease. Nanoparticles provide highly specialized, multifunctional platforms for targeted drug and diagnostic delivery to the brain. These systems have shown considerable promise in reducing amyloid-beta accumulation, modulating tau pathology, alleviating oxidative stress, and controlling neuroinflammation. Looking ahead, several exciting avenues are emerging:

7.1 Future Directions in Research and Development

Personalized Nanomedicine: Customizing nanoparticle systems based on individual genetic factors, such as APOE4 status, could enhance treatment outcomes and minimize side effects.

Stimuli-Responsive NPs: "Smart" nanocarriers that release their cargo in response to specific environmental triggers (e.g., pH, temperature, enzymes) in the AD brain could allow for precise, localized delivery.

Multifunctional Theranostics: Combining diagnostic and therapeutic functions in a single system (e.g., imaging via MRI or fluorescence) could enable early AD detection and continuous monitoring of disease progression.

Natural Compound Integration: Further research into plant-based or dietary compounds encapsulated in nanoparticles could provide low-toxicity, brain-penetrating treatments.

Cross-Disciplinary Collaborations: Incorporating artificial intelligence (AI) into nanomedicine could accelerate drug screening, predict nanoparticle behavior, and optimize the design of nanocarrier systems.

7.2 Clinical Translation and Challenges

Despite substantial progress in preclinical studies, translating these findings into human clinical trials remains challenging. To bridge this gap:

• Standardized models are needed to evaluate nanoparticle behavior in AD-specific conditions.

• Long-term safety data is vital, particularly in aging populations with comorbidities.

• Early-phase clinical trials focused on safety, tolerability, and pharmacokinetics will help build confidence in nanomedicine platforms.

• Global regulatory harmonization is necessary to streamline approval processes.

CONCLUSION

Alzheimer’s disease continues to be one of the most debilitating neurodegenerative disorders worldwide, placing a heavy burden on individuals, caregivers, and healthcare systems. A major obstacle in developing effective treatments for AD is the blood–brain barrier (BBB), which prevents most therapeutic agents from reaching the brain. Conventional drug delivery methods have struggled to address the multifactorial nature of AD, which includes amyloid-beta aggregation, tau hyperphosphorylation, oxidative stress, neuroinflammation, and cholinergic dysfunction. Nanotechnology offers a promising solution to these challenges. Nanoparticle-based systems can overcome the BBB through strategies like receptor-mediated transcytosis, adsorptive-mediated transport, and cell-based delivery. Additionally, nanoparticles can be engineered with targeting ligands, responsive drug release mechanisms, and imaging agents for both diagnostic and therapeutic purposes. These capabilities improve drug stability, brain penetration, and reduce systemic side effects. Innovative methods, such as green synthesis of nanoparticles, offer eco-friendly and biocompatible alternatives to traditional synthesis techniques. Encapsulating natural compounds like curcumin, resveratrol, and quercetin in nanoparticles also opens new avenues for clinically viable, antioxidant-rich treatments. These approaches align with current trends towards sustainable, patient-friendly therapies. However, significant challenges remain. Extensive preclinical and clinical evaluations are needed to assess the long-term safety, distribution, metabolism, and immunogenicity of nanoparticles. Additionally, regulatory frameworks must evolve to address the complexities of nanomedicines, especially for long-term AD treatments. Scalability and reproducibility also pose technical barriers to commercializing nanoparticle-based formulations. Nonetheless, the convergence of materials science, neuroscience, molecular biology, and pharmaceutical technologies positions nanomedicine at the forefront of next-generation Alzheimer’s treatments. The future lies in developing smarter, safer, and more effective nanoplatforms capable of not only delivering drugs but also diagnosing diseases earlier, monitoring therapeutic responses, and possibly reversing disease progression. In conclusion, while challenges remain, nanoparticle-based drug delivery systems are redefining Alzheimer’s treatment. Continued interdisciplinary research, regulatory support, and investment in translational studies are essential for bringing these innovations from research to clinical practice. With these efforts, personalized, targeted, and efficient Alzheimer’s therapies could soon be a reality.

REFERENCES

  1. Nagori, K., Nakhate, K.T., Yadav, K., et al. (2025). Innovative strategies for overcoming blood-brain barrier challenges in Alzheimer’s disease: A focus on green-synthesized metallic nanoparticles. Inorganic Chemistry Communications, 171, 113604. https://doi.org/10.1016/j.inoche.2024.113604
  2. Nady, D.S., Bakowsky, U., & Fahmy, S.A. (2023). Recent advances in brain delivery of synthetic and natural nano therapeutics: Reviving hope for Alzheimer’s disease patients. Journal of Drug Delivery Science and Technology, 89, 105047. https://doi.org/10.1016/j.jddst.2023.105047
  3. Song, Q., Li, J., Li, T., & Li, H.W. (2024). Nanomaterials that aid in the diagnosis and treatment of Alzheimer’s disease: Resolving blood–brain barrier crossing ability. Advanced Science, 11(38), 2403473. https://doi.org/10.1002/advs.202403473
  4. Ramalho, M.J., Andrade, S., Loureiro, J.A., & Pereira, M.C. (2025). Could encapsulation of natural compounds in BBB-permeable nanocarriers produce effective Alzheimer’s disease treatments? Nanomedicine, 20(5), 435-438. https://doi.org/10.1080/17435889.2024.2444859
  5. Cao, Y., & Zhang, R. (2022). The application of nanotechnology in treatment of Alzheimer’s disease. Frontiers in Bioengineering and Biotechnology, 10, 1042986. https://doi.org/10.3389/fbioe.2022.1042986
  6. Ceña, V., & Játiva, P. (2018). Nanoparticle crossing of blood–brain barrier: A road to new therapeutic approaches to central nervous system diseases. Nanomedicine, 13(13), 1513–1516. https://doi.org/10.2217/nnm-2018-0139
  7. Navarro Martínez, N., Toledo Hernández, J., & Morales, J.O. (2023). Nanoparticles for the potential treatment of Alzheimer’s disease: A physiopathological approach. Nanotechnology Reviews, 12, 20220548. https://doi.org/10.1515/ntrev-2022-0548
  8. Teleanu, D.M., Chircov, C., Grumezescu, A.M., et al. (2018). Blood-brain delivery methods using nanotechnology. Pharmaceutics, 10(4), 269. https://doi.org/10.3390/pharmaceutics10040269
  9. Poudel, P., & Park, S. (2022). Recent advances in the treatment of Alzheimer’s disease using nanoparticle-based drug delivery systems. Pharmaceutics, 14(4), 835. https://doi.org/10.3390/pharmaceutics14040835
  10. Zha, S., Liu, H., Li, H., et al. (2024). Functionalized nanomaterials capable of crossing the blood–brain barrier. ACS Nano, 18, 1820–1845. https://doi.org/10.1021/acsnano.3c10674
  11. Cummings, J., et al. (2019). Drug development in Alzheimer’s disease: The path to 2025. Alzheimer's Research & Therapy, 11(1), 60.
  12. Long, J.M., & Holtzman, D.M. (2019). Alzheimer disease: An update on pathobiology and treatment strategies. Cell, 179(2), 312–339.
  13. Sweeney, M.D., et al. (2018). The role of brain vasculature in neurodegenerative disorders. Nature Neuroscience, 21, 1318–1331.
  14. Saraiva, C., et al. (2016). Nanoparticle-mediated brain drug delivery: Overcoming blood-brain barrier to treat neurodegenerative diseases. Journal of Controlled Release, 235, 34–47.
  15. Anand, P., & Singh, B. (2020). A review on cholinesterase inhibitors for Alzheimer’s disease. Archives of Pharmacal Research, 43(8), 742–755.
  16. Kuo, Y.C., & Wang, C.C. (2014). Cationic solid lipid nanoparticles with surface ApoE can cross the BBB and enter microglial cells. Colloids and Surfaces B, 123, 610–617.
  17. Xu, L., et al. (2022). Intranasal delivery of nanoparticles for brain-targeted therapy. Frontiers in Pharmacology, 13, 837558.
  18. Pardridge, W.M. (2012). Drug transport across the blood–brain barrier. Journal of Cerebral Blood Flow & Metabolism, 32(11), 1959–1972.
  19. Shilo, M., Sharon, A., & Barenholz, Y. (2012). Cancer nanomedicine: Passive and active targeting. Nanomedicine, 7(5), 639–652.
  20. Tiwari, S., et al. (2022). Dendrimers in Alzheimer’s disease: Current status and future perspectives. International Journal of Pharmaceutics, 617, 121597.
  21. Saeedi, M., et al. (2019). Applications of dendrimers in drug delivery to the central nervous system. Current Neuropharmacology, 17(7), 668–681.
  22. Patel, T., Zhou, J., & Piepmeier, J.M. (2012). Polymeric nanoparticles for drug delivery to the central nervous system. Advanced Drug Delivery Reviews, 64(7), 701–705.
  23. Jiang, Y., et al. (2021). Redox-responsive nanoparticles for brain-targeted drug delivery. Advanced Functional Materials, 31(5), 2006942.
  24. Tiwari, D., & Sharma, R. (2020). Micelles as brain drug delivery carriers. Artificial Cells, Nanomedicine, and Biotechnology, 48(1), 69–78.
  25. Guterres, S.S., et al. (2018). Polymeric nanoparticles: Promising carriers for brain drug delivery. Current Pharmaceutical Design, 24(13), 1455–1466.
  26. Esfandyari-Manesh, M., et al. (2021). Niosome-based strategies for Alzheimer’s therapy. Journal of Drug Delivery Science and Technology, 66, 102785.
  27. Wong, H.L., Bendayan, R., Rauth, A.M., et al. (2007). A mechanistic study of enhanced doxorubicin uptake and retention in brain by polysorbate 80-coated nanoparticles. Journal of Controlled Release, 116(1), 49–59.
  28. Wang, L., et al. (2020). Solid lipid nanoparticles for brain drug delivery: Opportunities and challenges. Pharmaceutics, 12(12), 1055.
  29. Demeule, M., et al. (2008). Receptor-mediated brain delivery of morphine using a peptide vector. Journal of Neurochemistry, 106(4), 1534–1544.
  30. Arora, S., et al. (2022). Multifunctional gold nanoparticles for Alzheimer’s disease: Focus on targeting, diagnostics, and therapeutic potential. Biomedicine & Pharmacotherapy, 146, 112553.
  31. Patel, M., et al. (2022). BBB-penetrating nanoparticles: A boon for the treatment of brain disorders. Journal of Controlled Release, 351, 90–112.
  32. Zhang, Y., et al. (2020). The application of inorganic nanomaterials in Alzheimer’s disease diagnosis and therapy. Journal of Nanobiotechnology, 18, 1–20.

Reference

  1. Nagori, K., Nakhate, K.T., Yadav, K., et al. (2025). Innovative strategies for overcoming blood-brain barrier challenges in Alzheimer’s disease: A focus on green-synthesized metallic nanoparticles. Inorganic Chemistry Communications, 171, 113604. https://doi.org/10.1016/j.inoche.2024.113604
  2. Nady, D.S., Bakowsky, U., & Fahmy, S.A. (2023). Recent advances in brain delivery of synthetic and natural nano therapeutics: Reviving hope for Alzheimer’s disease patients. Journal of Drug Delivery Science and Technology, 89, 105047. https://doi.org/10.1016/j.jddst.2023.105047
  3. Song, Q., Li, J., Li, T., & Li, H.W. (2024). Nanomaterials that aid in the diagnosis and treatment of Alzheimer’s disease: Resolving blood–brain barrier crossing ability. Advanced Science, 11(38), 2403473. https://doi.org/10.1002/advs.202403473
  4. Ramalho, M.J., Andrade, S., Loureiro, J.A., & Pereira, M.C. (2025). Could encapsulation of natural compounds in BBB-permeable nanocarriers produce effective Alzheimer’s disease treatments? Nanomedicine, 20(5), 435-438. https://doi.org/10.1080/17435889.2024.2444859
  5. Cao, Y., & Zhang, R. (2022). The application of nanotechnology in treatment of Alzheimer’s disease. Frontiers in Bioengineering and Biotechnology, 10, 1042986. https://doi.org/10.3389/fbioe.2022.1042986
  6. Ceña, V., & Játiva, P. (2018). Nanoparticle crossing of blood–brain barrier: A road to new therapeutic approaches to central nervous system diseases. Nanomedicine, 13(13), 1513–1516. https://doi.org/10.2217/nnm-2018-0139
  7. Navarro Martínez, N., Toledo Hernández, J., & Morales, J.O. (2023). Nanoparticles for the potential treatment of Alzheimer’s disease: A physiopathological approach. Nanotechnology Reviews, 12, 20220548. https://doi.org/10.1515/ntrev-2022-0548
  8. Teleanu, D.M., Chircov, C., Grumezescu, A.M., et al. (2018). Blood-brain delivery methods using nanotechnology. Pharmaceutics, 10(4), 269. https://doi.org/10.3390/pharmaceutics10040269
  9. Poudel, P., & Park, S. (2022). Recent advances in the treatment of Alzheimer’s disease using nanoparticle-based drug delivery systems. Pharmaceutics, 14(4), 835. https://doi.org/10.3390/pharmaceutics14040835
  10. Zha, S., Liu, H., Li, H., et al. (2024). Functionalized nanomaterials capable of crossing the blood–brain barrier. ACS Nano, 18, 1820–1845. https://doi.org/10.1021/acsnano.3c10674
  11. Cummings, J., et al. (2019). Drug development in Alzheimer’s disease: The path to 2025. Alzheimer's Research & Therapy, 11(1), 60.
  12. Long, J.M., & Holtzman, D.M. (2019). Alzheimer disease: An update on pathobiology and treatment strategies. Cell, 179(2), 312–339.
  13. Sweeney, M.D., et al. (2018). The role of brain vasculature in neurodegenerative disorders. Nature Neuroscience, 21, 1318–1331.
  14. Saraiva, C., et al. (2016). Nanoparticle-mediated brain drug delivery: Overcoming blood-brain barrier to treat neurodegenerative diseases. Journal of Controlled Release, 235, 34–47.
  15. Anand, P., & Singh, B. (2020). A review on cholinesterase inhibitors for Alzheimer’s disease. Archives of Pharmacal Research, 43(8), 742–755.
  16. Kuo, Y.C., & Wang, C.C. (2014). Cationic solid lipid nanoparticles with surface ApoE can cross the BBB and enter microglial cells. Colloids and Surfaces B, 123, 610–617.
  17. Xu, L., et al. (2022). Intranasal delivery of nanoparticles for brain-targeted therapy. Frontiers in Pharmacology, 13, 837558.
  18. Pardridge, W.M. (2012). Drug transport across the blood–brain barrier. Journal of Cerebral Blood Flow & Metabolism, 32(11), 1959–1972.
  19. Shilo, M., Sharon, A., & Barenholz, Y. (2012). Cancer nanomedicine: Passive and active targeting. Nanomedicine, 7(5), 639–652.
  20. Tiwari, S., et al. (2022). Dendrimers in Alzheimer’s disease: Current status and future perspectives. International Journal of Pharmaceutics, 617, 121597.
  21. Saeedi, M., et al. (2019). Applications of dendrimers in drug delivery to the central nervous system. Current Neuropharmacology, 17(7), 668–681.
  22. Patel, T., Zhou, J., & Piepmeier, J.M. (2012). Polymeric nanoparticles for drug delivery to the central nervous system. Advanced Drug Delivery Reviews, 64(7), 701–705.
  23. Jiang, Y., et al. (2021). Redox-responsive nanoparticles for brain-targeted drug delivery. Advanced Functional Materials, 31(5), 2006942.
  24. Tiwari, D., & Sharma, R. (2020). Micelles as brain drug delivery carriers. Artificial Cells, Nanomedicine, and Biotechnology, 48(1), 69–78.
  25. Guterres, S.S., et al. (2018). Polymeric nanoparticles: Promising carriers for brain drug delivery. Current Pharmaceutical Design, 24(13), 1455–1466.
  26. Esfandyari-Manesh, M., et al. (2021). Niosome-based strategies for Alzheimer’s therapy. Journal of Drug Delivery Science and Technology, 66, 102785.
  27. Wong, H.L., Bendayan, R., Rauth, A.M., et al. (2007). A mechanistic study of enhanced doxorubicin uptake and retention in brain by polysorbate 80-coated nanoparticles. Journal of Controlled Release, 116(1), 49–59.
  28. Wang, L., et al. (2020). Solid lipid nanoparticles for brain drug delivery: Opportunities and challenges. Pharmaceutics, 12(12), 1055.
  29. Demeule, M., et al. (2008). Receptor-mediated brain delivery of morphine using a peptide vector. Journal of Neurochemistry, 106(4), 1534–1544.
  30. Arora, S., et al. (2022). Multifunctional gold nanoparticles for Alzheimer’s disease: Focus on targeting, diagnostics, and therapeutic potential. Biomedicine & Pharmacotherapy, 146, 112553.
  31. Patel, M., et al. (2022). BBB-penetrating nanoparticles: A boon for the treatment of brain disorders. Journal of Controlled Release, 351, 90–112.
  32. Zhang, Y., et al. (2020). The application of inorganic nanomaterials in Alzheimer’s disease diagnosis and therapy. Journal of Nanobiotechnology, 18, 1–20.

Photo
Shivendra Ughade
Corresponding author

SAGE University Bhopal

Photo
Dr. Rakhee Kapadia
Co-author

SAGE University Bhopal

Photo
Dr. Jitendra Banweer
Co-author

SAGE University Bhopal

Shivendra Ughade*, Dr. Rakhee Kapadia, Dr. Jitendra Banweer, Nanoparticles for Crossing the Blood–Brain Barrier in Alzheimer’s Disease Treatment, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 4, 3282-3294. https://doi.org/10.5281/zenodo.15301793

More related articles
Berberine And Its Study as An Antidiabetic Compoun...
Sonawane Sujata, Thorat Shreya, Talekar Yashaswini, Sonawane Vais...
Formulation And Evaluation of Multipurpose Polyher...
Dr Roopesh P. T., Anjali p., Aishwarya Lakshmi T., Suhana Muhamma...
Formulation and Evaluation of Herbal Mouthwash ...
Pallavi Wagh, Samiksha Wankhade, Aditi Tikait, Dr. Swati Deshmukh...
A Review On Epilepsy Its Treatment...
Misal Vaishnavi , Deokar Shivprasad S, Thombre Vaishnavi B, Bansod Vaishnavi A, Bhande Vaishnavi B, ...
Formulation And Evaluation of Neem Based Shampoo ...
Shivani Awachar, Shivani Wankhade, Aditi Tikait, Dr. Swati Deshmukh, ...
Related Articles
The Role of Vitamin-D Receptor in Alzheimer’s Disease: A Molecular and Clinica...
Vrushali Murari, Arun Pachauri, Kailash Singh Bisht, Anupama Sinha, Lokesh Paranjape, Omkar Sutar, ...
Clinical Review of Antidiabetic Drugs: implications for Type 2 Diabetes Mellitus...
Sahil Nivangune, Shradha Singhi , Shravani Kulkarni, Abhijeet Satpute , Vedanti Walzade , Prathmesh...
Incidence of Type 2 Respiratory Failure in Hypothyroidism...
Sayad maseera fathima , Arshad shaik, ...
Ayurvedic Remedies of Bacterial Meningitis...
Komal Ushir, Dinesh Rishipathak, Pragati Bhosale, Manisha Raut, ...
Berberine And Its Study as An Antidiabetic Compound ...
Sonawane Sujata, Thorat Shreya, Talekar Yashaswini, Sonawane Vaishnavi , Soni Om, ...
More related articles
Berberine And Its Study as An Antidiabetic Compound ...
Sonawane Sujata, Thorat Shreya, Talekar Yashaswini, Sonawane Vaishnavi , Soni Om, ...
Formulation And Evaluation of Multipurpose Polyherbal Powder Shampoo ...
Dr Roopesh P. T., Anjali p., Aishwarya Lakshmi T., Suhana Muhammad Kunji, Jumana Haseen, ...
Formulation and Evaluation of Herbal Mouthwash ...
Pallavi Wagh, Samiksha Wankhade, Aditi Tikait, Dr. Swati Deshmukh, ...
Berberine And Its Study as An Antidiabetic Compound ...
Sonawane Sujata, Thorat Shreya, Talekar Yashaswini, Sonawane Vaishnavi , Soni Om, ...
Formulation And Evaluation of Multipurpose Polyherbal Powder Shampoo ...
Dr Roopesh P. T., Anjali p., Aishwarya Lakshmi T., Suhana Muhammad Kunji, Jumana Haseen, ...
Formulation and Evaluation of Herbal Mouthwash ...
Pallavi Wagh, Samiksha Wankhade, Aditi Tikait, Dr. Swati Deshmukh, ...