Engineering Nanoparticles for Efficient Vaccine Delivery and Immune Modulation

Engineering Nanoparticles for Efficient Vaccine Delivery and Immune Modulation

Introduction to Nanoparticle Vaccine Delivery Systems

Concept and Importance of Nanoparticle-Based Vaccination

Nanoparticle vaccine delivery systems represent a cutting-edge biotechnological approach that utilizes particles ranging from 1 to 1000 nanometers as carriers for antigens and immune modulators. These systems are highly significant because they can mimic the natural size and structure of pathogens, enhancing their recognition and processing by the immune system. The engineered nanoparticles allow precise control over antigen presentation, which can direct specific types of immune responses. Compared to traditional vaccine platforms, nanoparticle systems offer greater flexibility and designability, making them particularly promising for tackling emerging or refractory infectious diseases. As such, this technology platform has become a focal point for the next generation of vaccine development.

Limitations of Conventional Vaccine Formulations

Traditional vaccine formulations, especially soluble protein subunit vaccines, often encounter several challenges in vivo. These antigens are typically unstable in physiological conditions, susceptible to enzymatic degradation, and rapidly cleared from the body, limiting their bioavailability. Although soluble antigens can be drained by the lymphatic system, they lack the physical properties needed for efficient uptake by antigen-presenting cells, necessitating higher doses or multiple doses to elicit an adequate immune response. More importantly, these antigens primarily induce humoral immunity but struggle to effectively activate cellular immunity, which is crucial for eliminating intracellular pathogens like viruses and certain bacteria. While adjuvants can partly enhance immune responses, the difficulty in achieving co-delivery of antigens and adjuvants to the same immune cell remains a significant limitation of conventional vaccine formulations.

Advantages of Nanoparticles in Antigen Delivery and Immune Activation

Nanoparticles exhibit several advantages over traditional formulations due to their unique physicochemical properties. Their nanoscale size facilitates diffusion through tissue interstitial spaces and efficient capture by lymphatic vessels, leading to accumulation in lymph nodes, which are central to immune responses. The surface characteristics of nanoparticles make them readily recognizable and internalized by dendritic cells and other antigen-presenting cells, significantly enhancing antigen uptake. Furthermore, nanoparticles can co-encapsulate or conjugate both antigens and adjuvants, ensuring their simultaneous delivery to the same cell and generating synergistic immune activation effects. This structure also protects antigens from degradation and enables sustained or pulsed release through material design, simulating the effect of repeated immunizations. Additionally, certain nanoparticles can promote cross-presentation of antigens, activating cytotoxic T cells, which is particularly relevant for developing vaccines against cancer and chronic infectious agents.

Fig.1 Schematic showing nanoparticle-based vaccine delivery methods.Fig.1 Schematic of various nanoparticle vaccine delivery systems1,2.

Design Principles of Nanoparticle Vaccine Carriers

Antigen Encapsulation and Presentation Strategies

The method of antigen loading in nanoparticles directly impacts the delivery efficiency and immunogenicity. The primary strategies include internal encapsulation and surface display. Internal encapsulation involves enclosing the antigen within the nanoparticle core, using biodegradable polymers like PLGA to form nanoparticles, which effectively protect the antigen and provide sustained release. In contrast, surface display involves chemically coupling or physically adsorbing antigens onto the nanoparticle surface, such as recombinant antigens displayed on virus-like particles after self-assembly, facilitating direct recognition by B cell receptors to induce a robust antibody response. Hybrid strategies are also commonly employed, such as encapsulating mRNA antigens within lipid nanoparticles while functionalizing their surface with targeting molecules, allowing dual control over antigen expression and presentation.

Surface Functionalization for Immune Cell Targeting

Surface functionalization is a critical strategy for achieving precise immune cell targeting. By conjugating specific ligands, nanoparticles can directly bind to receptors on the surface of target immune cells, enhancing delivery efficiency. For instance, modifying nanoparticles with mannosylated derivatives can target the mannose receptors on dendritic cells, improving cellular uptake. Other approaches include using CD40 antibodies or cytokines like GM-CSF as targeting heads to bind to receptors on dendritic cells or cells expressing GM-CSF receptors. Additionally, adjusting the surface charge and hydrophobicity of nanoparticles can influence their targeting behavior. For example, positively charged particles tend to interact more readily with the negatively charged cell membrane, while polyethylene glycol (PEG) modifications can reduce non-specific protein adsorption, extend circulation time, and promote migration to lymph nodes.

Controlled Release and Stability Enhancement Mechanisms

Achieving controlled release of antigens and adjuvants is one of the central goals of nanoparticle design. This can be accomplished by selecting environment-sensitive materials that trigger specific release behaviors in different physiological environments. For example, pH-sensitive polymers can degrade in the acidic environment of endosomes, facilitating intracellular antigen release, which is particularly important for activating MHC-I cross-presentation. Enzyme-sensitive peptides can also be used to design nanoparticles that release antigens upon cleavage in enzyme-rich microenvironments. Moreover, adjusting the crystallinity, molecular weight, or crosslinking density of materials allows precise control over release kinetics, ranging from days to weeks. Regarding stability, nanoparticle formulations can effectively protect temperature-sensitive antigens, such as mRNA, by providing a stable hydrophobic environment that shields fragile molecules from degradation by ribonucleases during storage and transportation.

Adjuvant Incorporation and Immune Modulation

Incorporating adjuvants into the nanoparticle structure is a powerful strategy for efficient immune modulation. This co-delivery system ensures that both the antigen and adjuvant are taken up by the same antigen-presenting cell, simultaneously activating innate immune receptors and antigen processing pathways to produce a synergistic effect. For example, Toll-like receptor (TLR) agonists like CpG ODN can be encapsulated in nanoparticles alongside antigens, and upon release in the cell, they activate the TLR9 signaling pathway, triggering a strong inflammatory response and upregulation of co-stimulatory molecules. Another approach involves incorporating adjuvants as part of the nanoparticle structure, such as in the case of ISCOM matrices, which are both delivery vehicles and potent adjuvants. Nanoparticles can also co-deliver multiple types of adjuvants to simultaneously activate different immune pathways, such as combining cyclic dinucleotides that activate the cGAS-STING pathway with TLR agonists to polarize specific T cell subsets, enabling fine-tuned regulation of the immune response.

Interested in Advanced Nanoparticle Delivery Strategies?

BOC Sciences provides versatile nanoparticles with diverse compositions and functional modifications, customized solutions for your delivery needs.

Mechanisms of Immune Activation by Nanoparticle Vaccines

The initial step in nanoparticle vaccine efficacy is the uptake of nanoparticles by professional antigen-presenting cells (APCs), primarily dendritic cells (DCs). Due to their small size, nanoparticles can either be directly drained to the lymph nodes via lymphatic vessels or captured by dendritic cells in tissues. The pattern recognition receptors (PRRs) on the surface of dendritic cells can identify specific physical and chemical features of nanoparticles, perceiving them as "danger signals" and triggering active uptake mechanisms such as phagocytosis or macropinocytosis. Once internalized, the nanoparticles enter the endosomal-lysosomal pathway, where they are degraded in acidic environments and by enzymes, releasing the encapsulated antigens. These antigens are then processed into short peptides and bound to major histocompatibility complex (MHC) molecules. The MHC-II complexes are presented on the surface of the dendritic cell, activating CD4+ helper T cells, while some antigens are also processed via "cross-presentation" and delivered to the cytosol, where they are degraded by the proteasome and presented by MHC-I complexes to activate CD8+ cytotoxic T cells, essential for eliminating intracellular pathogens and cancerous cells.

Nanoparticles and their encapsulated adjuvants are potent activators of the innate immune response, which serves as the foundation for initiating an effective adaptive immune response. After uptake by dendritic cells, nanoparticles or their adjuvants (e.g., Toll-like receptor agonists) bind to intracellular pattern recognition receptors, triggering the activation of inflammasomes and the secretion of pro-inflammatory cytokines such as interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α). Concurrently, activated dendritic cells upregulate co-stimulatory molecules such as CD80 and CD86 on their surface. These "second signals" are critical for the full activation of naive T cells. Once fully activated, antigen-loaded dendritic cells migrate to the T cell areas of lymphoid organs, presenting the processed antigens to T cells, driving their differentiation into various effector T cells, and stimulating B cells to produce high-affinity antibodies. This process bridges innate immunity to adaptive immunity, facilitating a comprehensive immune response.

The lymph nodes are central sites for immune cell interactions and the initiation of adaptive immune responses. As such, the efficient accumulation of vaccine nanoparticles in the lymph nodes is crucial. Nanoparticles within the optimal size range of 20 to 200 nm are able to migrate through interstitial spaces and lymphatic vessels to the lymph nodes. Nanoparticles that are surface-modified with PEG or hydrophilic materials can minimize non-specific interactions with tissue matrixes, allowing for more efficient transport to the lymph nodes. Upon reaching the lymph nodes, nanoparticles can either be captured directly by resident dendritic cells or be processed by dendritic cells migrating from peripheral tissues. The sustained high concentration of nanoparticles in lymphoid organs significantly increases the likelihood of antigen encounter with rare antigen-specific T and B cells, thus efficiently triggering immune responses.

The success of a vaccine is not only determined by the induction of a strong primary immune response but also by the establishment of long-lasting immune memory. Nanoparticle vaccines facilitate the formation of durable immune memory through multiple mechanisms. The controlled antigen release from nanoparticles provides continuous antigen stimulation, which is crucial for the maturation of germinal centers and affinity maturation of B cells, leading to the generation of long-lived plasma cells and memory B cells. These cells are capable of persistently secreting high-affinity antibodies or rapidly responding to reinfection with the same pathogen. Additionally, efficient cross-presentation and strong co-stimulatory signals promote the differentiation of central memory and effector memory T cells, which circulate in lymphoid organs and peripheral tissues, providing fast and multi-layered defense. The precise spatiotemporal control over the delivery of both antigens and adjuvants by engineered nanoparticles lays a solid foundation for generating high-quality, long-lasting immune memory.

Types of Nanoparticle Platforms for Vaccine Delivery

Lipid-Based Nanoparticles (LNPs) and Liposomes

LNPs are a class of nanocarriers made from lipids, with liposomes being the most well-known form. Liposomes possess a phospholipid bilayer structure, wherein the hydrophilic core and hydrophobic layer enable the encapsulation of antigens with varying properties, such as water-soluble proteins or hydrophobic immunomodulatory molecules. The next generation of lipid nanoparticles features more complex structures, typically composed of ionizable lipids, phospholipids, cholesterol, and PEG-lipids. Ionizable lipids, which carry a positive charge in acidic environments, allow for the efficient encapsulation of negatively charged nucleic acid-based antigens, such as mRNA. Upon entering cells, these nanoparticles revert to a neutral state at physiological pH, reducing toxicity, and regain a positive charge in the acidic environment of endosomes, promoting membrane fusion and antigen release. Due to their excellent biocompatibility, versatility in drug loading, and ease of large-scale production, lipid-based systems have become a critical platform for current vaccine technologies.

Polymeric Nanocarriers for Antigen and Adjuvant Co-Delivery

Polymeric nanocarriers offer a highly customizable platform for the co-delivery of antigens and adjuvants, enabling precise control over the release profiles. These carriers are typically composed of biocompatible or biodegradable polymer materials such as poly(lactic-co-glycolic acid) (PLGA), poly(lactic acid) (PLA), and polycaprolactone (PCL). Nanoparticles or micelles encapsulating both antigens and adjuvants can be fabricated through methods like emulsion solvent evaporation or nanoprecipitation. The key advantage of polymeric carriers lies in their tunable release kinetics. By adjusting the polymer's molecular weight, crystallinity, or monomer ratio, antigens can be released either rapidly or over an extended period of weeks, mimicking the effects of multiple immunizations. For instance, polymers with pH-sensitive linkers can be used to design "smart" carriers that degrade and release their cargo only in the acidic environment of antigen-presenting cells' endosomes, thereby efficiently activating cellular immune responses.

Inorganic Nanoparticles and Hybrid Nanostructures

Inorganic nanoparticles, such as mesoporous silica nanoparticles (MSNs), gold nanoparticles, and iron oxide nanoparticles, have shown great promise in vaccine delivery due to their unique physical properties. Mesoporous silica nanoparticles possess an exceptionally high surface area and ordered pore structure, which allows for the physical adsorption or covalent coupling of large amounts of antigen molecules, thereby achieving high payloads. Gold nanoparticles, with their controllable size and shape, exhibit surface plasmon resonance effects that can be leveraged to enhance antigen release from endosomes via photothermal effects, thereby boosting immune responses. However, the relatively poor biodegradability of inorganic materials often limits their use. To address this, hybrid nanostructures have been developed. For example, mesoporous silica nanoparticles can serve as a core to efficiently load antigens, with a biodegradable polymer coating used to control release rates. Similarly, gold nanoparticles can be integrated into lipid-based carriers to combine the biocompatibility of lipids with the functional capabilities of inorganic materials, creating synergistic delivery systems with enhanced performance.

Virus-Like Particles and Biomimetic Nanoplatforms

Virus-like particles (VLPs) are self-assembled structures formed by one or more viral proteins, which mimic the highly organized and repetitive surface topology of the virus. Despite lacking viral genetic material, VLPs are efficiently recognized by the immune system, triggering robust B cell and T cell responses. This platform is particularly suitable for presenting target antigens. A prime example of the successful application of VLPs is the human papillomavirus (HPV) vaccine. Biomimetic nanoplatforms take inspiration even further by directly utilizing elements derived from natural biological systems. For instance, exosomes or cellular membrane fragments can be used as vaccine carriers. Exosomes, which are naturally secreted nanovesicles, carry the recognition signals of their parent cells, enabling them to facilitate intercellular communication and material transfer. By employing genetic engineering techniques to express antigens on the surface of exosomes or fuse them with synthetic nanoparticles, high-performance vaccine carriers can be designed that combine both natural characteristics and artificial functionalities.

Table 1. Nanoparticle Vaccine Delivery Products from BOC Sciences.

Product Name Description Applicable Scenarios Inquiry
Lipid Nanoparticles (LNPs)Lipid-based nanocarriers, particularly suitable for encapsulating mRNA or water-soluble proteins, with good biocompatibility and controlled release properties.mRNA vaccine delivery, especially for combating viruses that mutate rapidlyInquiry
Gold NanoparticlesAdjustable particle size and shape, can enhance immune response through surface plasmon resonance effects. Suitable for efficient antigen release and delivery.Nanovaccine design, particularly antigen release combined with thermal effectsInquiry
PLGA Nanoparticles Biodegradable polymer-based nanoparticles suitable for antigen encapsulation. Commonly used in vaccine delivery, providing sustained antigen release.Vaccine delivery, especially antigen encapsulation and controlled release systemsInquiry
ISCOM Matrix Nanoparticles made of lipids and sugars with potent immune-enhancing effects, often used for co-delivery of antigens and adjuvants.Immuno-enhancer delivery, vaccine adjuvant deliveryInquiry
Virus-Like Particles (VLPs) Self-assembling nanoparticles that mimic viral surface structures without a viral genome. Serve as an efficient antigen display platform.Cancer immunotherapy, vaccine design (e.g., HPV vaccine)Inquiry
Mesoporous Silica Nanoparticles (MSNs) High surface area, porous structure, suitable for high antigen payloads. Often used for antigen adsorption and co-delivery.High payload vaccines, co-delivery of antigens and adjuvantsInquiry

Analytical Techniques for Nanoparticle Vaccine Characterization

Accurate physical characterization of nanoparticle vaccines is fundamental for ensuring both quality control and reproducibility. The particle size and distribution directly influence the nanoparticle's lymphatic drainage and cellular uptake behavior in vivo. Dynamic light scattering (DLS) is commonly used to rapidly measure the hydrodynamic diameter and polydispersity index (PDI) of particles in solution, providing insights into the uniformity of the formulation. Laser diffraction is also employed for broader particle size analysis. To visualize particle morphology, transmission electron microscopy (TEM) and scanning electron microscopy (SEM) offer high-resolution imaging, allowing for detailed assessment of nanoparticle shape, whether spherical, rod-like, or other complex structures, and surface smoothness. Atomic force microscopy (AFM) can provide three-dimensional surface morphology and roughness data under conditions close to their natural state. Combined, these techniques enable a comprehensive evaluation of the physical stability of nanoparticles and provide crucial parameters for understanding their biological behavior.

The integrity of the antigen during preparation and storage is critical for vaccine potency. Encapsulation efficiency refers to the percentage of the total input antigen that is successfully loaded into or adsorbed onto the surface of the nanoparticle. This is typically determined by separating free antigens from the nanoparticle-bound antigens using methods such as ultracentrifugation, gel filtration chromatography, or dialysis. The antigen content in the supernatant can then be quantified using techniques like BCA protein assays, ELISA, or high-performance liquid chromatography (HPLC), which provide indirect estimates of encapsulation efficiency. For evaluating antigen stability, it is essential to assess whether its conformation has changed within the nanoparticle. Circular dichroism (CD) and fluorescence spectroscopy are commonly used to analyze the secondary and tertiary structure of the antigen. Techniques such as sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and size-exclusion chromatography are used to detect antigen degradation or aggregation. These analyses ensure that nanoparticles effectively protect the antigen and prevent its inactivation before reaching the target site.

Simulating the in vivo environment to study the release of antigens from nanoparticles is crucial for predicting the strength and duration of the immune response. In vitro release studies typically involve placing nanoparticles in release media that mimic physiological conditions, with samples collected at predetermined time points to measure the amount of antigen released using previously mentioned analytical methods. Release curves can be plotted to assess the release kinetics. To more accurately replicate the intracellular environment, the release media may be adjusted to different pH values or supplemented with specific proteases to evaluate pH-sensitive or enzyme-sensitive nanoparticle-triggered release properties. Additionally, stability studies accelerate the evaluation process by storing nanoparticles under stress conditions, such as high temperature or exposure to light, and periodically measuring changes in particle size, encapsulation efficiency, and antigen activity. This helps predict the nanoparticle vaccine's storage stability and shelf life under standard conditions.

Table 2. BOC Sciences Nanoparticle Vaccine Development & Customization Services.

Service Name Description Inquiry
Nanoparticle Vaccine Development and Customization Provides customized nanoparticle platform development services, helping clients select appropriate carrier materials, design antigen loading systems, and optimize vaccine efficacy.Inquiry
Immunomodulation and Adjuvant Co-Development Offers development of antigen and adjuvant co-delivery systems, including optimization of adjuvant selection and co-delivery system design.Inquiry
Nanoparticle Surface Functionalization Services Provides surface modification services for nanoparticles to enhance targeted delivery efficiency, functionalizing nanoparticles for specific immune cells or pathogen targets.Inquiry
Antigen Loading and Release Studies Studies the loading efficiency and release characteristics of antigens within nanoparticles, optimizing vaccine release rates to maximize immunogenicity.Inquiry
Nanoparticle Stability and Storage Optimization Provides stability testing and storage condition optimization for nanoparticles to ensure vaccine efficacy during transport and storage.Inquiry
Preclinical Nanovaccine Research Services Provides support during early-stage research, including immunological mechanism validation, animal model evaluation, and vaccine optimization.Inquiry
Nanoparticle Characterization and Analysis Services Provides comprehensive characterization and analysis of nanoparticle properties, such as size, morphology, and surface characteristics, to ensure that nanoparticle carriers meet R&D requirements.Inquiry

Before in vivo testing, in vitro cell models are employed to study the interactions between nanoparticles and immune cells, helping to predict immunogenicity and optimize design. By co-incubating nanoparticles labeled with different fluorescent markers with dendritic cells or macrophages, flow cytometry can quantify cellular uptake efficiency, distinguishing between endocytosis and surface adsorption. Confocal microscopy offers visual confirmation, revealing the intracellular localization of nanoparticles, such as whether they escape from endosomes. To assess the ability of nanoparticles to activate immune cells, the activation state of antigen-presenting cells can be monitored after exposure to nanoparticles. Flow cytometry can measure the expression levels of surface co-stimulatory molecules, while enzyme-linked immunosorbent assays (ELISA) are used to detect cytokine secretion in cell culture supernatants. These in vitro studies provide direct evidence of the mechanisms through which nanoparticle vaccines activate immune responses.

Applications and Research Directions

Infectious Disease Vaccine Development Models

Nanoparticle technology provides an innovative platform for addressing infectious diseases that traditional vaccine strategies struggle to control. In the realm of viral infections, lipid nanoparticle-based mRNA vaccines have proven effective in combating rapidly mutating pathogens. This platform is capable of encoding key antigens, such as the spike protein of the SARS-CoV-2 virus, which is then expressed in vivo to induce immune responses. For highly mutable viruses like influenza, researchers are designing nanoparticle vaccines that display multiple conserved antigens from different serotypes, eliciting broad-spectrum neutralizing antibodies and circumventing the need for annual updates. In the fight against complex pathogens like malaria and HIV, nanoparticles are being used to present fragile multi-protein conformational epitopes, such as presenting the Plasmodium falciparum circumsporozoite protein with an adjuvant on the surface of virus-like particles, aiming to induce protective immune responses that have previously been difficult to generate.

Cancer and Immune Therapy Research

In cancer immunotherapy, nanoparticle vaccines are being explored as promising personalized treatment strategies. The fundamental concept is to activate the patient's immune system to recognize and attack tumor cells. By loading nanoparticle carriers with tumor-specific neoantigens identified through sequencing, personalized neoantigen vaccines can be created that efficiently prime T cell responses against these neoantigens. Nanoparticles can also be used to deliver tumor-associated antigens, such as Melan-A or MUC1, in combination with potent adjuvants to break immune tolerance. Additionally, combining nanoparticle vaccines with immune checkpoint inhibitors is a current area of significant interest. Nanoparticles can trigger and expand T cell populations within the tumor microenvironment, while checkpoint inhibitors can relieve the immune suppression imposed by the tumor, resulting in enhanced anti-tumor effects. This synergy holds great promise in improving therapeutic outcomes.

Veterinary and Environmental Immunology Studies

Nanoparticle vaccines are not limited to human medicine and are showing considerable potential in veterinary and environmental immunology. In livestock, nanoparticle-based vaccines for high-impact infectious diseases, such as foot-and-mouth disease and avian influenza, are under development, offering faster and stronger protection than traditional inactivated vaccines. This approach could significantly reduce economic losses and reliance on antibiotics. In aquaculture, nanoparticle oral vaccines targeting bacterial pathogens like Aeromonas hydrophila can resist gastrointestinal degradation, providing mucosal immunity and improving fish health. In the emerging field of environmental immunology, researchers are investigating the use of nanoparticle vaccines to control infectious diseases in wildlife populations. For instance, edible nanoparticle vaccines could be distributed to control rabies in fox populations or manage white-nose syndrome in bats, providing an ecologically friendly tool for disease management.

Summary

Nanoparticle vaccine delivery systems, with their unique physicochemical properties, demonstrate tremendous potential in immune activation and antigen delivery. Compared to traditional vaccine formulations, nanoparticles more effectively mimic the natural structure of pathogens, enhancing the immune system's recognition and processing, thereby significantly improving the specificity and durability of the immune response. Furthermore, the design of nanoparticles allows for the co-delivery of antigens and adjuvants, further enhancing immune effects, particularly in activating cellular immunity, which is crucial for addressing challenging diseases such as cancer and chronic infections. Although nanoparticle vaccine delivery technology still faces some challenges, as research deepens and technology advances, nanoparticle platforms are poised to become a key tool in the development of future vaccines, driving the innovation and application of next-generation vaccines. In this context, BOC Sciences, as a leading biotechnology company, is dedicated to providing advanced nanomaterials and vaccine delivery solutions, supporting the continuous progress and innovation in vaccine development and the biopharmaceutical industry.

References

  1. Image retrieved from Figure 1 "Schematic representation of different nanoparticle delivery systems", Gregory, Anthony E., et al., 2013, used under [CC BY 3.0] (https://creativecommons.org/licenses/by/3.0/). The title was changed to "Schematic of various nanoparticle vaccine delivery systems".
  2. Gregory, Anthony E., et al., Vaccine delivery using nanoparticles., Frontiers in cellular and infection microbiology 3 (2013): 13. https://doi.org/10.3389/fcimb.2013.00013
  3. Aikins, Marisa E., et al., Engineered nanoparticles for cancer vaccination and immunotherapy., Accounts of chemical research 53.10 (2020): 2094-2105. https://doi.org/10.1021/acs.accounts.0c00456
* Please kindly note that our services can only be used to support research purposes (Not for clinical use).
Online Inquiry
Verification code