View Article

Abstract

Vaccination enormously contributes to maintain global health. Resistance for infectious diseases is growing quickly during treatment. Vaccines alone are not able to give enough strong and long-lasting protection against infections. To overcome this nanovaccines are developed. Nanoparticle (NP)-based delivery vehicles, such as dendrimers, liposomes, micelles, virosomes, nanogels, and microemulsions, offer numerous advantages over traditional vaccine adjuvants. The nanovaccines (50–250 nm in size) are considered most efficient in terms of tissue targeting, staying in the bloodstream for a long time. Nanovaccines are proved to in improving antigen presentation, targeted delivery, stimulating body’s innate immune system, and a strong T-cell response which helps in fighting against infectious diseases and cancers. Also,nanovaccines are very helpful for making cancer treatments that use immunotherapy. So, this review highlights the various types of NPs used in the new paradigm in viral vaccinology for infectious diseases and a brief comparison of conventional vaccines and nanovaccines. It focuses on the current NP-based vaccines, their potential as adjuvants, their potential in inducing innate and humoral immunity and the ways they can be delivered to cells. It also provides an overview that in future nanovaccines can be modified in order to attach more antigens and to get maximized therapeutic effect.

Keywords

Vaccine, Nanotechnology, Targeted Delivery, Immunity, Nanocarrier.

Introduction

The preparation of pathogen containing either disabled or killed form of virus or a part of the pathogen's structure is called vaccine. It which helps in promoting the formation of antibodies or cellular immunity against the pathogen when administered1.An idea of vaccination has changed over ages and this has led to a wide variety of vaccinations. Vaccinations include live viruses in attenuated form, inactivated pathogens, inactivated toxins, or pathogen parts like sub-unit and conjugate vaccines can all be included2. As far as public health vaccination offers a reliable line of defense against infectious disorders by delivering specific agents, known as Antigens, into the body3 which initiates the immune response and produce adaptive immunity.

Nanovaccines

Nanovaccines made of nanoparticles, are an emerging class of vaccines. In nanovaccines, carry vaccine proteins are carried to the body very small particles that mimic the structures of pathogens and boost the immune system. Nanovaccines contain already determined antigen which is attached to a nanomaterial and an adjuvant and this antigen is responsible for making the immune system to react2. Nanovaccines surface may contain number of antigen epitopes. Nanovaccines induce both humoral and cell-mediated immune responses which makes them more effective than traditional vaccinations. COVID-19, HIV, Merkel Cell Polyomavirus, influenza A virus, and a lot of other vaccines are made with nanoparticles.

Advantages Of Nanovaccines4

Nanovaccines offer several advantages over conventional vaccines.

a) Nanovaccines provide site-specific targeting which help in minimizing the potential aftereffects.

b) Nanovaccines are able to activate of the immune cells and ultimately, immune-stimulatory response even if administered in small amount.

c) Nanovaccines have potential to protect the antigen from protease degradation thus, increasing the amount of antigen available for initiating immune response.

d) Soluble antigens are less effective at eliciting protective immunity because of their inability to be endocytosed by cells, so nanovaccines help in improving antigen immunogenicity by providing targeted antigen delivery to Antigen Presenting Cells (APCs).

e) Nanovaccines have potential to distribute and release of the antigens to the site of administration in precisely controlled manner which in turn diminish the need for booster doses.  

f) Nanovaccines contain Toll-like receptor (TLR) ligands and agonists as immune-stimulatory chemicals which improve adjuvanticity substantially.

g) Nanovaccines help in loading antigens onto major histocompatibility complex (MHC), therefore improving cross-presentation.

h) Nanovaccines provide controllable drug release rates.

i) Nanovaccines offer better stability as compared to conventional vaccines.

j) Nanovaccines can easily enter the cellular mechanism by endocytosis process because of the similarity in size to numerous cellular components and thus, they can elicit a long-lasting and durable response.

Table 1: Advantages and Disadvantages of Nanovaccines

Advantages

  • Dose required is low and thus producing low side effects          
  • Protecting and stabilizing antigens in the extracellular or intracellular environment                
  • Antigen immunogenicity is improved.
  • Enhanced immune response and reducing the need for booster doses.  
  • Precisely controlled antigen release and targeting antigens to APCs.    
  • Other immune-stimulatory molecules can be incorporated as necessary.                           
  • Better stability.    
  • Long-lasting response.

Disadvantages

  • Difficulty with bulk production.
  • Short shelf life.
  • Obtaining uniform size is not possible.   

Requirements for Nanovaccines4

In order to employ nanovaccines for enhancing immune responses, several conditions must be fulfilled.

i) The most crucial requirements for Nanovaccines are safety, minimal reacto-genicity and production of  selective and sustained immune response which will result in improved memory responses. This response is the outcome of controlled release from nanovaccines.

ii) MHC class I molecules must do cross-presentation on Dendritic Cells (DCs).

iii) Cost effective and bulk production should be feasible.

Properties of Nanovaccines5:

i) Nanovaccines possess the size-dependent properties which have crucial role in bio-distribution and immune-modulation within the body. For Example, nano-sized particles of size 146 nm generated a stronger response in mice as compared to 64-nm-sized particles.

ii) Because of their nano size, it is easier for them to easily penetrate cells and distribute.

iii) Nanovaccines trigger the production of antibodies when nano-sized particles and their conjugates are considered as foreign substances by B-cells. So, formation of antibodies is affected by composition of nanovaccines, surface coating, absorption, and processing by immune system cells.

iv) Another crucial factor that determines efficacy and effectiveness of vaccination is an adjuvanticity of nanovaccines. The studies reveal that nanovaccines stimulate the Antigen-Presenting Cells (APC) to enhance the uptake of antigens.

v) Nanovaccines specifically target immune cells or organs. Targeting can be done by adding ligands or antibodies to the nanocarrier surface, particular receptors on immune cells can be bound. Targeting of vaccine helps in boosting the effectiveness of the vaccination and reducing the side effects.

Composition of Nanovaccines:

Nanovaccines consist of three main components:

(1) Antigens,

(2) Adjuvants,

(3) Nano carriers.

Antigens and Adjuvants:

The source of antigens can be from natural tumor antigens, synthetic peptides, mRNA, and DNA encoding tumor antigens. Adjuvants are added to enhance the immunogenicity of vaccine antigens, the without compromising vaccine safety. Adjuvants are able to produce a more robust immune response in combination with a specific antigen. Thus, nanoparticles can deliver either antigens or adjuvants to the targeted cells at predetermined rates3. In conventional vaccines, antigens and adjuvants are separate components, which sometimes lead to wastage of adjuvants, reduced vaccine immunogenicity, and an increased risk of adverse immune reactions. Nanoparticle-based delivery vehicles can facilitate the co-delivery of antigens and adjuvants as a whole, which significantly enhances the efficacy, safety of vaccines and achieve targeted delivery of vaccine.

Nanocarriers:

Nanocarriers are promising component of nanovaccine that can carry single or multiple therapeutic agents, thus increasing the solubility, stabilizing therapeutic activity in vivo, enhancing the aim to deliver the drug at desired site, and reducing drug- related side effects. However, some carriers possess adjuvant properties, which aid in activating the immune system and enhancing immune responses, thereby improving the efficacy of vaccines. Liposomes, polymer nanocarriers, inorganic nanocarriers, and exosomes nanocarriers are among the commonly used nanocarriers.

Mode of action of Nanovaccines6:

The nanovaccine can be coated with specific ligands or antigens such as Fc receptor, C-type lectin receptor, and anti-DEC-205 which actively get attached to the Dendritic Cells (DCs) receptors. The addition of immune-stimulatory molecules are to nanoparticles could increase the immunogenicity and improve the targeted delivery of antigens. The most prominent immune-stimulatory chemicals are mannose, β-Glucans, chitosan, glycolipids, and saponins. Another important consideration is the stability of the antigen in the vaccine formulation because modern antigens are vulnerable to enzymatic, pH, and temperature degradation, especially peptides, proteins, and nucleic acids. These are degraded before their presentation to antigen-presenting cells (APCs). There is need to develop as delivery systems which provide them protection against these degrading agents and antigen can be delivered to (APCs) efficiently. The mode of action of nanovaccines involves a series of steps that enhance antigen delivery, its processing, immune system activation, and long-term protection. Here's a breakdown:

1. Targeted Delivery & Uptake

The purpose of nanovaccines is to transport adjuvants and antigens to immune cells. They are engineered in such a way that they can be efficiently up taken by antigen-presenting cells (APCs), such as dendritic cells and macrophages, by process phagocytosis or endocytosis.

2. Antigen Processing & Presentation

The uptake of nanovaccine by antigen-presenting cells (APCs) causes release of nanovaccines in a controlled manner and is followed by processing and presentation of the antigens on Major Histocompatibility Complex (MHC) molecules and it allows T-cells to recognize the pathogen and start the immune activation.

MHC-I Pathway → Activates CD8+ T cells, leading to cytotoxic responses.

MHC-II Pathway → Activates CD4+ T cells, helping B cell activation.

3. Activation of Immune Responses

  • CD8+ T Cells → Differentiate into Cytotoxic T Lymphocytes (CTLs), which destroy infected cells.
  • CD4+ T Cells → Stimulate B cells, enhancing antibody production.
  • B Cells → Differentiate into plasma cells, secreting antigen-specific antibodies.

4. Induction of Memory Immunity

Some T and B cells develop into memory cells. Thus, providing assurance for a prompt and powerful immune response upon future exposure to the same virus.

5. Immune Modulation & Adjuvant Effect

Nanoparticles have the ability to function as adjuvants, improving immune activation, and they can be engineered to release antigens slowly, offering sustained stimulation.

Figure 1 Schematic Diagram for Cross-Presentation of Exogenous Antigen of Nanovaccine to Dendritic Cells4.

How nanovaccines induce immunity7:

The use of nanotechnology helps nanovaccines to induce immunity by improving the distribution and presentation of antigens to the immune system. These nanoparticle-based vaccines help in
augmenting primary and secondary immune responses. Nanovaccines achieve this
enhancement through various mechanisms, including activation of innate, cellular, and
humoral immunity.Here's a more detailed breakdown of how they induce immunity:

Activation of innate immunity

Nanovaccines have pathogen-associated molecular patterns (PAMPs) on their surface to interact with immune cells. PAMPs act as ligands for pattern recognition receptors (PRRs) which are abundantly present in these immune cells. Consequently, PAMP-PRR interactions trigger endocytosis, with larger particles being engulfed by macrophages while smaller particles are
engulfed by dendritic cells (DCs). Nanovaccine particles can be modified for enhancing their survival from macrophage degradation and to facilitate their direct delivery to APCs. Moreover, the type of PAMPs present influences cytokine and chemokine secretion by neutrophils and macrophages, which further help in activation and maturation of APCs, thereby initiating strong cellular and humoral immune responses.

Activation of cellular immunity

Following vaccination, cellular immunity is boosted, resulting in the development of immunological memory and pathogen neutralization. Once activated,dendritic cells after binding to nanovaccine particles migrate to the lymphatic organs to start this process. Through MHC class I receptors, activated DCs present antigens to CD8+ CTLs, triggering potent cell-mediated immune responses and the apoptotic destruction of target cells. Additionally, activated DCs use the MHC-II receptor to deliver antigens to CD4+ T-helper helper (Th) cells. The cells released cytokines are used to classify them into Th1 and Th2 subsets. The Th1 fraction mostly generates pro-inflammatory cytokines, which promote the growth of CTLs and strengthen cell-mediated immunity. In humoral immunity, the Th2 cell subset secretes a different class of cytokines that stimulate the generation of B-cells. The total preventive or therapeutic potential of a proposed nanovaccine is greatly influenced by the delicate balance between Th1/Th2 activity. In certain situations, nanovaccines may work by inhibiting T-regulatory (Treg) cells, which inherently prevent the body's effector T cells from activating and proliferating.

Activation of humoral immunity

Nanovaccines can induce strong Th2 cytokine responses that stimulate B cells in the lymph nodes and spleen. B cells recognize soluble antigens and undergo proliferation in the germinal center. Activated B cells become either antibody-secreting plasma cells producing soluble antibodies or memory cells providing immunity for future encounters with the same antigen. Due to the short lifespan of plasma cells, antibody titers gradually decrease over time. Memory cells that are kept in the bone marrow or lymphatic organs become active and offer defense against reinfection with the same antigen. Memory B cells proliferate quickly and change into cells that secrete antibodies, mostly IgG antibodies that fight the antigen. Likewise, memory T cells, such as CD4+ and CD8+ cells, enhance humoral and cellular immune responses by helping to produce more cytokine and chemokine signals. However, memory B and T cells may not be able to produce adequate protection if the antigen (epitope) undergoes major structural alterations. Even though the initial vaccination can offer about 90% protection, the residual 10% may still have negative consequences, requiring booster shots to reach 100% protection.

Figure 2:  Activation of cellular and humoral immune responses by nanovaccines

Types of Nanovaccines

Nanovaccines are the vaccines of today’s age which are designed for enhancing the immune response by providing delivery of antigens in a controlled and targeted manner. Nanovaccines are classified on the basis of various parameters such as composition, type of construction, shape, and their mode of action against desired diseases. Nanovaccines employ nano sized particles, which are of size range from 1 to 100 nanometers and help to carry the vaccine components to specific cells in the body8.

i) Organic NPs9

Organic NPs are made of organic molecules of natural or synthetic origin. Most organic materials used are preferred because they are biodegradable, nontoxic, and biocompatible. These organic NPs offer advantages such as antigens' and adjuvants' potential to self-fabricate under physiologically favorable circumstances and their versatility in accommodating a broad range of sizes, compositions, surface modifications, and modalities. This organic NP vaccine delivery platform can be easily modified, such as virus-like particles (VLPs) and liposomes, which are polymeric NPs (PNPs).

a) Virus-like particles (VLPs) are a type of nanoparticle that closely mimic the structure of viruses but are non-infectious in nature due to lack of genetic material. VLPs vaccines have antigens on their surface, which stimulate an immune response without causing disease. Various examples of VLP-based vaccines including human papillomavirus (HPV),hepatitis B, influenza, norovirus, and coronavirus.

VLP vaccines have several advantages over traditional vaccines, which include:

*Safety: VLP vaccines are non-infectious because they do not contain genetic material which makes them safe for use in humans,

*Efficacy: Due to the antigens are present in highly structured and repetitive manner replicating the natural structure of viruses, VLP vaccinations elicit a potent and sustained immune response.


*Versatility; VLPs can be modified to display multiple antigens on their surface, which can help in enhancing the immune response and providing protection against multiple strains or types of viruses.

*Stability: VLP vaccines are stable at a wide range of temperatures, thus facilitating storage and distribution.

Figure 3: Schematic representation of Virus-like particle

b) Polymeric Nanoparticles (PNPs)6

Polymeric nanoparticles are that type of vaccine delivery system where antigens and adjuvants are encapsulated in a biocompatible and biodegradable polymer matrix. PNPs are divided into two main categories: natural PNPs and synthetic PNPs. Polymers are derived from biological sources in natural PNPs such as proteins, polysaccharides, and lipids, while in synthetic PNPs synthetic polymers are used for encapsulation. Natural polymers are more preferred over synthetic polymers as they are biodegradable, nontoxic, biocompatible and less likely to cause adverse reactions or long-term toxicity. Poly (lactic-co-glycolic acid) (PLGA), polyethyleneimine (PEI), and polypropylene sulphide (PPS) are examples of synthetic polymeric NPs that are used to make vaccines.Examples of natural polymeric NPs used in vaccine development include chitosan, alginate, and silk fibroin. But on the other hand, highly effective vaccine delivery systems can be created by carefully controlling the size, shape, and surface characteristics of synthetic PNPs. Nevertheless, they could be more hazardous and have a larger chance of causing immunological responses.

Advantages of PNPs over conventional vaccine delivery methods:

  1. Enhanced antigen storage,
  2. Protection against deterioration,
  3. Controlled antigen and adjuvant release,
  4. Capacity to target certain cells or tissues.

Figure 4: Schematic representation of Polymeric Nanoparticles

c) Liposomes9:

Liposomes are a special type of organic NPs which are made up of biodegradable phospholipids which get self-assembled into a lipid bilayer on contact with water. This lipid bilayer on the surface of a particle makes the particle more stable. That’s why liposomes are mostly impermeable like salts and macromolecules. Liposomes have ability to hold both hydrophobic and hydrophilic molecules, like antigenic proteins and peptides, in their lipid bilayer and aqueous core, respectively. Liposomes can protect the vaccine against degradation. Moreover, liposomes are able to carry single or multiple hydrophilic and lipophilic antigens and provide controlled release of antigens. Liposomes also show enhanced cellular uptake and improved antigen- specific immune response.

Figure 5: Schematic Representation of Liposomes

ii) Lipid nanoparticles (LNPs)6:

LNPs consist of a lipid bilayer that surrounds an aqueous core.LNP can be encapsulated to protect nucleic acids, such as mRNA or DNA, from degradation and thus enables a longer-lasting immune response. LNPs possess the ability to target particular cells, such as immune cells or liver cells, is one benefit of them. Lipid-based nanoparticles are the most preferred drug delivery system because they are biocompatible, self-assembled, have a substantial drug capacity, provide enhanced drug availability, are inexpensive, easy to create and release can be controlled. However, LNPs possess some potential limitations. LNPs can induce immune responses and cause inflammation, which can limit their effectiveness. Moreover, manufacturing of LNPs can be difficult and expensive on a large scale.

Figure 6: Schematic representation of Lipid Nanoparticles

iii) Inorganic Nanoparticles10:

Inorganic NPs find their benefits for vaccine distribution due to their hard structure and predictable fabrication; however, they are predominantly non-biodegradable. The main inorganic NPs are carbon, silica,calcium phosphate, aluminum-based, gold and magnetic nanoparticles. Size and shape of an inorganic nanoparticle affects vaccine delivery. It is easy to synthesize Gold nanoparticles (AuNPs) are easily into a variety of shapes (spherical, rods, cubic, etc.) which can induce humoral and cellular responses. Examples of gold nanoparticles are DNA vaccine adjuvants for human immunodeficiency virus (HIV) or as carriers for antigens from viruses, such as influenza and foot-and-mouth disease. Nanotubes and Mesoporous spheres are synthesized from Carbon nano-particles (CNTs) and are able to enhance the IgG response. Carbon nanoparticles get conjugated to multiple copies of protein and peptide antigens. Silica-based nanoparticles are considered another promising nano-carrier in vaccine delivery because of their ability to target a selective tumor and real-time imaging. The structural properties of nanoparticles can be changed to modify their interaction with cells while their synthesization. Calcium phosphate nanoparticles are another type of inorganic nanoparticle which is formed on mixing calcium chloride, dibasic sodium phosphate, and sodium citrate under certain conditions. They are nontoxic and show excellent biocompatibility and can be formed in size range between 50 and 100 nm. They are useful as adjuvants for DNA vaccines and mucosal immunity.

Figure 7: Schematic representation of Gold Nanoparticles

Iron Oxide Nanoparticles Due to their distinct magnetic characteristics, iron oxide nanoparticles function exceptionally well and offer a great deal of promise for usage in biological applications including targeted medication administration and magnetic resonance imaging. Iron oxide nanoparticles can be used as immune adjuvants to improve antigen processing or as a delivery mechanism to transfer antigens to the immune system. Magnetic nanoparticles can be coated with single layer ligands, polymers, mixtures of polymers and biomolecules (phospholipids and carbohydrates), or inorganic materials (silica and gold) to stop them from oxidizing and aggregating after manufacture. In addition to stabilizing nanoparticles in solution, these coatings help different biological ligands—such as proteins, antibodies, transferrin, and folate—bind to the surface of the nanoparticles, which is necessary for medicinal applications. These ligands can be chemically connected to magnetic nanoparticles coated with polymers, giving them the ability to target. Iron oxide nanoparticles can therefore be used in nanovaccines.

Figure 8: Schematic representation of Iron Oxide Nanoparticles

iv) Emulsion: Emulsion vaccines employ an oil-in-water emulsion for the delivery of the antigen. An emulsion is typically composed of an oil phase containing an antigen, and water phase contains a surfactant and other adjuvants that enhance the immune response. These vaccines show improved stability and uptake of the vaccine antigen that leads to a stronger and longer-lasting immune
response. Various examples of emulsion vaccines include influenza, hepatitis B, and human papillomavirus (HPV).

v) Exosomes11: Exosome-based vaccine delivery systems are an emerging field of research for the development of vaccines against infectious diseases. Exosomes are small, membrane-bound vesicles released by cells for intercellular communication. They contain various bioactive molecules, including proteins, lipids, and nucleic acids, that can be modified and engineered to deliver therapeutic agents, including vaccine antigens. Exosome membranes can easily bind to target cells and thus increase the bioavailability of the loaded drugs. The phospholipid bilayer structures of exosomes help in enabling stable drug transport, extending drug half-life during delivery, and protecting drugs from enzymatic degradation that’s why exosome-based vaccines can be stored easily if cold storage is not available. Exosome-based vaccine delivery systems are more preferred over traditional vaccine delivery methods because exosomes are biocompatible and biodegradable which makes them safe for use in the human body. Because of their small size they are able to penetrate tissues and cross biological barriers, such as the blood-brain barrier. Moreover, they are immunogenic and are able to stimulate an immune response to the vaccine antigen.

Figure 9: Schematic representation of Exosomes

vi) Dendrimers: Dendrimers are well-defined synthetic globular macromolecules that possess various chemical and biological properties. Chemical properties include the presence of various functional groups on the surface that can be easily accessed for linking the molecules and these surface functional groups can be changed to precisely change the properties. This in turn will help in controlling their interaction with bio-membrane and their bio-distribution. Dendrimers bears adjuvant properties that provide multivalent scaffolds to produce highly defined conjugates with small molecule immune-stimulators and/or antigens.

Figure 10: Schematic representation of Dendrimers

vii) Self-assembling NP9:

These vaccines are synthesized from protein complexes. These proteins get themselves arranged into complex three-dimensional structures in water. SANPs possess advantages of self-assemblies and better over antigen shape. One application of this science is in SANPs.

Figure 11: Schematic representation of SANPs

viii) Protein/peptide NPs9:

Virus capsid proteins are used to synthesize protein/peptide NP vaccines and these proteins have ability to get self-assembled. They are considered copy of their parent viruses, but they lack any genetic material and are not able to spread disease. These are considered as “viruslike particles” (VLPs) which can be enveloped (eVLP) or not enveloped (non-eVLP),

Figure 12: Schematic representation of Protein/peptide NPs

Table 2. Roles of different types of NPs in vaccine development

 

Types

Vaccine NPs

Roles in vaccine development

Disease cured

 

 

 

 

 

 

 

 

Organic NPs

PNPs

Provides better immunogenicity by modifying surface proteins, biodegradable and targeted antigen delivery

Hepatitis B virus (HBV), malaria,Ebola, and Mycobacterium

tuberculosis (M.tb)

Liposomes

They protect vaccine against degradation,
able to carry hydrophilic and lipophilic
antigen, control the release of antigen, increased cellular uptake, and improved antigen- specific immune response

HBV, hepatitis A virus (HAV),Human Immunodeficiency Virus(HIV), influenza A viruses (IAVs),

and influenza B

viruses (IBVs)

Dendrimers

Also have adjuvant properties and provide small,highly defined conjugates as immune-stimulators and/or antigens

Ebola; hemagglutinin type 1 and

neuraminidase type 1-influenza A

(H1N1);Toxoplasma gondii

VLPs

The particles copy their parent pathogen, with
high gastrointestinal stability,and possess self-adjuvant properties

Human papillomavirus (HPV),H1N1 IAV,

HIV, H5N1 IAV

 

 

 

Inorganic
NPs

Gold NPs

Increase immune response, act as both

delivery systems and as adjuvants

West Nile virus, foot and mouth disease virus, H1N1 IAV,

Streptococcus pneumonia, and

Burkholderia mallei

Iron oxides

NPs (IONPs)

Activate immune cells, humoral and cellular immune responses and cytokine production

Mycobacterium

tuberculosis, malaria,

HBV

Self-assembled NPs

 

Have multiple binding sites, improved antigen stability and immunogenicity

EBV, malaria

Protein/peptide NPs

 

Have multiple binding sites, improved antigen stability and immunogenicity

HIV, influenza, and malaria

Table 3: Advantages and disadvantages of different nanoparticles in vaccine delivery

 

Types of Nanoparticle Systems

Advantages

Disadvantages

Virus-like
particles (VLP)

  • Devoid of pathogenic agents
  • Immunomodulator shielding reduces off-target effects
  •  Self-adjuvant
     
  • Poly dispersed particle size
  • Limited encapsulation
  • Lack of reproducibility

 

Liposomes

  • Phospholipids have inherent adjuvant  characteristics and is  stable in GI fluids when altered
  • Able to absorb both hydrophilic and hydrophobic antigens
  • Limited antigen loading;
  • Poor stability of GI exposed liposomes;
  • Less stable than polymer particles

Polymeric

  • It is biodegradable
  • Immunogenicity can be easily improve by surface properties modifications
  • Release of antigens at
    targeted sites
  • Premature release of antigens;
  • Low antigen protection;
  •  Inadequate protection against antigens

 
 

Inorganic
nanoparticles

  • Improved resistance

towards adsorbed antigens

  • There is less chance of an early release;
  •  Surface modification is simple.
  • Non-biodegradable
  • Low aqueous solubility

Emulsion

  • Can take in both hydrophilic and
    hydrophobic antigens
  • Self-adjuvant
  • Poor GI stability
  • Premature release of antigens

Lipid nanoparticle

  • Safe and versatile vehicles for drugs
  • Biodegradable and biocompatible
  • Improved oral drug bioavailability
  • Low loading efficiency
  • Drug expulsion during storage

Exosomes

  • Delivery of vaccine to targeted site
  • Modification is possible by transferring active molecules between cells
  • Methods for

quantification are not
sensitive enough

  • High cost

Summary:

Research on vaccines has advanced significantly in recent years examining their enormous potential to treat a wide range of illnesses. In particular, nanovaccines have potential of developing less harmful and more effective vaccines due to the strong interactions of nanoparticles with the immune system. Strong antigenicity combined with extended retention and release makes nanovaccines producing memory effector response and both cell-mediated and antibody-mediated antibody responses, and thus reducing the need for repeated booster doses.

Challenges and future prospects:

The production of nanovaccines is limited by the toxicity of nanomedicines, scaling-up procedures, and the absence of regulatory requirements. Because pre-clinical and clinical investigations have demonstrated that nanovaccines causes dose-dependent acute and chronic toxicities with preferential bioaccumulation based on the route of administration, so the use of nanovaccines is controversial. Inorganic nanoparticles, such as metallic nanoparticles, are one of the types of nanoparticles that are inherently hazardous after extended exposure. Another key issue that has been somewhat mitigated by technical advancements is scaling up; nonetheless, scaling up in a sterile environment remains a considerable obstacle. In this era of modern medicine, where aim is to save countless lives. The persistent increase in antibiotic resistance and the lack of effective cancer treatment because of ongoing causative strain changes and disease heterogeneity has led to personalized medicine. Similar to this, vaccines against cancers and bacterial infections could be very beneficial; nevertheless, the existing vaccine's lack of potency and antigenic breadth are its main drawbacks. In order to create biomimetic nanovaccines as personalized medications, researchers have recently turned to biomimetic nanotechnology. These nanovaccines may be naturally multi-antigenic and immunostimulatory. When bacterial outer membrane vesicles (OMVs) are coated onto nanoparticles for anti-virulence immunization, the pathogen is surpassed by employing its own survival strategy and bacterial toxins are delivered and neutralized. This can lessen development of antibiotic resistance and successfully stop bacterial colonization. By altering these biomimetic nanovaccines' outer membrane coating, several nano-toxoid formulations can be created. Although cancer nano-based immunotherapy has advanced, the main obstacle remains the toxicity evaluations of these nanovaccines. It is necessary to evaluate autoimmune adverse effects that result from immune activation triggered by nanovaccines beforehand. Nanovaccines that over-activate antigen-presenting cells may be harmful because they can kill dendritic cells. Finally, the successful implementation of nano-based immunotherapy necessitates strategies to reduce the increasing complexity, manufacturing expense, and commercialization challenge associated with nanovaccine therapy.

Abbreviations

HIV: Human Immunodeficiency Virus

APCs: Antigen Presenting Cells

TLR: Toll-like receptor  

MHC: Major Histocompatibility Complex

DCs: Dendritic Cells

mRNA: Messanger Ribonucleic Acid

DNA: Deoxyribonucleic Acid

CTLs: Cytotoxic T Lymphocytes

PAMPs: Pathogen-Associated Molecular Patterns

PRRs:Pattern Recognition Receptors

VLPs:Virus-Like Particles

PNPs:Polymeric NanoParticles

HPV: Human Papilloma Virus

PLGA:Poly lactic-co-glycolic acid

PEI:Polyethyleneimine

PPS: Poly Propylene Sulphide

AuNPs: Gold nanoparticles

CNTs: Carbon nano-particles

OMVs: Outer Membrane Vesicles

IONPs: Iron oxides Nano Particles

SANPs: Self-Assembling Nano Particles

REFERENCES

        1. Bhupinder Singh et al.-An Overview, IJPFR, April-June 2011; 1(1): 101-109
        2. Ahmad S, et al. A Comprehensive Review on Types of Vaccines: From Classic to Cutting-Edge. Vaccines Vacccin 2023, 8(2): 000164
        3. Daniel Lozano et al.- An Overview of the Use of Nanoparticles in Vaccine Development. Nanomaterials 2023, 13, 1828.
        4. Cheol-Gyun et al- Enhancement of Immune Responses Elicited by Nanovaccines through a Cross-Presentation Pathway, Tissue Eng Regen Med, (2023) 20(3):355–370
        5. Manju et al Nanovaccines to combat drug resistance: the next-generation immunization, Future Journal of Pharmaceutical Sciences (2023) 9:64
        6. Priyanka et al Nanovaccines: A game changing approach in the fight against infectious diseases, Biomedicine & Pharmacotherapy 167 (2023) 115597
        7. Fredmoore L. Orosco-Review article : Recent advances in peptide-based nanovaccines for reemerging and emerging infectious diseases, J Adv Biotechnol Exp Ther. 2024 Jan; 7(1):106-117
        8. Prateek Bhardwaj et al-Review article:Advancements in prophylactic and therapeutic nanovaccines, Acta Biomaterialia 108 (2020) 1–21
        9. Chittaranjan Baruah et al- The emergence of nanovaccines as a new paradigm in virological vaccinology: A review, Explor Immunol. 2023;3:361–83
        10. Rajashri Bezbaruah et al- Review Nanoparticle-Based Delivery Systems for Vaccines, Vaccines 2022, 10, 1946.
        11. Sangiliyandi Gurunathan et al Review Nanovaccines: An effective therapeutic approach for cancer therapy, Biomedicine & Pharmacotherapy 170 (2024) 115992
        12. Min Yao et al-Research progress of nanovaccine in antitumor immunotherapy, Front. Oncol. 13:1211262
        13. Deka B, Babu A, Baruah C, Barthakur M. Nanopesticides: a systematic review of their prospects with special reference to tea pest management. Front Nutr. 2021;8:686131.
        14. Deka B, Baruah C, Babu A, Kalita P. Biological and non-conventional synthesis of Zinc oxide nanoparticles (ZnO-NPs): their potential applications. J Nanotechnol Nanomaterials. 2022;3:79–89.
        15. Deka B, Nisha SN, Baruah C, Babu A, Sarkar S, Phukan H, et al. Agricultural pest management with plant-derived nanopesticides: prospects and challenges. J App Nanotech. 2022;1:1–9.
        16. Cordeiro AS, Patil-Sen Y, Shivkumar M, Patel R, Khedr A, Elsawy MA. Nanovaccine delivery approaches and advanced delivery systems for the prevention of viral infections: from development to clinical application. Pharmaceutics. 2021;13:2091.
        17. Doll TA, Raman S, Dey R, Burkhard P. Nanoscale assemblies and their biomedical applications. J Soc Interface. 2013;10:20120740.
        18. Kelly HG, Kent SJ, Wheatley AK. Immunological basis for enhanced immunity of nanoparticle vaccines. Expert Rev Vaccines. 2019;18:269–80.
        19. Zaheer T, Pal K, Zaheer I. Topical review on nano-vaccinology: biochemical promises and key challenges. Process Biochem. 2021;100:237–44.
        20. Facciolà A, Visalli G, Laganà P, La Fauci V, Squeri R, Pellicanò GF, et al. The new era of vaccines: the “nanovaccinology”. Eur Rev Med Pharmacol Sci. 2019;23:7163–82.
        21. Irvine DJ, Read BJ. Shaping humoral immunity to vaccines through antigen-displaying nanoparticles. Curr Opin Immunol. 2020;65:1–6.
        22. Zhao L, Seth A, Wibowo N, Zhao CX, Mitter N, Yu C, et al. Nanoparticle vaccines. Vaccine. 2014;32:327–37.
        23. Aikins ME, Bazzill J, Moon JJ. Vaccine nanoparticles for protection against HIV infection. Nanomedicine (Lond). 2017;12:673–82.
        24. Gregoriadis G. Engineering liposomes for drug delivery: progress and problems. Trends Biotechnol.1995;13:527–37.
        25. Poon C, Gallo J, Joo J, Chang T, Bañobre-López M, Chung EJ. Hybrid, metal oxide-peptide amphiphile micelles for molecular magnetic resonance imaging of atherosclerosis. J Nanobiotechnology.2018;16:92.
        26. Singha S, Shao K, Ellestad KK, Yang Y, Santamaria P. Nanoparticles for immune stimulation against infection, cancer, and autoimmunity. ACS Nano. 2018;12:10621–35.
        27. Gaucher G, Dufresne MH, Sant VP, Kang N, Maysinger D, Leroux JC. Block copolymer micelles: preparation, characterization and application in drug delivery. J Control Release. 2005;109:169–88.
        28. Al-Halifa S, Gauthier L, Arpin D, Bourgault S, Archambault D. Nanoparticle-based vaccines against respiratory viruses. Front Immunol. 2019;10:22.
        29. Huang J, Ding Y, Yao J, Peng K, Deng K, Zhang M, et al. The SARS-CoV-2 rS1-E-PLGA nanovaccine and evaluation of its immune effect in BALB/c mice. Eur Rev Med Pharmacol Sci. 2022;26:5255–63.
        30. Huang X, Zhu Q, Huang X, Yang L, Song Y, Zhu P, et al. In vivo electroporation in DNA-VLP primeboost preferentially enhances HIV-1 envelope-specific IgG2a, neutralizing antibody and CD8 T cell responses. Vaccine. 2017;35:2042–51.
        31. Moon JJ, Suh H, Polhemus ME, Ockenhouse CF, Yadava A, Irvine DJ. Antigen-displaying lipidenveloped PLGA nanoparticles as delivery agents for a Plasmodium vivax malaria vaccine. PLoS One.2012;7:e31472.
        32. Fan Y, Stronsky SM, Xu Y, Steffens JT, van Tongeren SA, Erwin A, et al. Multilamellar vaccine particle elicits potent immune activation with protein antigens and protects mice against Ebola virus infection. ACS Nano. 2019;13:11087–96.
        33. Bano I, Arshad M, Yasin T, Ghauri MA, Younus M. Chitosan: a potential biopolymer for wound management. Int J Biol Macromol. 2017;102:380–3.
        34. Mohammed MA, Syeda JTM, Wasan KM, Wasan EK. An overview of chitosan nanoparticles and its application in non-parenteral drug delivery. Pharmaceutics. 2017;9:53.
        35. M Ways TM, Lau WM, Khutoryanskiy VV. Chitosan and its derivatives for application in mucoadhesive drug delivery systems. Polymers (Basel). 2018;10:267.
        36. Feng G, Jiang Q, Xia M, Lu Y, Qiu W, Zhao D, et al. Enhanced immune response and protective effects of nano-chitosan-based DNA vaccine encoding T cell epitopes of Esat-6 and FL against Mycobacterium tuberculosis infection. PLoS One. 2013;8:e61135.
        37. Karayianni M, Sentoukas T, Skandalis A, Pippa N, Pispas S. Chitosan-based nanoparticles for nucleic acid delivery: technological aspects, applications, and future perspectives. Pharmaceutics.2023;15:1849.
        38. Thomas C, Rawat A, Hope-Weeks L, Ahsan F. Aerosolized PLA and PLGA nanoparticles enhance humoral, mucosal and cytokine responses to hepatitis B vaccine. Mol Pharm. 2011;8:405–15.
        39. Prego C, Paolicelli P, Díaz B, Vicente S, Sánchez A, González-Fernández A, et al. Chitosan-based nanoparticles for improving immunization against hepatitis B infection. Vaccine. 2010;28:2607–14.
        40. Han J, Zhao D, Li D, Wang X, Jin Z, Zhao K. Polymer-based nanomaterials and applications for vaccines and drugs. Polymers (Basel). 2018;10:31.
        41. Clarke PD, Adams P, Ibáñez R, Herzog C. Rate, intensity, and duration of local reactions to a virosome-adjuvanted vs. an aluminium-adsorbed hepatitis A vaccine in UK travellers. Travel Med Infect Dis. 2006;4:313–8.
        42. Glück R, Mischler R, Finkel B, Que JU, Scarpa B, Cryz SJ Jr. Immunogenicity of new virosome influenza vaccine in elderly people. Lancet. 1994;344:160–3.
        43. Das I, Padhi A, Mukherjee S, Dash DP, Kar S, Sonawane A. Biocompatible chitosan nanoparticles as an efficient delivery vehicle for Mycobacterium tuberculosis lipids to induce potent cytokines and antibody response through activation of γδ T cells in mice. Nanotechnology. 2017;28:165101.
        44. Wang N, Chen M, Wang T. Liposomes used as a vaccine adjuvant-delivery system: From basics to clinical immunization. J Control Release. 201;303:130–50.
        45. Heegaard PM, Boas U, Sorensen NS. Dendrimers for vaccine and immunostimulatory uses. A review.Bioconjug Chem. 2010;21:405–18.
        46. Chahal JS, Khan OF, Cooper CL, McPartlan JS, Tsosie JK, Tilley LD, et al. Dendrimer-RNA nanoparticles generate protective immunity against lethal Ebola, H1N1 influenza, and Toxoplasma gondii challenges with a single dose. Proc Natl Acad Sci U S A. 2016;113:E4133–42.
        47. Chowdhury S, Toth I, Stephenson RJ. Dendrimers in vaccine delivery: recent progress and advances. Biomaterials. 2022;280:121303.
        48. Hassett KJ, Meinerz NM, Semmelmann F, Cousins MC, Garcea RL, Randolph TW. Development of a highly thermostable, adjuvanted human papillomavirus vaccine. Eur J Pharm Biopharm. 2015;94:220–8.
        49. Chattopadhyay S, Chen JY, Chen HW, Hu CJ. Nanoparticle vaccines adopting virus-like features for enhanced immune potentiation. Nanotheranostics. 2017;1:244–60.
        50. Niikura K, Matsunaga T, Suzuki T, Kobayashi S, Yamaguchi H, Orba Y, et al. Gold nanoparticles as a vaccine platform: influence of size and shape on immunological responses in vitro and in vivo. ACS Nano. 2013;7:3926–38.
        51. Chen YS, Hung YC, Lin WH, Huang GS. Assessment of gold nanoparticles as a size-dependent vaccine carrier for enhancing the antibody response against synthetic foot-and-mouth disease virus peptide. Nanotechnology. 2010;21:195101.
        52. Tao W, Gill HS. M2e-immobilized gold nanoparticles as influenza A vaccine: role of soluble M2e and longevity of protection. Vaccine. 2015;33:2307–15.
        53. Safari D, Marradi M, Chiodo F, Th Dekker HA, Shan Y, Adamo R, et al. Gold nanoparticles as carriers for a synthetic Streptococcus pneumoniae type 14 conjugate vaccine. Nanomedicine (Lond).2012;7:651–62.
        54. Carabineiro SAC. Applications of gold nanoparticles in nanomedicine: recent advances in vaccines. Molecules. 2017;22:857.
        55. Zhao Y, Zhao X, Cheng Y, Guo X, Yuan W. Iron oxide nanoparticles-based vaccine delivery for cancer treatment. Mol Pharm. 2018;15:1791–9.
        56. Pusic K, Aguilar Z, McLoughlin J, Kobuch S, Xu H, Tsang M, et al. Iron oxide nanoparticles as a clinically acceptable delivery platform for a recombinant blood-stage human malaria vaccine. FASEB J. 2013;27:1153–66.
        57. Nie J, Wang Q, Jin S, Yao X, Xu L, Chang Y, et al. Self-assembled multiepitope nanovaccine based on NoV P particles induces effective and lasting protection against H3N2 influenza virus. Nano Res.2023;16:7337–46.
        58. Malhi H, Homad LJ, Wan YH, Poudel B, Fiala B, Borst AJ, et al. Immunization with a self-assembling nanoparticle vaccine displaying EBV gH/gL protects humanized mice against lethal viral challenge.Cell Rep Med. 2022;3:100658.
        59. Burkhard P, Lanar DE. Malaria vaccine based on self-assembling protein nanoparticles. Expert Rev Vaccines. 2015;14:1525–7.
        60. Sung HD, Kim N, Lee Y, Lee EJ. Protein-based nanoparticle vaccines for SARS-CoV-2. Int J Mol Sci.2021;22:13445.

Reference

  1. Bhupinder Singh et al.-An Overview, IJPFR, April-June 2011; 1(1): 101-109
  2. Ahmad S, et al. A Comprehensive Review on Types of Vaccines: From Classic to Cutting-Edge. Vaccines Vacccin 2023, 8(2): 000164
  3. Daniel Lozano et al.- An Overview of the Use of Nanoparticles in Vaccine Development. Nanomaterials 2023, 13, 1828.
  4. Cheol-Gyun et al- Enhancement of Immune Responses Elicited by Nanovaccines through a Cross-Presentation Pathway, Tissue Eng Regen Med, (2023) 20(3):355–370
  5. Manju et al Nanovaccines to combat drug resistance: the next-generation immunization, Future Journal of Pharmaceutical Sciences (2023) 9:64
  6. Priyanka et al Nanovaccines: A game changing approach in the fight against infectious diseases, Biomedicine & Pharmacotherapy 167 (2023) 115597
  7. Fredmoore L. Orosco-Review article : Recent advances in peptide-based nanovaccines for reemerging and emerging infectious diseases, J Adv Biotechnol Exp Ther. 2024 Jan; 7(1):106-117
  8. Prateek Bhardwaj et al-Review article:Advancements in prophylactic and therapeutic nanovaccines, Acta Biomaterialia 108 (2020) 1–21
  9. Chittaranjan Baruah et al- The emergence of nanovaccines as a new paradigm in virological vaccinology: A review, Explor Immunol. 2023;3:361–83
  10. Rajashri Bezbaruah et al- Review Nanoparticle-Based Delivery Systems for Vaccines, Vaccines 2022, 10, 1946.
  11. Sangiliyandi Gurunathan et al Review Nanovaccines: An effective therapeutic approach for cancer therapy, Biomedicine & Pharmacotherapy 170 (2024) 115992
  12. Min Yao et al-Research progress of nanovaccine in antitumor immunotherapy, Front. Oncol. 13:1211262
  13. Deka B, Babu A, Baruah C, Barthakur M. Nanopesticides: a systematic review of their prospects with special reference to tea pest management. Front Nutr. 2021;8:686131.
  14. Deka B, Baruah C, Babu A, Kalita P. Biological and non-conventional synthesis of Zinc oxide nanoparticles (ZnO-NPs): their potential applications. J Nanotechnol Nanomaterials. 2022;3:79–89.
  15. Deka B, Nisha SN, Baruah C, Babu A, Sarkar S, Phukan H, et al. Agricultural pest management with plant-derived nanopesticides: prospects and challenges. J App Nanotech. 2022;1:1–9.
  16. Cordeiro AS, Patil-Sen Y, Shivkumar M, Patel R, Khedr A, Elsawy MA. Nanovaccine delivery approaches and advanced delivery systems for the prevention of viral infections: from development to clinical application. Pharmaceutics. 2021;13:2091.
  17. Doll TA, Raman S, Dey R, Burkhard P. Nanoscale assemblies and their biomedical applications. J Soc Interface. 2013;10:20120740.
  18. Kelly HG, Kent SJ, Wheatley AK. Immunological basis for enhanced immunity of nanoparticle vaccines. Expert Rev Vaccines. 2019;18:269–80.
  19. Zaheer T, Pal K, Zaheer I. Topical review on nano-vaccinology: biochemical promises and key challenges. Process Biochem. 2021;100:237–44.
  20. Facciolà A, Visalli G, Laganà P, La Fauci V, Squeri R, Pellicanò GF, et al. The new era of vaccines: the “nanovaccinology”. Eur Rev Med Pharmacol Sci. 2019;23:7163–82.
  21. Irvine DJ, Read BJ. Shaping humoral immunity to vaccines through antigen-displaying nanoparticles. Curr Opin Immunol. 2020;65:1–6.
  22. Zhao L, Seth A, Wibowo N, Zhao CX, Mitter N, Yu C, et al. Nanoparticle vaccines. Vaccine. 2014;32:327–37.
  23. Aikins ME, Bazzill J, Moon JJ. Vaccine nanoparticles for protection against HIV infection. Nanomedicine (Lond). 2017;12:673–82.
  24. Gregoriadis G. Engineering liposomes for drug delivery: progress and problems. Trends Biotechnol.1995;13:527–37.
  25. Poon C, Gallo J, Joo J, Chang T, Bañobre-López M, Chung EJ. Hybrid, metal oxide-peptide amphiphile micelles for molecular magnetic resonance imaging of atherosclerosis. J Nanobiotechnology.2018;16:92.
  26. Singha S, Shao K, Ellestad KK, Yang Y, Santamaria P. Nanoparticles for immune stimulation against infection, cancer, and autoimmunity. ACS Nano. 2018;12:10621–35.
  27. Gaucher G, Dufresne MH, Sant VP, Kang N, Maysinger D, Leroux JC. Block copolymer micelles: preparation, characterization and application in drug delivery. J Control Release. 2005;109:169–88.
  28. Al-Halifa S, Gauthier L, Arpin D, Bourgault S, Archambault D. Nanoparticle-based vaccines against respiratory viruses. Front Immunol. 2019;10:22.
  29. Huang J, Ding Y, Yao J, Peng K, Deng K, Zhang M, et al. The SARS-CoV-2 rS1-E-PLGA nanovaccine and evaluation of its immune effect in BALB/c mice. Eur Rev Med Pharmacol Sci. 2022;26:5255–63.
  30. Huang X, Zhu Q, Huang X, Yang L, Song Y, Zhu P, et al. In vivo electroporation in DNA-VLP primeboost preferentially enhances HIV-1 envelope-specific IgG2a, neutralizing antibody and CD8 T cell responses. Vaccine. 2017;35:2042–51.
  31. Moon JJ, Suh H, Polhemus ME, Ockenhouse CF, Yadava A, Irvine DJ. Antigen-displaying lipidenveloped PLGA nanoparticles as delivery agents for a Plasmodium vivax malaria vaccine. PLoS One.2012;7:e31472.
  32. Fan Y, Stronsky SM, Xu Y, Steffens JT, van Tongeren SA, Erwin A, et al. Multilamellar vaccine particle elicits potent immune activation with protein antigens and protects mice against Ebola virus infection. ACS Nano. 2019;13:11087–96.
  33. Bano I, Arshad M, Yasin T, Ghauri MA, Younus M. Chitosan: a potential biopolymer for wound management. Int J Biol Macromol. 2017;102:380–3.
  34. Mohammed MA, Syeda JTM, Wasan KM, Wasan EK. An overview of chitosan nanoparticles and its application in non-parenteral drug delivery. Pharmaceutics. 2017;9:53.
  35. M Ways TM, Lau WM, Khutoryanskiy VV. Chitosan and its derivatives for application in mucoadhesive drug delivery systems. Polymers (Basel). 2018;10:267.
  36. Feng G, Jiang Q, Xia M, Lu Y, Qiu W, Zhao D, et al. Enhanced immune response and protective effects of nano-chitosan-based DNA vaccine encoding T cell epitopes of Esat-6 and FL against Mycobacterium tuberculosis infection. PLoS One. 2013;8:e61135.
  37. Karayianni M, Sentoukas T, Skandalis A, Pippa N, Pispas S. Chitosan-based nanoparticles for nucleic acid delivery: technological aspects, applications, and future perspectives. Pharmaceutics.2023;15:1849.
  38. Thomas C, Rawat A, Hope-Weeks L, Ahsan F. Aerosolized PLA and PLGA nanoparticles enhance humoral, mucosal and cytokine responses to hepatitis B vaccine. Mol Pharm. 2011;8:405–15.
  39. Prego C, Paolicelli P, Díaz B, Vicente S, Sánchez A, González-Fernández A, et al. Chitosan-based nanoparticles for improving immunization against hepatitis B infection. Vaccine. 2010;28:2607–14.
  40. Han J, Zhao D, Li D, Wang X, Jin Z, Zhao K. Polymer-based nanomaterials and applications for vaccines and drugs. Polymers (Basel). 2018;10:31.
  41. Clarke PD, Adams P, Ibáñez R, Herzog C. Rate, intensity, and duration of local reactions to a virosome-adjuvanted vs. an aluminium-adsorbed hepatitis A vaccine in UK travellers. Travel Med Infect Dis. 2006;4:313–8.
  42. Glück R, Mischler R, Finkel B, Que JU, Scarpa B, Cryz SJ Jr. Immunogenicity of new virosome influenza vaccine in elderly people. Lancet. 1994;344:160–3.
  43. Das I, Padhi A, Mukherjee S, Dash DP, Kar S, Sonawane A. Biocompatible chitosan nanoparticles as an efficient delivery vehicle for Mycobacterium tuberculosis lipids to induce potent cytokines and antibody response through activation of γδ T cells in mice. Nanotechnology. 2017;28:165101.
  44. Wang N, Chen M, Wang T. Liposomes used as a vaccine adjuvant-delivery system: From basics to clinical immunization. J Control Release. 201;303:130–50.
  45. Heegaard PM, Boas U, Sorensen NS. Dendrimers for vaccine and immunostimulatory uses. A review.Bioconjug Chem. 2010;21:405–18.
  46. Chahal JS, Khan OF, Cooper CL, McPartlan JS, Tsosie JK, Tilley LD, et al. Dendrimer-RNA nanoparticles generate protective immunity against lethal Ebola, H1N1 influenza, and Toxoplasma gondii challenges with a single dose. Proc Natl Acad Sci U S A. 2016;113:E4133–42.
  47. Chowdhury S, Toth I, Stephenson RJ. Dendrimers in vaccine delivery: recent progress and advances. Biomaterials. 2022;280:121303.
  48. Hassett KJ, Meinerz NM, Semmelmann F, Cousins MC, Garcea RL, Randolph TW. Development of a highly thermostable, adjuvanted human papillomavirus vaccine. Eur J Pharm Biopharm. 2015;94:220–8.
  49. Chattopadhyay S, Chen JY, Chen HW, Hu CJ. Nanoparticle vaccines adopting virus-like features for enhanced immune potentiation. Nanotheranostics. 2017;1:244–60.
  50. Niikura K, Matsunaga T, Suzuki T, Kobayashi S, Yamaguchi H, Orba Y, et al. Gold nanoparticles as a vaccine platform: influence of size and shape on immunological responses in vitro and in vivo. ACS Nano. 2013;7:3926–38.
  51. Chen YS, Hung YC, Lin WH, Huang GS. Assessment of gold nanoparticles as a size-dependent vaccine carrier for enhancing the antibody response against synthetic foot-and-mouth disease virus peptide. Nanotechnology. 2010;21:195101.
  52. Tao W, Gill HS. M2e-immobilized gold nanoparticles as influenza A vaccine: role of soluble M2e and longevity of protection. Vaccine. 2015;33:2307–15.
  53. Safari D, Marradi M, Chiodo F, Th Dekker HA, Shan Y, Adamo R, et al. Gold nanoparticles as carriers for a synthetic Streptococcus pneumoniae type 14 conjugate vaccine. Nanomedicine (Lond).2012;7:651–62.
  54. Carabineiro SAC. Applications of gold nanoparticles in nanomedicine: recent advances in vaccines. Molecules. 2017;22:857.
  55. Zhao Y, Zhao X, Cheng Y, Guo X, Yuan W. Iron oxide nanoparticles-based vaccine delivery for cancer treatment. Mol Pharm. 2018;15:1791–9.
  56. Pusic K, Aguilar Z, McLoughlin J, Kobuch S, Xu H, Tsang M, et al. Iron oxide nanoparticles as a clinically acceptable delivery platform for a recombinant blood-stage human malaria vaccine. FASEB J. 2013;27:1153–66.
  57. Nie J, Wang Q, Jin S, Yao X, Xu L, Chang Y, et al. Self-assembled multiepitope nanovaccine based on NoV P particles induces effective and lasting protection against H3N2 influenza virus. Nano Res.2023;16:7337–46.
  58. Malhi H, Homad LJ, Wan YH, Poudel B, Fiala B, Borst AJ, et al. Immunization with a self-assembling nanoparticle vaccine displaying EBV gH/gL protects humanized mice against lethal viral challenge.Cell Rep Med. 2022;3:100658.
  59. Burkhard P, Lanar DE. Malaria vaccine based on self-assembling protein nanoparticles. Expert Rev Vaccines. 2015;14:1525–7.
  60. Sung HD, Kim N, Lee Y, Lee EJ. Protein-based nanoparticle vaccines for SARS-CoV-2. Int J Mol Sci.2021;22:13445.

Photo
Gurpreet Kaur
Corresponding author

Rayat Bahra Institute of Pharmacy, Hoshiarpur.

Photo
Ashita Pawaiya
Co-author

Rayat Bahra Institute of Pharmacy, Hoshiarpur.

Photo
Damandeep Kaur
Co-author

Rayat Bahra Institute of Pharmacy, Hoshiarpur.

Photo
Sumit Pasricha
Co-author

Rayat Bahra Institute of Pharmacy, Hoshiarpur.

Photo
Priyanka Rani
Co-author

Rayat Bahra Institute of Pharmacy, Hoshiarpur.

Gurpreet Kaur*, Ashita Pawaiya, Damandeep Kaur, Sumit Pasricha, Priyanka Rani , Nanovaccines: A Revolutionary Paradigm in Immunization, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 4, 1271-1289 https://doi.org/10.5281/zenodo.15189996

More related articles
Review on lipid based carrier as a drug delivery s...
Pranali Subhash jadhav, Pranali Hatwar, Gajanan Sanap, ...
Melatonin Loaded Nanocarriers...
Raslamol K., Saranya K. G., Amayasree Suresh, Archa Ani, A. S. At...
Chikungunya: A Comprehensive Narrative Review on E...
Komal Pimpalshende, Kunal Darote, Sejal Sontakke, Dr. Swati Gaikw...
The Impact Of Nanotechnology On Drug Delivery System...
Pranjal tukaram lahare, Shiddhesh sunil chavan, ...
Chikungunya: A Comprehensive Narrative Review on Epidemiology, Pathogenesis and ...
Komal Pimpalshende, Kunal Darote, Sejal Sontakke, Dr. Swati Gaikwad, ...
Advancements of Cancer Treatments and Cure...
Priyanka Tadkase, Shital Bhimewar, Pallavi Bawanthade, Fiza Firoz Karvinkar, Dhanashri Chelmelwar, ...
Related Articles
Cancer Vaccines Delivery Systems: Strategies For Efficient Targeting And Immuniz...
Parikshit Nagda, Krishna Hingad, Ghisulal Dewasi, Nirmal Sodha , Nihal Singh Rao, Bhaktraj Singh Cha...
A Comprehensive Review On Cubosomes...
Sahil Nandkumar kamble , Radhika Subhedar, Nilesh B. Chougule, ...
Analysis Of Common Vaccines and Associated Adverse Events in Immunized Pediatric...
Hari Singh Rathore, Deepanmita Wakalwar, Y. N. Verma, Piyush Agrawal, Mahendra Singh Rathore, ...
The Inhalable Dry Powder Nano- Formulation Advancing Lung Disease Therapy ...
Muskare Mayuri , Pandhare Jayashri, Majge Shashank , Pandhare Priya, ...
Review on lipid based carrier as a drug delivery system...
Pranali Subhash jadhav, Pranali Hatwar, Gajanan Sanap, ...
More related articles
Review on lipid based carrier as a drug delivery system...
Pranali Subhash jadhav, Pranali Hatwar, Gajanan Sanap, ...
Melatonin Loaded Nanocarriers...
Raslamol K., Saranya K. G., Amayasree Suresh, Archa Ani, A. S. Athira, Febiya Joy, Gayathri Prasad, ...
Chikungunya: A Comprehensive Narrative Review on Epidemiology, Pathogenesis and ...
Komal Pimpalshende, Kunal Darote, Sejal Sontakke, Dr. Swati Gaikwad, ...
Review on lipid based carrier as a drug delivery system...
Pranali Subhash jadhav, Pranali Hatwar, Gajanan Sanap, ...
Melatonin Loaded Nanocarriers...
Raslamol K., Saranya K. G., Amayasree Suresh, Archa Ani, A. S. Athira, Febiya Joy, Gayathri Prasad, ...
Chikungunya: A Comprehensive Narrative Review on Epidemiology, Pathogenesis and ...
Komal Pimpalshende, Kunal Darote, Sejal Sontakke, Dr. Swati Gaikwad, ...