Abstract
Vaccination programmes provide a safe, effective and cost‐efficient strategy for maintaining population health. In veterinary medicine, vaccination not only reduces disease within animal populations but also serves to enhance public health by targeting zoonoses. Nevertheless, for many pathogens, an effective vaccine remains elusive. Recently, nanovaccines have proved to be successful for various infectious and non‐infectious diseases of animals. These novel technologies, such as virus‐like particles, self‐assembling proteins, polymeric nanoparticles, liposomes and virosomes, offer great potential for solving many of the vaccine production challenges. Their benefits include low immunotoxicity, antigen stability, enhanced immunogenicity, flexibility sustained release and the ability to evoke both humoral and cellular immune responses. Nanovaccines are more efficient than traditional vaccines due to ease of control and plasticity in their physio‐chemical properties. They use a highly targeted immunological approach which can provide strong and long‐lasting immunity. This article reviews the currently available nanovaccine technology and considers its utility for both infectious diseases and non‐infectious diseases such as auto‐immunity and cancer. Future research opportunities and application challenges from bench to clinical usage are also discussed.
Keywords: nanotechnology, nanovaccine, veterinary, zoonosis control
Nanovaccines are successful because of the enhanced lowest immunotoxicity, antigen stability, better immunogenicity, the flexibility of the physical characteristics of nanomaterials and sustained release.

1. INTRODUCTION
Nanotechnology is a new field of science with various applications, including synthesizing different nanoparticle sizes and structures. In veterinary medicine, nanotechnology has enabled us to find treatments and develop powerful immune responses against infectious and non‐infectious diseases (Lei & Karim, 2021; Sahoo et al., 2021; Woldeamanuel et al., 2021). Nanotechnology finds its most notable application in cancer research, where it is primarily employed to enhance the detection and treatment of cancerous cells (Yang et al., 2021). According to recent studies (Alghuthaymi et al., 2021; Hu et al., 2020; Ross et al., 2019; Soliman et al., 2019), nanoparticles have emerged as a promising area of research for combating various types of diseases caused by microorganisms, such as fungi, bacteria and viruses. Nanoparticles possess unique properties that make them a potential tool for targeted drug delivery, diagnostic imaging and other therapeutic approaches. Nanoscience improves the imaging and diagnosis of diseases by removing biological restrictions (Tatli Seven et al., 2018). Various types of nanoparticles in veterinary medicine have been developed and characterized. Nanoparticles, such as liposomes, emulsions, proteasomes, nano‐beads, ISCOMs and polymeric biological nanoparticles like exosomes and bacteriophages, are being used to treat both non‐infectious and infectious diseases (Bai et al., 2018; Chariou et al., 2020; da Silva et al., 2021; Youssef et al., 2019). Nanoparticles are a powerful option for vaccine commerce due to the inactivity of surface change and the capacity to efficiently co‐transfer the adjuvants (Zaheer et al., 2021). Furthermore, the nano‐adjuvants in biological systems save the goal antigen from decay and improve absorption by immune intermediates (Sahu et al., 2018). This system leads to steady immunogenic effects and potentially delivers the antigen repetitively (Celis‐Giraldo et al., 2021). The size of nanoparticles significantly impacts their interaction with the immune system and how they are dispersed throughout the body. Nanoparticles that are smaller than 100 nanometres are readily taken up by dendritic cells (Shi et al., 2017). The nanoparticles can function as beneficial adjuvants, stimulating both cellular and humoral immune responses. As a result, the biomaterial can circulate more quickly, be more bioavailable, and have their biological components protected from degradation. In nanovaccines, regulating adjuvant and antigen size, surface charge, shape, hydrophobicity, flexibility and charge density is the key to improving a strong immune reaction by transferring nanovaccines to the lymphatic vessel. Polyethylene glycol (PEG) can slow down drug release from cells, which can increase the circulation time of the drug and help accumulate medication at the site of injury or damage (Prasad, Charmode, et al., 2021). Biodegradable nanovaccines can enhance slow release, antigen shelf life, immunogenicity and regular delivery to target cells (Cerbu et al., 2021). Nano‐medication is helpful in the prevention and treatment of diseases (Suzana Gonçalves Carvalho et al., 2020). Nanovaccines are far more efficient than conventional vaccines, and they can destroy infectious agents by activating the immune system in the blood and cells (Facciolà et al., 2019). Indeed, the stimulation of the immune responses can happen by transferring antigens to the immune system and providing the body with more powerful responses to the primary pathogen (Bhardwaj et al., 2020; Yadav et al., 2018). Nanoscience application in vaccine development has provided a novel method for solving the past questions. Nanovaccines also can deal with cancer treatment (Bragazzi, 2019; Ni et al., 2020). Tumour immunotherapy is a new medicinal way to inhibit tumour metastasis and recognize and kill cancer cells, by stimulating the patient's immune responses ( Asouli et al., 2023; Kheirollahpour et al., 2020; Prajapati et al., 2022). Within the field of immuno‐oncology, various modalities have emerged as promising approaches to enhance anti‐tumour immune responses. These include tumour vaccines, chimeric antigen receptor T‐Cell adoptive immunotherapy, and immune checkpoint inhibitors. These modalities have shown considerable efficacy in preclinical and clinical studies, offering new hope in the fight against cancer (Billingsley et al., 2020; Mi et al., 2019). In the past few years, cancer vaccines have proven to have hopeful therapeutic effects in scientific trials due to their convenient preparation, high specificity and relatively low cost (Cuzzubbo et al., 2021; El‐Sayed & Kamel, 2020). The main procedure of cancer vaccines is triggering and rapid increase of tumour antigen‐specific (CTL), cytotoxic T lymphocytes to destroy tumour tissues (Chen et al., 2021).
Nanoscience has gained significant attention in the field of veterinary medicine, with particular emphasis on its potential applications in diagnosis, vaccination and treatment. This article will provide a review of several Nano‐Biotech programs that are focused on advancing vaccination science in veterinary medicine.
2. TYPE OF NANOPARTICLES AND THEIR APPLICATIONS
A nanoparticle is a material with unique properties and a size range of 1–1000 nanometres. Nanoparticles with distinct chemical structures and physical properties could deliver molecules for their anthelmintic effects. They could be labelled as follows.
2.1. Polymeric structures
Generally, polymers are chain‐like structures with diverse characteristics. They can be natural polymers like polysaccharides, such as inulin, chitosan or synthetic, like PEG, other polyanhydrides, and polyesters (Han et al., 2018). Some synthetic polymers, like aliphatic polyesters, are environmentally friendly because of their decay characteristics. Moreover, hydrophobic and hydrophilic macromolecules can be encapsulated in polyester‐based polymers. Furthermore, natural biopolymers demonstrate benefits, such as low toxicity, biodegradability, abundance and biocompatibility (Davoodi et al., 2022). The most common polysaccharides in nano‐pharmacology are chitosan, dextran, agarose, hyaluronic acid and protein polymers, such as albumin, gelatin or soy (Cordeiro et al., 2015). Until now, chitosan has contributed to most studies, thanks to its non‐immunogenic impacts, great biocompatibility and the capacity to combine with other polymers. Due to these effects, chitosan is one of the most common molecules for cancer treatment (Chua et al., 2012). Polymeric nanoparticles include natural, artificial or semi‐synthetic polymers, so some challenges for preparing those structures will be the drug balance or compatibility and inert substance with the polymers (Guo et al., 2015). Several pure polymers can improve drug delivery systems, including pectin, carboxymethyl cellulose, alginate, hyaluronic acid, hypromellose phthalate, dextran and chitosan (Luo et al., 2017). The production technique performs a vital role in attaining the ideal properties of the nanoparticles, far from the natural features of polymers (Su et al., 2022). Polymeric nanoparticles have gained attention recently due to their potential applications in drug delivery, diagnostics and imaging. Various techniques have been developed for producing these nanoparticles, including reversed‐phase microemulsion, ionotropic gelation, emulsification solvent evaporation, emulsion polymerization, nanoprecipitation and supercritical fluid technology (George et al., 2019).
2.2. Lipid‐based nanoparticles
These nanoparticles are primarily created for lipophilic medication delivery and consist of a solid lipid matrix, surfactant and stabilizer. The lipid matrix components include glyceride mixtures, triglycerides and waxes (Thi et al., 2021). A further benefit of lipid nanoparticles and liposomes is that most of them are already authorized by European Medicines Agency and the Food and Drug Administration (García‐Pinel et al., 2019).
2.2.1. Liposomes
Liposomes were presented in the 1970s as drug carriers, and today they are contained in some EU‐approved and USA‐formulated medications to decrease the harmful impacts of powerful medicines (Samimi et al., 2019). Liposomes are the most broadly defined nanoparticles in literature for veterinary applications. They are nanostructures shaped through double lipid layers composed of phospholipids (Ickenstein & Garidel, 2019). These nanoparticles offer numerous advantages, including their ability to interact with both water‐soluble and lipid‐soluble medications, their ability to be regulated by electric charge, their size range of 25–1000 nm and their capacity for functionalization (Kumar, 2019). Bangham & Horne (1964) defined these nanoparticles. Since then, numerous research with this nanodevice has been developed, displaying promise in multiple areas, like veterinary medicine. Nevertheless, several problems limit their use, like drug leakage, stability and upscaling (Ahmad et al., 2021).
2.2.2. Solid lipid nanoparticles (SLN)
As an alternative to liposomes, solid lipid nanoparticles (SLN) appeared after 1990 (size levels 50–1000 nm). SLN may be controlled through all application methods; their manufacturing usually distributes natural liquids and is readily useful (Wang & Luo, 2019). Furthermore, biodegradable solid lipids can be utilized to form SLN, which is supported by surfactants to maintain stability. Therefore, the SLN nanoparticle technology is an innovative approach for creating NE (oil‐in‐water) formulations by replacing the liquid lipid with a solid form, resulting in even greater stability (Wang et al., 2021). Hence, it is possible to regulate the release and balance of the system under environmentally harmful situations. SLNs demonstrate several benefits, such as the capacity for parenteral routes, excellent stability, release control, specific targeting, and the physical safety of easily destroyable drugs (Geszke‐Moritz & Moritz, 2016). On the other hand, some problems stay, such as drug expulsion and limited medicine loading.
2.2.3. Nanostructured lipid carriers (NLC)
In the mid‐2000s, nanostructured lipid carriers (NLCs) appeared as enhanced lipid‐based carriers to defeat the previous problems of SLN and liposomes. Nano lipid carriers account for the second‐generation SLN and the third‐generation NE (Tang et al., 2018). The biophysical stability of solid lipids prevents the deactivation of medication and overcomes issues related to its precursor, such as problems with drug transportation and storage. Similar to SLN, NLCs are particles with an aqueous external medium, which is stabilized with amphiphilic elements called surfactants in constructional solid lipids (Fernandes et al., 2021). However, a fragment of this solid lipid phase can be replaced by (oil) in the liquid phase, which prepares a formless structure, preventing its removal and promoting better drug encapsulation (Chuang et al., 2018). For nanotoxicology factors, the great biocompatibility of SLN and NLC was similar to that of Doktorovova et al. (2014).
2.3. Dendrimers
Dendrimers were broadly examined in drug delivery for anticancer and antimicrobials purposes, gene transfer, immunization and MRI imaging, but only a few methods were used for anthelmintic medicines (Tabatabaie et al., 2018). These molecules are hyper‐branched, containing an initiator centre, an internal layer of repetitive parts, and surrounding, multifunctional groups. The core both illustrates the molecules’ shape and encapsulates various types of particles (Hari et al., 2012). Each new layer or branch is named ‘generation’, and the second layer can transfer small particles. Furthermore, outer functional groups are connected to their surroundings. Their solubility, low cost, tuning ability and cell membrane make them appropriate for different use as drug carriers (Paleos et al., 2010). The first study for dendrimer was on polyamidoamine (PAMAM), adjusted by ethylenediamine or ammonia cores. The PAMAM functional characteristics and shape are highly similar to natural proteins, for example haemoglobin or insulin (Kheraldine et al., 2021). Recently, more dendrimer groups were introduced, for instance, tecto, polipropilenimine, multilingual, amphiphilic, chiral and micellar dendrimers (Sherje et al., 2018).
2.4. Metallic nanoparticles
Metallic nanoparticles’ size ranges from 1 to 100 nm, and they can couple proteins, medicines, antibodies and other biomedical particles (Li et al., 2022). Combining nano‐metal with drugs can reduce the negative effects of drugs on the immune system. Some nano‐metal particles have antimicrobial and anthelmintic properties, which can improve treatment outcomes alone or with other medicines (Reddy et al., 2020). The primary biomedicine use of these nanoparticles is bioimaging, biosensing, hyperthermy, gene transfer, drug delivery systems and cell labelling (Manjula et al., 2022). The most commonly used metallic nanoparticles for drug delivery are silver, gold, iron oxide, copper and zinc (Chouhan & Mandal, 2021). These nanoparticles in veterinary science have been chiefly used as antiviral and antimicrobial agents (Liew et al., 2022).
2.5. Micellar nanostructures
Micelles are drug carriers that consist of amphiphilic polymers and have two parts, the internal part (nucleus) constitutes a hydrophobic sector, and the external part (crown) consists of a hydrophilic sector that has the size of (10–100 nm) (Sanabria, 2021). The particle size can be controlled in different ways, achieving methodology, the aggregates number, the polymers molecular weight and the number of polymers (Li & Zhang, 2021). Other kinds can make polymeric micellar nanoparticles, which divide into four categories by the division of the hydrophilic part. There are four classes of micelles. The first class is phospholipidic micelles, which have phospholipids in their crown. The second class is pluronic micelles, the third class is poly‐l‐amino acid micelles and the fourth class is polyester micellar nanoparticles, which are biocompatible (Carvalho et al., 2020).
2.6. Nanoemulsion
Nanoemulsions (NEs) are formed from the colloidal dispersion of droplets, developed in at least three parts: water, oil and the emulsifier, and the size might differ (50–1000 nm). It is a kinetically steady and thermodynamically irregular method. Some research has shown the benefit of NE as a nano‐delivery approach for veterinary purposes (Manocha et al., 2022). NE is applicable to transferring medicines into the liquid portions of breeding living organisms or steadily releasing poorly injectable hydrophilic medications (Costa et al., 2021). Nanoemulsions can help improve the bioactivity and steadiness of antiparasitic drugs for treating animal parasitic diseases (Rehman et al., 2020) (Figure 1).
FIGURE 1.

Current applications used in the veterinary medicine field for nanovaccine development.
3. WHY NANO‐VACCINOLOGY?
In recent years, nano‐vaccinology has developed quickly, leading to the production of ‘Nano‐Vaccines’. Numerous peptides and proteins have recently been introduced as a new treatment method (Askarizadeh et al., 2020). Some of their positive features were: controlled drug release, increased drug stability and improved targeting capacity (Jazayeri et al., 2021). Antigen‐carrying nanoparticles can influence the immune system and improve the T‐cell cytotoxic response against antigens linked to nanoparticle (Huang et al., 2019). This occurs due to the biological potential of several antigen‐expressing cells, which can strongly attract foreign particles like bacteria and microparticles (Cordeiro & Alonso, 2016). Another advantage is regarding lymphoid tissues, the nanoscale size of the particle and proper antigen production and diagnosis (Prasanna et al., 2021). Regarding nanovaccines, the nanoparticles interact with the immunogen in one of three ways: absorption, conjugation or encapsulation (Shen et al., 2018). This cellular nutrition procedure detects antigens to express foreign antigens to other cells in the immune system. Nano‐metric particles can improve the efficacy of vaccines due to their biomedical uses (Du & Sun, 2020). Nanoparticles also have various characteristics as adjuvants for vaccines. However, these materials have some problems, like immunity toxicity and unspecific absorption by the intracranial system (Yun & Cho, 2020). Nanotechnology presents opportunities to use molecules for scientific purposes, where traditional strategies are limited.
4. NANOVACCINES INTERVENTION IN VETERINARY MEDICINE SCIENCE AND ANALYSING CURRENT STUDIES
Nanoparticles have received much attention recently because they use a release system or immune system enhancers (Rosales‐Mendoza & González‐Ortega, 2019). In addition, they have emerged as a novel method for vaccination. Moreover, nanovaccines are more efficient than conventional vaccines and trigger blood and cell immune reactions (Loera‐Muro & Angulo, 2018). Nanovaccines inhibit infection by killing infectious agents controlling the immune system. Furthermore, vaccines significantly benefit from nanoparticles formulations, enhancing antigen perception, targeted APCs administration, immunogenicity and slow release of antigens (Sun et al., 2018). Because of their biodegradability, bioavailability and minimal toxicity, most vaccine models containing NPs can be safe and effective alternatives to traditional vaccine formulas (Seyfoori et al., 2021). With the help of nanoparticles as adjuvants or vectors, a new generation of vaccines is produced. For instance, they can efficiently activate the immune response due to the similar size of pathogens and nanoparticles (Xiang et al., 2006). The stimulation of each humoral and cellular immune response followed the application of this kind of vaccine. The advantages of nanovaccines are their best stability in the bloodstream, improved immune system activation, cold chain and no boosters (Prasad, Ghosh, et al., 2021). Using nanoparticles in vaccinology presents two important benefits (Shah et al., 2014). The first benefit is that NPs act as adjuvants and antigens, which increase the antigenicity of conjugated and adsorbed antigens. In this case, they can imitate the characteristics of pathogens like viruses (Dmour et al., 2022). Second, the results of inflammatory cytokines mediate many immune responses activated by nanoparticles. Consequently, different nanoparticle frames are currently advanced for various applications (Chen et al., 2017). These structures can be artificially engineered or found in nature. The engineered particles can mainly be created to select the immune system or avoid interactions. This benefit is linked to the nano‐size (which stimulates absorption by phagocytic cells), mucosa‐associated lymphoid tissues and effective presentation and recognition of antigens (Vinay et al., 2018). The various types of NPs, including virus‐like particles, liposomes, inorganic particles and polymeric particles, have become widely recognized in medical science, particularly in the field of vaccinology (Lei et al., 2020). The emulsion ability of nanoparticles helps synthetic vaccines to enhance immunity and to perform as a vaccine delivery system for different materials. It can cause pliable natural and immune responses. They are used as antigen carriers to improve antigen processing because of their functionality and surface space. These qualities lead to controlled antigen release and effective cell targeting. Nanoparticles have the potential to both increase the half‐life of most vaccines and release antigens in a controlled way (Azadi et al., 2020). Additionally, they can act individually as immune enhancers. However, mixed with immune enhancers, classical adjuvants have more sophistication than improved immunogenicities (Table 1) (Figure 2).
TABLE 1.
Current status of nano‐vaccinology in veterinary medicine science.
| Target | Nanoparticle carrier | Model of study | Result | References |
|---|---|---|---|---|
| Anaplasma marginale | Silica vesicles: thin wall of 6 nm and pore size of 5.8 nm | Mouse | Activate both B‐ and T‐cell immunity | Zhao et al. (2016) |
| Leishmania major | TSA recombinant plasmid and poly(methyl methacrylate) nanoparticles | Mouse | Inducing specific antibody responses | Zarrati et al. (2014) |
| L. major | PLGA nanoparticle | Mouse | Significant induction of nitric oxide production by peritoneal macrophages and increase in splenocyte IFN‐γ production showed the protective effect of PLGA‐CPA/CPB vaccination. The conjugation of the antigen with the PLGA can activate immune responses against L. major | Noormehr et al. (2018) |
| Echinococcus granulosus | Polymeric nanoparticles | Dog | Higher cell internalization in murine macrophages and dendritic cells as well as a higher penetration into Caco‐2 cells in vitro | |
| Haemonchus contortus | Poly(lactic‐co‐glycolic acid) (PLGA) nanoparticle | Goat | The level of IL‐17 and TGF‐β increased and the faecal eggs and the abomasal worm burdens was reduced | Wang et al. (2021) |
| Theileria parva | Silica vesicle (SV)‐p67C | Cattle | Primed strong Ab and T‐cell responses, stimulation of both high Ab titres and CD4 T‐cell response to p67C | Lacasta et al. (2021) |
| Rhipicephalus microplus |
Silica vesicles (SVs) Rhodamine‐labelled SV‐140‐C18 (Rho‐SV‐140‐C18) vesicles |
Mouse | Greater accumulation of SVs at the site of injection. The Bm86 antigen biodistribution was traced in lymph nodes, kidney, and liver, this delivery platform successfully elicits antibody responses | Mody et al. (2021) |
| Bacillus anthracis | Cyclic di‐GMP (CDG), polyanhydride nanoparticle | Mouse | Elicited rapid, high titre and immunization that persisted for at least 108 DPI | Kelly et al. (2021) |
| B. anthracis | Amphiphilic polyanhydride nanoparticles | Mouse | Provide controlled antigen delivery, antigen stability, immune modulation and protection in a single dose against a pathogenic challenge and also can stabilize and release functional protective antigen | Petersen et al. (2012) |
| Brucella | PLGA nanoparticles (LPS‐PLGA and OPS‐PLGA) | Mouse | Immune responses have increased on immunoglobulin production. Immune systems were able to remove more bacteria from the body. So improve the immunization process | Afshari et al. (2020) |
| Escherichia coli | Chitosan (CS) and ascorbate chitosan (AsCS) nanoparticles | Broiler chickens | Improving vaccine efficacy via the induction of strong immunity | Mohammed et al. (2021) |
| Clostridium perfringens type D | Chitosan nanoparticle | Mouse | IgA antibody serum level increased sufficiently and IgG antibody titres increased | Poorhassan et al. (2021) |
| Leptospira interrogans | Halloysite clay nanotubes (HNTs) and amine‐functionalised multi‐walled carbon nanotubes (NH2‐MWCNTs) | Golden Syrian hamsters (Mesocricetus auratus) | Transfecting CHO cells, inducing IgG immune response | Oliveira et al. (2020) |
| Salmonella enteritidis | Outer membrane proteins (OMP) and flagellin (FLA) of SE [CS‐NP (OMP + FLA)] | Broiler chickens | Antigen‐specific splenocyte proliferation, mucosal and systemic antibody response and the frequency of IFN‐γ‐producing T cells were increased; mRNA levels of toll‐like receptor (TLR) 2 and TLR 4, and cytokines IL‐4 and IL‐10 were up‐regulated | Han et al. (2020) |
| Salmonella | Outer membrane proteins (OMPs) and flagellin (F) protein loaded and F‐protein surface coated chitosan nanoparticles (CS NPs) (OMPs‐F‐CS NPs) | Chicken | Enhanced the specific systemic IgG and mucosal IgA antibody responses as well as reduced the challenge Salmonella load in the intestines | Renu, Han et al. (2020) |
| S. enteritidis | Chitosan‐nanoparticle | Broiler chickens | Induce an antigen‐specific immune response against SE and decrease SE caecal load in broilers | Acevedo‐Villanueva et al. (2021) |
| Salmonella | Outer membrane proteins (OMPs) and flagellin (F) protein (OMPs‐F‐CS NPs) | Layer chickens | Increased the expression of toll‐like receptor in chicken caecal tonsils. (Targets immune cells of chickens and induced antigen‐specific B‐ and T‐cell responses) | Renu, Markazi et al. (2020) |
| Mycobacterium tuberculosis | Polymeric nanocapsules (NCs) with an oily core and a polymer shell made of chitosan (CS) or inulin/polyarginine (INU/pArg) | Cell lines? | INU:pArg:Ag NCs induced an adequate immunoglobulin A (IgA) response | Diego‐González et al. (2020) |
| Pseudomonas aeruginosa | Poly lactic‐co‐glycolic acid | Rabbit | Effective in immunization | Safari Zanjani et al. (2018) |
| Brucella melitensis | Poly(lactic‐co‐glycolic acid) (PLGA) | Mouse | Increase in the total IgG and IgM antibody titres and high level of protection | Maleki et al. (2019) |
| Flavobacterium columnare | Mucoadhesive polymer chitosan‐complexed nanovaccine (CS‐NE) | Red tilapia (Oreochromis sp.) | Better adsorption onto the mucosal surfaces, elicited great vaccine efficacy and modulated the MALT immune response. Provided effective delivery system for the induction of a mucosal immune response against columnaris disease in tilapia | Kitiyodom et al. (2021) |
| Staphylococcus aureus | Mesoporous silica nanoparticles (MSN) | Mouse | Improved CD8+ T‐cell responses while maintaining CD4+ T‐cell responses and humoral immunity. Prevented and treated superficial infection and decreased bacterial invasiveness | Chen et al. (2020) |
| Yersinia pestis | Polyanhydride nanoparticle | Mouse | Nanovaccine delivery system enables induction of both rapid and long‐lived protective immunity against Y. pestis | Wagner et al. (2019) |
| Avian pathogenic E. coli (APEC) | Nano‐sized proteolipids enriched with various immunogenic molecules | Broiler chicken | Involvement of both non‐specific immune responses and specific antibody and cytokine responses in the APEC_OMV‐mediated protection | Hu et al. (2020) |
| Mycobacterium avium subsp. paratuberculosis | Polyanhydride nanoparticles (PAN) | Mouse | Increase in the levels of antigen‐specific T‐cell responses. High percentages of triple cytokine (IFN‐γ, IL‐2, TNF‐α) producing CD8+ T cells. Significant levels of double (IFN‐γ, TNF‐α) and single cytokine (IFN‐γ) secreting CD8+ T cells | Thukral et al. (2020) |
| BRSV | Polyanhydride nanoparticles (CPTEG:CPH nanoparticles) | Neonatal calf | Reduced pathology in the lungs, reduced viral burden and decreased virus shedding | McGill et al. (2018) |
| Swine influenza virus | Biodegradable polyanhydride nanoparticles | Mouse | Augmented antigen‐specific cellular immune response in pigs, with promise to induce cross‐protective immunity | Dhakal et al. (2017) |
| Avian influenza virus | Polymer (2‐hydroxyethyl methacrylate) | Hy‐Line chickens | Reduction in virus shedding from the oropharynx | Shan et al. (2010) |
| Infectious bursal disease virus | Unilamellar liposome | Broiler chickens | Elevated humoral antibody titre of IgG, cellular immunity (INF‐γ levels) increased and induce highly protective level of immune response | |
| Rhabdovirus | Targeted carbon nanotubes | Fish | Enhanced uptake by carp macrophages and immune‐related tissues, which would then trigger strong immune responses against spring viremia of carp virus (SVCV) infection | Zhang et al. (2020) |
| Zika virus | Nanoparticles made with a recombinant form of Z. virus nonstructural protein (NP‐∆NS1) | Mouse | Boost antibody production and increase the efficiency of subunit vaccines | Favaro et al. (2021) |
| Classical swine fever virus (CSFV) | Ferritin nanoplatform | Mouse | High ferritin‐assisted humoral and cellular immunities | Zhao et al. (2021) |
| Bovine viral diarrhoea virus (BVDV) | Silica nanoparticles | Mouse | Higher antibody response and higher cell‐mediated response | Mody et al. (2014) |
| Bovine viral diarrhoea virus 1 (BVDV‐1) | Hollow type mesoporous silica nanoparticles with surface amino functionalisation (HMSA) | Sheep | The long‐term cell‐mediated immune responses were detectable up to 4 months. The cell‐mediated immune responses were consistently high. Gives balanced immune responses in a production animal | Mahony et al. (2015) |
| Newcastle disease virus | Chitosan nanoparticles containing the lentogenic live virus vaccine against NDV (NDV‐CS‐NPs) | Chicken | Induced better protection of immunized specific pathogen free chickens | Zhao et al. (2012) |
| Foot‐and‐mouth disease virus | Gold nanoparticles | Guinea pigs | The titre and sensitivity of the raised antibodies were maximal. Antibody biosynthesis was accompanied by increased production of pro‐inflammatory cytokines (especially IFN‐γ) and by stimulation of the respiratory activity of peritoneal macrophages | Dykman et al. (2015) |
| Porcine reproductive and respiratory syndrome virus (PRRSV) | PLGA [poly(lactide‐co‐glycolides)] nanoparticles | Pigs | Reduced the lung pathology and viremia, and the viral load in the lungs. Immunologically, enhanced innate and adaptive immune cell population and associated cytokines with decreased secretion of immunosuppressive mediators were observed at both mucosal sites and blood | Dwivedi et al. (2012) |
| Ovarian cancer | Poly(lactic‐co‐glycolic acid) (PLGA) | Cell lines | Delaying the growth and inhibiting the metastasis of OC | Zhang et al. (2022) |
| Cancer | Poly(d, l‐lactide‐co‐glycolide) (PLGA) nanoparticles | Mouse | Increase antigen cells uptake, increase efficiency to delay tumour development | Yang et al. (2018) |
| Cancer | (InAc‐NPs) using inulin acetate (InAc) | Mouse | Produce storing serum antibody titres against encapsulated antigen. Experimentally, increase cytokines. Complete protection from metastasis of intravenously injected melanoma cells to lungs | Rajput et al. (2018) |
FIGURE 2.

Nanovaccine mechanism of action.
5. CHALLENGES IN THE NANOVACCINES MANUFACTURING PROCESS, QUALITY CONTROL AND REGISTRATION
Considering the cost‐control processes when designing nanovaccines is necessary since NPs production is valuable compared with typical drugs (Pinheiro et al., 2011). The primary challenge in mass‐producing nanovaccines is the high cost of scaling up production and the production process itself. Analysing methods, large‐scale production, in‐process quality control and final product testing all require a significant investment of resources, both in terms of time and money (Usman et al., 2020). Outsourcing the nanovaccine manufacturing line is another way to defeat the barrier, as numerous companies worldwide specialize in various nanoparticles production (Ferrari, 2005). Another possible alternative is collaboration with research and academic organizations. Therefore, the manufacturing process of nanovaccines must reveal adequate strength and consistent quality in mass production (Ahmad et al., 2020). Another challenge is the lack of harmony in quality control tests and manufacturing. For example, there are no standard methods in the pharmaceutical industry for in‐process quality control and the end products because innovative nano‐drugs are typically complex and challenging to separate (Dacoba et al., 2020). Nano‐drugs’ size is defined by the critical quality attribute (CQA). Some factors, such as charge, surface coating, morphology and shape, are other CQA to nano‐drugs (Rawal et al., 2019). Even minor variations in particle size or large size dispersal in the scale might change the product's bioavailability. Furthermore, the small size creates particular conditions for the analytical methods (Fifis et al., 2004). In addition, particle shape can affect the kinetics of cell uptake, the mechanisms of uptake, the level of uptake and intracellular distribution; therefore, toxicity can be changed (Clogston et al., 2020). For instance, the shape of (Au) impacts the nanoparticle cellular internalization (Rodríguez‐León et al., 2019; Salatin et al., 2015; Spadavecchia et al., 2016). In one study, the cellular uptake was most significant for triangle‐shaped particles, and for star‐shaped ones was lowest (Jiang et al., 2012). The surface layer also influences cell capture nanoparticles. For example, phagocytic cells catch nanoparticles with a negative charge faster than neutrals and are removed more quickly than the latter (Djemaa et al., 2018; Sercombe et al., 2015). Meanwhile, nanoparticles with a positive charge can link with negatively charged proteins and stay longer in the blood (Djemaa et al., 2019; Park et al., 2016). One of the major challenges is predicting the relationship between in vivo and in vitro manners of nanoparticles. The main elements that need to be studied using in vivo samples are tissue accumulation, cellular interactions, transportation and biocompatibility (Pokrajac et al., 2021). The study of the accumulation and the long‐time release profile must be idealized due to the persistence of nanoparticles in the blood or their removal in tissues and the lack of knowledge of long‐term impacts (Lowry et al., 2019).
6. NEW DEVELOPMENTS FOR A BRIGHTER FUTURE
Vaccination includes a series of microbiology, immunology, molecular biology, dose application rules, production cost and return on investment (Qin et al., 2021). Creating a production against infections for human and animal immune systems is the highest purpose of any novel vaccination method. Despite having a significant influence on humans, vaccination for veterinary is also concerned with great production and animals’ health. Nanoscience assists with veterinary parasite vaccination, enhancing their potency. Vaccines that use nanoparticles as adjuvants have more power to stimulate both cellular immune reactions and antibody responses (Singh, 2021). In addition, nanotechnology can improve the activation of the immune system by creating greater antibody protection and concentration. This activation occurs by simultaneously triggering the MHC class I and II routes (major histocompatibility complex), or improving the antigen lifetime and their visibility, to reach the host immunity (Beg et al., 2020; Wang et al., 2018). One of the best methods to raise the efficacy of unique and eradicated vaccines is to use unique carriers, including living vectors, micro‐ or nanoparticles and plasmid DNA (Jia et al., 2018; Zhang et al., 2019). The crucial part of natural immunity and the responses of vaccine aids are important points in vaccine production that are often ignored. Understanding the T‐cell immune response is crucial, and studying pathogen‐specific T‐cell immunity could provide valuable insights and signatures to aid in the development of vaccines against infectious diseases (Fajardo et al., 2022). Specific mechanisms for immune signalling must be confirmed for reprogramming of immune cascade. Vaccines are designed innovatively to reach complete protection. Antibody‐dependent enhancement can hinder vaccine development by worsening the immune response. Therefore, it is important to strive for a balanced introduction of neutralizing antibodies, T‐cell immunity against various pathogens and antibody effector functions to optimize vaccine efficacy (Najafi‐Hajivar et al., 2016) (Figure 3). The use of nanoparticles in veterinary vaccines is less restricted compared to human vaccines. This means that there is more flexibility in choosing from a variety of particles with different formulas for veterinary vaccination. In contrast, only a limited number of vaccine particles are currently used for human vaccines.
FIGURE 3.

T‐cell immunity against different pathogens.
7. CONCLUSION
‘Nano‐Vaccinology’ is the science of nano‐sized materials with great capacity. The clinical and laboratory scale can expand beyond the boundaries by a more immunogenic and stabilized release against non‐infectious and infectious diseases. In the storage and synthesis of nanovaccines, quality is a concern in nanovaccines commerce regarding desirable surface effects. For efficiently preventing non‐infectious pathogenic and cancerous diseases in immune‐tolerant patients, the nanovaccines have opened an entrance to boundless hopes. More studies focusing on partnerships with commercial companies lead to the rapid commercializing of nanovaccines.
AUTHOR CONTRIBUTIONS
Conceptualization; supervision: Hassan Borji. Methodology; formal analysis and investigation; writing – original draft preparation; writing – review and editing: Soheil Sadr. Formal analysis and investigation; writing – original draft preparation; writing – review and editing: Parian Poorjafari Jafroodi. Writing – original draft preparation: Mohammad Javad Haratizadeh and Zahra Ghasemi. Funding acquisition: No Funding. Resources: No Resources. Figure Producer: Ashkan Hajjafari.
CONFLICT OF INTEREST STATEMENT
The authors declare no conflicts of interest.
FUNDING INFORMATION
No funding was received for conducting this study.
ETHICS STATEMENT
All applicable international, national and/or institutional guidelines for the care and use of animals were followed.
PEER REVIEW
The peer review history for this article is available at https://publons.com/publon/10.1002/vms3.1221.
ACKNOWLEDGEMENTS
We would like to thank the research deputy of the Ferdowsi University of Mashhad for your support.
Sadr, S. , Poorjafari Jafroodi, P. , Haratizadeh, M. J. , Ghasemi, Z. , Borji, H. , & Hajjafari, A. (2023). Current status of nano‐vaccinology in veterinary medicine science. Veterinary Medicine and Science, 9, 2294–2308. 10.1002/vms3.1221
DATA AVAILABILITY STATEMENT
The data sets generated during and/or analysed during the current study are available from the corresponding author on reasonable request.
REFERENCES
- Acevedo‐Villanueva, K. , Renu, S. , Gourapura, R. , & Selvaraj, R. (2021). Efficacy of a nanoparticle vaccine administered in‐ovo against Salmonella in broilers. PLoS One, 16(4), e0247938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Afshari, H. , Maleki, M. , & Salouti, M. (2020). Immunological effects of two new nano vaccines against Brucella based on OPS and LPS antigens conjugated with PLGA nanoparticles. European Polymer Journal, 139, 110021. [Google Scholar]
- Ahmad, M. Z. , Ahmad, J. , Alasmary, M. Y. , Abdel‐Wahab, B. A. , Warsi, M. H. , Haque, A. , & Chaubey, P. (2021). Emerging advances in cationic liposomal cancer nano vaccines: Opportunities and challenges. Immunotherapy, 13(6), 491–507. [DOI] [PubMed] [Google Scholar]
- Ahmad, M. Z. , Ahmad, J. , Haque, A. , Alasmary, M. Y. , Abdel‐Wahab, B. A. , & Akhter, S. (2020). Emerging advances in synthetic cancer nano‐vaccines: Opportunities and challenges. Expert Review of Vaccines, 19(11), 1053–1071. [DOI] [PubMed] [Google Scholar]
- Alghuthaymi, M. A. , Hassan, A. A. , Kalia, A. , Sayed El Ahl, R. M. , El Hamaky, A. A. , Oleksak, P. , Kuca, K. , & Abd‐Elsalam, K. A. (2021). Antifungal nano‐therapy in veterinary medicine: Current status and future prospects. Journal of Fungi, 7(7), 494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Askarizadeh, A. , Badiee, A. , & Khamesipour, A. (2020). Development of nanocarriers for Leishmania vaccine delivery. Expert Opinion on Drug Delivery, 17(2), 167–187. 10.1080/17425247.2020.1713746 [DOI] [PubMed] [Google Scholar]
- Asouli, A. , Sadr, S. , Mohebalian, H. , & Borji, H. (2023). Anti‐tumor effect of protoscolex hydatid cyst somatic antigen on inhibition cell growth of K562. Acta Parasitologica, 68, 385–392. [DOI] [PubMed] [Google Scholar]
- Azadi, Y. , Ahmadpour, E. , & Ahmadi, A. (2020). Targeting strategies in therapeutic applications of toxoplasmosis: Recent advances in liposomal vaccine delivery systems. Current Drug Targets, 21(6), 541–558. [DOI] [PubMed] [Google Scholar]
- Bai, D.‐P. , Lin, X.‐Y. , Huang, Y.‐F. , & Zhang, X.‐F. (2018). Theranostics aspects of various nanoparticles in veterinary medicine. International Journal of Molecular Sciences, 19(11), 3299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bangham, A. D. , & Horne, R. W. (1964). Negative staining of phospholipids and their structural modification by surface‐active agents as observed in the electron microscope. Journal of Molecular Biology, 8(5), 660–IN10. [DOI] [PubMed] [Google Scholar]
- Beg, S. , Alharbi, K. S. , Alruwaili, N. K. , Alotaibi, N. H. , Almalki, W. H. , Alenezi, S. K. , Altowayan, W. M. , Alshammari, M. S. , & Rahman, M. (2020). Nanotherapeutic systems for delivering cancer vaccines: Recent advances. Nanomedicine, 15(15), 1527–1537. [DOI] [PubMed] [Google Scholar]
- Bhardwaj, P. , Bhatia, E. , Sharma, S. , Ahamad, N. , & Banerjee, R. (2020). Advancements in prophylactic and therapeutic nanovaccines. Acta Biomaterialia, 108, 1–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Billingsley, M. M. , Singh, N. , Ravikumar, P. , Zhang, R. , June, C. H. , & Mitchell, M. J. (2020). Ionizable lipid nanoparticle‐mediated mRNA delivery for human CAR T cell engineering. Nano Letters, 20(3), 1578–1589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bragazzi, N. L. (2019). Nanomedicine: Insights from a bibliometrics‐based analysis of emerging publishing and research trends. Medicina, 55(12), 785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carvalho, S. G. , Araujo, V. H. S. , Dos Santos, A. M. , Duarte, J. L. , Silvestre, A. L. P. , Fonseca‐Santos, B. , Villanova, J. C. O. , Gremião, M. P. D. , & Chorilli, M. (2020). Advances and challenges in nanocarriers and nanomedicines for veterinary application. International Journal of Pharmaceutics, 580, 119214. 10.1016/j.ijpharm.2020.119214 [DOI] [PubMed] [Google Scholar]
- Carvalho, S. G. , Araujo, V. H. S. , Dos Santos, A. M. , Duarte, J. L. , Silvestre, A. L. P. , Fonseca‐Santos, B. , Villanova, J. C. O. , Gremião, M. P. D. , & Chorilli, M. (2020). Advances and challenges in nanocarriers and nanomedicines for veterinary application. International Journal of Pharmaceutics, 580, 119214. [DOI] [PubMed] [Google Scholar]
- Celis‐Giraldo, C. T. , López‐Abán, J. , Muro, A. , Patarroyo, M. A. , & Manzano‐Román, R. (2021). Nanovaccines against animal pathogens: The latest findings. Vaccines, 9(9), 988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cerbu, C. , Kah, M. , White, J. C. , Astete, C. E. , & Sabliov, C. M. (2021). Fate of biodegradable engineered nanoparticles used in veterinary medicine as delivery systems from a one health perspective. Molecules (Basel, Switzerland), 26(3), 523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chariou, P. L. , Ortega‐Rivera, O. A. , & Steinmetz, N. F. (2020). Nanocarriers for the delivery of medical, veterinary, and agricultural active ingredients. ACS Nano, 14(3), 2678–2701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen, F. , Wang, Y. , Gao, J. , Saeed, M. , Li, T. , Wang, W. , & Yu, H. (2021). Nanobiomaterial‐based vaccination immunotherapy of cancer. Biomaterials, 270, 120709. [DOI] [PubMed] [Google Scholar]
- Chen, G. , Bai, Y. , Li, Z. , Wang, F. , Fan, X. , & Zhou, X. (2020). Bacterial extracellular vesicle‐coated multi‐antigenic nanovaccines protect against drug‐resistant Staphylococcus aureus infection by modulating antigen processing and presentation pathways. Theranostics, 10(16), 7131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen, W. , Zuo, H. , Mahony, T. J. , Zhang, B. , Rolfe, B. , & Xu, Z. P. (2017). Efficient induction of comprehensive immune responses to control pathogenic E. coli by clay nano‐adjuvant with the moderate size and surface charge. Scientific Reports, 7(1), 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chouhan, D. , & Mandal, P. (2021). Applications of chitosan and chitosan based metallic nanoparticles in agrosciences—A review. International Journal of Biological Macromolecules, 166, 1554–1569. [DOI] [PubMed] [Google Scholar]
- Chua, B. Y. , Al Kobaisi, M. , Zeng, W. , Mainwaring, D. , & Jackson, D. C. (2012). Chitosan microparticles and nanoparticles as biocompatible delivery vehicles for peptide and protein‐based immunocontraceptive vaccines. Molecular Pharmaceutics, 9(1), 81–90. [DOI] [PubMed] [Google Scholar]
- Chuang, S.‐Y. , Lin, C.‐H. , Huang, T.‐H. , & Fang, J.‐Y. (2018). Lipid‐based nanoparticles as a potential delivery approach in the treatment of rheumatoid arthritis. Nanomaterials, 8(1), 42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clogston, J. D. , Hackley, V. A. , Prina‐Mello, A. , Puri, S. , Sonzini, S. , & Soo, P. L. (2020). Sizing up the next generation of nanomedicines. Pharmaceutical Research, 37(1), 1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cordeiro, A. S. , & Alonso, M. J. (2016). Recent advances in vaccine delivery. Pharmaceutical Patent Analyst, 5(1), 49–73. [DOI] [PubMed] [Google Scholar]
- Cordeiro, A. S. , Alonso, M. J. , & de la Fuente, M. (2015). Nanoengineering of vaccines using natural polysaccharides. Biotechnology Advances, 33(6), 1279–1293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Costa, C. P. , Moreira, J. N. , Sousa Lobo, J. M. , & Silva, A. C. (2021). Intranasal delivery of nanostructured lipid carriers, solid lipid nanoparticles and nanoemulsions: A current overview of in vivo studies. Acta Pharmaceutica Sinica B, 11(4), 925–940. 10.1016/j.apsb.2021.02.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cuzzubbo, S. , Mangsbo, S. , Nagarajan, D. , Habra, K. , Pockley, A. G. , & McArdle, S. E. (2021). Cancer vaccines: Adjuvant potency, importance of age, lifestyle, and treatments. Frontiers in Immunology, 11, 3850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- da Silva, C. F. , Almeida, T. , de Melo Barbosa, R. , Cardoso, J. C. , Morsink, M. , Souto, E. B. , & Severino, P. (2021). New trends in drug delivery systems for veterinary applications. Pharmaceutical Nanotechnology, 9(1), 15–25. [DOI] [PubMed] [Google Scholar]
- Dacoba, T. G. , Ruiz‐Gatón, L. , Benito, A. , Klein, M. , Dupin, D. , Luo, M. , Menta, M. , Teijeiro‐Osorio, D. , Loinaz, I. , & Alonso, M. J. (2020). Technological challenges in the preclinical development of an HIV nanovaccine candidate. Drug Delivery and Translational Research, 10(3), 621–634. [DOI] [PubMed] [Google Scholar]
- Davoodi, B. , Goodarzi, V. , Hosseini, H. , Tirgar, M. , Shojaei, S. , Asefnejad, A. , Saeidi, A. , Oroojalian, F. , & Zamanlui, S. (2022). Design and manufacturing a tubular structures based on poly (ɛ‐caprolactone)/poly (glycerol‐sebacic acid) biodegradable nanocomposite blends: Suggested for applications in the nervous, vascular and renal tissue engineering. Journal of Polymer Research, 29(2), 54. [Google Scholar]
- Dhakal, S. , Goodman, J. , Bondra, K. , Lakshmanappa, Y. S. , Hiremath, J. , Shyu, D.‐L. , Ouyang, K. , Kang, K.‐I. , Krakowka, S. , & Wannemuehler, M. J. (2017). Polyanhydride nanovaccine against swine influenza virus in pigs. Vaccine, 35(8), 1124–1131. [DOI] [PubMed] [Google Scholar]
- Diego‐González, L. , Crecente‐Campo, J. , Paul, M. J. , Singh, M. , Reljic, R. , Alonso, M. J. , González‐Fernández, Á. , & Simón‐Vázquez, R. (2020). Design of polymeric nanocapsules for intranasal vaccination against Mycobacterium tuberculosis: Influence of the polymeric shell and antigen positioning. Pharmaceutics, 12(6), 489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Djemaa, S. B. , David, S. , Hervé‐Aubert, K. , Falanga, A. , Galdiero, S. , Allard‐Vannier, E. , Chourpa, I. , & Munnier, E. (2018). Formulation and in vitro evaluation of a siRNA delivery nanosystem decorated with gH625 peptide for triple negative breast cancer theranosis. European Journal of Pharmaceutics and Biopharmaceutics, 131, 99–108. [DOI] [PubMed] [Google Scholar]
- Djemaa, S. B. , Munnier, E. , Chourpa, I. , Allard‐Vannier, E. , & David, S. (2019). Versatile electrostatically assembled polymeric siRNA nanovectors: Can they overcome the limits of siRNA tumor delivery? International Journal of Pharmaceutics, 567, 118432. [DOI] [PubMed] [Google Scholar]
- Dmour, I. , & Islam, N. (2022). Recent advances on chitosan as an adjuvant for vaccine delivery. International Journal of Biological Macromolecules, 200, 498–519. [DOI] [PubMed] [Google Scholar]
- Doktorovova, S. , Souto, E. B. , & Silva, A. M. (2014). Nanotoxicology applied to solid lipid nanoparticles and nanostructured lipid carriers – A systematic review of in vitro data. European Journal of Pharmaceutics and Biopharmaceutics, 87(1), 1–18. 10.1016/j.ejpb.2014.02.005 [DOI] [PubMed] [Google Scholar]
- Du, G. , & Sun, X. (2020). Engineering nanoparticulate vaccines for enhancing antigen cross‐presentation. Current Opinion in Biotechnology, 66, 113–122. [DOI] [PubMed] [Google Scholar]
- Dwivedi, V. , Manickam, C. , Binjawadagi, B. , Joyappa, D. , & Renukaradhya, G. J. (2012). Biodegradable nanoparticle‐entrapped vaccine induces cross‐protective immune response against a virulent heterologous respiratory viral infection in pigs. PLoS One, 7(12), e51794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dykman, L. A. , Staroverov, S. A. , Mezhenny, P. V. , Fomin, A. S. , Kozlov, S. V. , Volkov, A. A. , Laskavy, V. N. , & Shchyogolev, S. Y. (2015). Use of a synthetic foot‐and‐mouth disease virus peptide conjugated to gold nanoparticles for enhancing immunological response. Gold Bulletin, 48(1), 93–101. [Google Scholar]
- El‐Sayed, A. , & Kamel, M. (2020). Advanced applications of nanotechnology in veterinary medicine. Environmental Science and Pollution Research, 27(16), 19073–19086. [DOI] [PubMed] [Google Scholar]
- Facciolà, A. , Visalli, G. , Laganà, P. , La Fauci, V. , Squeri, R. , Pellicanò, G. , Nunnari, G. , Trovato, M. , & Di Pietro, A. (2019). The new era of vaccines: The “nanovaccinology”. European Review for Medical and Pharmacological Sciences, 23(16), 7163–7182. [DOI] [PubMed] [Google Scholar]
- Fajardo, C. , Martinez‐Rodriguez, G. , Blasco, J. , Mancera, J. M. , Thomas, B. , & De Donato, M. (2022). Nanotechnology in aquaculture: Applications, perspectives and regulatory challenges. Aquaculture and Fisheries, 7(2), 185–200. [Google Scholar]
- Favaro, M. T. P. , Rodrigues‐Jesus, M. J. , Venceslau‐Carvalho, A. A. , Alves, R. P. D. S. , Pereira, L. R. , Pereira, S. S. , Andreata‐Santos, R. , & de Souza Ferreira, L. C. (2021). Nanovaccine based on self‐assembling nonstructural protein 1 boosts antibody responses to Zika virus . Nanomedicine: Nanotechnology, Biology and Medicine, 32, 102334. [DOI] [PubMed] [Google Scholar]
- Fernandes, F. , Dias‐Teixeira, M. , Delerue‐Matos, C. , & Grosso, C. (2021). Critical review of lipid‐based nanoparticles as carriers of neuroprotective drugs and extracts. Nanomaterials, 11(3), 563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferrari, M. (2005). Cancer nanotechnology: Opportunities and challenges. Nature Reviews Cancer, 5(3), 161–171. [DOI] [PubMed] [Google Scholar]
- Fifis, T. , Gamvrellis, A. , Crimeen‐Irwin, B. , Pietersz, G. A. , Li, J. , Mottram, P. L. , McKenzie, I. F. , & Plebanski, M. (2004). Size‐dependent immunogenicity: Therapeutic and protective properties of nano‐vaccines against tumors. The Journal of Immunology, 173(5), 3148–3154. [DOI] [PubMed] [Google Scholar]
- García‐Pinel, B. , Porras‐Alcalá, C. , Ortega‐Rodríguez, A. , Sarabia, F. , Prados, J. , Melguizo, C. , & López‐Romero, J. M. (2019). Lipid‐based nanoparticles: Application and recent advances in cancer treatment. Nanomaterials, 9(4), 638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- George, A. , Shah, P. A. , & Shrivastav, P. S. (2019). Natural biodegradable polymers based nano‐formulations for drug delivery: A review. International Journal of Pharmaceutics, 561, 244–264. [DOI] [PubMed] [Google Scholar]
- Geszke‐Moritz, M. , & Moritz, M. (2016). Solid lipid nanoparticles as attractive drug vehicles: Composition, properties and therapeutic strategies. Materials Science and Engineering: C, 68, 982–994. [DOI] [PubMed] [Google Scholar]
- Guo, Y. , Wang, D. , Song, Q. , Wu, T. , Zhuang, X. , Bao, Y. , Kong, M. , Qi, Y. , Tan, S. , & Zhang, Z. (2015). Erythrocyte membrane‐enveloped polymeric nanoparticles as nanovaccine for induction of antitumor immunity against melanoma. ACS Nano, 9(7), 6918–6933. [DOI] [PubMed] [Google Scholar]
- Han, J. , Zhao, D. , Li, D. , Wang, X. , Jin, Z. , & Zhao, K. (2018). Polymer‐based nanomaterials and applications for vaccines and drugs. Polymers, 10(1), 31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Han, Y. , Renu, S. , Patil, V. , Schrock, J. , Feliciano‐Ruiz, N. , Selvaraj, R. , & Renukaradhya, G. J. (2020). Immune response to Salmonella enteritidis infection in broilers immunized orally with chitosan‐based Salmonella subunit nanoparticle vaccine. Frontiers in Immunology, 11, 935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hari, B. V. , Kalaimagal, K. , Porkodi, R. , Gajula, P. K. , & Ajay, J. Y. (2012). Dendrimer: Globular nanostructured materials for drug delivery. International Journal of PharmTech Research, 4(1), 432–451. [Google Scholar]
- Hu, R. , Liu, H. , Wang, M. , Li, J. , Lin, H. , Liang, M. , Gao, Y. , & Yang, M. (2020). An OMV‐based nanovaccine confers safety and protection against pathogenic Escherichia coli via both humoral and predominantly Th1 immune responses in poultry. Nanomaterials, 10(11), 2293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang, P. , Wang, X. , Liang, X. , Yang, J. , Zhang, C. , Kong, D. , & Wang, W. (2019). Nano‐, micro‐, and macroscale drug delivery systems for cancer immunotherapy. Acta Biomaterialia, 85, 1–26. 10.1016/j.actbio.2018.12.028 [DOI] [PubMed] [Google Scholar]
- Ickenstein, L. M. , & Garidel, P. (2019). Lipid‐based nanoparticle formulations for small molecules and RNA drugs. Expert Opinion on Drug Delivery, 16(11), 1205–1226. [DOI] [PubMed] [Google Scholar]
- Jazayeri, S. D. , Lim, H. X. , Shameli, K. , Yeap, S. K. , & Poh, C. L. (2021). Nano and microparticles as potential oral vaccine carriers and adjuvants against infectious diseases. Frontiers in Pharmacology, 12, 682286. 10.3389/fphar.2021.682286 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jia, J. , Zhang, Y. , Xin, Y. , Jiang, C. , Yan, B. , & Zhai, S. (2018). Interactions between nanoparticles and dendritic cells: From the perspective of cancer immunotherapy. Frontiers in Oncology, 8, 404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang, Q. , Song, C. , Nangreave, J. , Liu, X. , Lin, L. , Qiu, D. , Wang, Z.‐G. , Zou, G. , Liang, X. , Yan, H. , & Ding, B. (2012). DNA origami as a carrier for circumvention of drug resistance. Journal of the American Chemical Society, 134(32), 13396–13403. [DOI] [PubMed] [Google Scholar]
- Kelly, S. M. , Larsen, K. R. , Darling, R. , Petersen, A. C. , Bellaire, B. H. , Wannemuehler, M. J. , & Narasimhan, B. (2021). Single‐dose combination nanovaccine induces both rapid and durable humoral immunity and toxin neutralizing antibody responses against Bacillus anthracis . Vaccine, 39(29), 3862–3870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kheirollahpour, M. , Mehrabi, M. , Dounighi, N. M. , Mohammadi, M. , & Masoudi, A. (2020). Nanoparticles and vaccine development. Pharmaceutical Nanotechnology, 8(1), 6–21. [DOI] [PubMed] [Google Scholar]
- Kheraldine, H. , Rachid, O. , Habib, A. M. , Al Moustafa, A. E. , Benter, I. F. , & Akhtar, S. (2021). Emerging innate biological properties of nano‐drug delivery systems: A focus on PAMAM dendrimers and their clinical potential. Advanced Drug Delivery Reviews, 178, 113908. [DOI] [PubMed] [Google Scholar]
- Kitiyodom, S. , Trullàs, C. , Rodkhum, C. , Thompson, K. D. , Katagiri, T. , Temisak, S. , Namdee, K. , Yata, T. , & Pirarat, N. (2021). Modulation of the mucosal immune response of red tilapia (Oreochromis sp.) against columnaris disease using a biomimetic‐mucoadhesive nanovaccine. Fish & Shellfish Immunology, 112, 81–91. [DOI] [PubMed] [Google Scholar]
- Kumar, R. (2019). Lipid‐based nanoparticles for drug‐delivery systems. In Nanocarriers for drug delivery (pp. 249–284). Elsevier. [Google Scholar]
- Lacasta, A. , Mody, K. T. , De Goeyse, I. , Yu, C. , Zhang, J. , Nyagwange, J. , Mwalimu, S. , Awino, E. , Saya, R. , & Njoroge, T. (2021). Synergistic effect of two nanotechnologies enhances the protective capacity of the Theileria parva sporozoite p67C antigen in cattle. The Journal of Immunology, 206(4), 686–699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lei, X. , Cai, X. , & Yang, Y. (2020). Genetic engineering strategies for construction of multivalent chimeric VLPs vaccines. Expert Review of Vaccines, 19(3), 235–246. [DOI] [PubMed] [Google Scholar]
- Lei, Z. , & Karim, A. (2021). The challenges and applications of nanotechnology against bacterial resistance. Journal of Veterinary Pharmacology and Therapeutics, 44(3), 281–297. [DOI] [PubMed] [Google Scholar]
- Liew, K. B. , Janakiraman, A. K. , Sundarapandian, R. , Khalid, S. H. , Razzaq, F. A. , Ming, L. C. , & Ng, P. W. (2022). A review and revisit of nanoparticles for antimicrobial drug delivery. Journal of Medicine and Life, 15(3), 328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li, J. , Ren, H. , & Zhang, Y. (2022). Metal‐based nano‐vaccines for cancer immunotherapy. Coordination Chemistry Reviews, 455, 214345. [Google Scholar]
- Li, M. , & Zhang, X. (2021). Nanostructure‐based surface‐enhanced raman spectroscopy techniques for pesticide and veterinary drug residues screening. Bulletin of Environmental Contamination and Toxicology, 107(2), 194–205. 10.1007/s00128-020-02989-5 [DOI] [PubMed] [Google Scholar]
- Loera‐Muro, A. , & Angulo, C. (2018). New trends in innovative vaccine development against Actinobacillus pleuropneumoniae . Veterinary Microbiology, 217, 66–75. [DOI] [PubMed] [Google Scholar]
- Lowry, G. V. , Avellan, A. , & Gilbertson, L. M. (2019). Opportunities and challenges for nanotechnology in the agri‐tech revolution. Nature Nanotechnology, 14(6), 517–522. [DOI] [PubMed] [Google Scholar]
- Luo, M. , Wang, H. , Wang, Z. , Cai, H. , Lu, Z. , Li, Y. , Du, M. , Huang, G. , Wang, C. , & Chen, X. (2017). A STING‐activating nanovaccine for cancer immunotherapy. Nature Nanotechnology, 12(7), 648–654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mahony, D. , Mody, K. T. , Cavallaro, A. S. , Hu, Q. , Mahony, T. J. , Qiao, S. , & Mitter, N. (2015). Immunisation of sheep with bovine viral diarrhoea virus, E2 protein using a freeze‐dried hollow silica mesoporous nanoparticle formulation. PLoS One, 10(11), e0141870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maleki, M. , Salouti, M. , Shafiee Ardestani, M. , & Talebzadeh, A. (2019). Preparation of a nanovaccine against Brucella melitensis M16 based on PLGA nanoparticles and oligopolysaccharide antigen. Artificial Cells, Nanomedicine, and Biotechnology, 47(1), 4248–4256. [DOI] [PubMed] [Google Scholar]
- Manjula, N. G. , Sarma, G. , Shilpa, B. M. , & Suresh Kumar, K. (2022). Environmental applications of green engineered copper nanoparticles. In Phytonanotechnology (pp. 255–276). Springer Nature Singapore. [Google Scholar]
- McGill, J. L. , Kelly, S. M. , Kumar, P. , Speckhart, S. , Haughney, S. L. , Henningson, J. , Narasimhan, B. , & Sacco, R. E. (2018). Efficacy of mucosal polyanhydride nanovaccine against respiratory syncytial virus infection in the neonatal calf. Scientific Reports, 8(1), 1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mi, Y. , Hagan, C. T., IV , Vincent, B. G. , & Wang, A. Z. (2019). Emerging nano‐/microapproaches for cancer immunotherapy. Advanced Science, 6(6), 1801847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mody, K. T. , Mahony, D. , Zhang, J. , Cavallaro, A. S. , Zhang, B. , Popat, A. , Mahony, T. J. , Yu, C. , & Mitter, N. (2014). Silica vesicles as nanocarriers and adjuvants for generating both antibody and T‐cell mediated immune resposes to Bovine Viral Diarrhoea Virus E2 protein. Biomaterials, 35(37), 9972–9983. [DOI] [PubMed] [Google Scholar]
- Mody, K. T. , Zhang, B. , Li, X. , Fletcher, N. L. , Akhter, D. T. , Jarrett, S. , Zhang, J. , Yu, C. , Thurecht, K. J. , & Mahony, T. J. (2021). Characterization of the biodistribution of a silica vesicle nanovaccine carrying a Rhipicephalus (Boophilus) microplus protective antigen with in vivo live animal imaging. Frontiers in Bioengineering and Biotechnology, 8, 606652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mohammed, G. M. , ElZorkany, H. E. , Farroh, K. Y. , Abd El‐Aziz, W. R. , & Elshoky, H. A. (2021). Potential improvement of the immune response of chickens against E. coli vaccine by using two forms of chitosan nanoparticles. International Journal of Biological Macromolecules, 167, 395–404. [DOI] [PubMed] [Google Scholar]
- Manocha, S. , Dhiman, S. , Grewal, A. S. , & Guarve, K. (2022). Nanotechnology: An approach to overcome bioavailability challenges of nutraceuticals. Journal of Drug Delivery Science and Technology, 72, 103418. [Google Scholar]
- Najafi‐Hajivar, S. , Zakeri‐Milani, P. , Mohammadi, H. , Niazi, M. , Soleymani‐Goloujeh, M. , Baradaran, B. , & Valizadeh, H. (2016). Overview on experimental models of interactions between nanoparticles and the immune system. Biomedicine & Pharmacotherapy, 83, 1365–1378. [DOI] [PubMed] [Google Scholar]
- Ni, Q. , Zhang, F. , Liu, Y. , Wang, Z. , Yu, G. , Liang, B. , Niu, G. , Su, T. , Zhu, G. , & Lu, G. (2020). A bi‐adjuvant nanovaccine that potentiates immunogenicity of neoantigen for combination immunotherapy of colorectal cancer. Science Advances, 6(12), eaaw6071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Noormehr, H. , Hosseini, A. Z. , Soudi, S. , & Beyzay, F. (2018). Enhancement of Th1 immune response against Leishmania cysteine peptidase A, B by PLGA nanoparticle. International Immunopharmacology, 59, 97–105. [DOI] [PubMed] [Google Scholar]
- Oliveira, T. L. , Bacelo, K. L. , Forster, K. M. , Ilha, V. , Rodrigues, O. E. , & Hartwig, D. D. (2020). DNA nanovaccines prepared using LemA antigen protect Golden Syrian hamsters against Leptospira lethal infection. Memórias do Instituto Oswaldo Cruz, 115, e190396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paleos, C. M. , Tsiourvas, D. , Sideratou, Z. , & Tziveleka, L. A. (2010). Drug delivery using multifunctional dendrimers and hyperbranched polymers. Expert opinion on Drug Delivery, 7(12), 1387–1398. [DOI] [PubMed] [Google Scholar]
- Park, D. H. , Cho, J. , Kwon, O. J. , Yun, C. O. , & Choy, J. H. (2016). Biodegradable inorganic nanovector: Passive versus active tumor targeting in siRNA transportation. Angewandte Chemie International Edition, 55(14), 4582–4586. [DOI] [PubMed] [Google Scholar]
- Petersen, L. , Phanse, Y. , Ramer‐Tait, A. E. , Wannemuehler, M. , & Narasimhan, B. (2012). Amphiphilic polyanhydride nanoparticles stabilize Bacillus anthracis protective antigen. Molecular Pharmaceutics, 9(4), 874–882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pinheiro, A. V. , Han, D. , Shih, W. M. , & Yan, H. (2011). Challenges and opportunities for structural DNA nanotechnology. Nature Nanotechnology, 6(12), 763–772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pokrajac, L. , Abbas, A. , Chrzanowski, W. , Dias, G. M. , Eggleton, B. J. , Maguire, S. , Maine, E. , Malloy, T. , Nathwani, J. , & Nazar, L. (2021). Nanotechnology for a sustainable future: Addressing global challenges with the international network4sustainable nanotechnology. ACS Publications. [DOI] [PubMed] [Google Scholar]
- Poorhassan, F. , Nemati, F. , Saffarian, P. , Mirhosseini, S. A. , & Motamedi, M. J. (2021). Design of a chitosan‐based nano vaccine against epsilon toxin of Clostridium perfringens type D and evaluation of its immunogenicity in BALB/c mice. Research in Pharmaceutical Sciences, 16(6), 575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Prajapati, S. K. , Malaiya, A. , Kesharwani, P. , Soni, D. , & Jain, A. (2022). Biomedical applications and toxicities of carbon nanotubes. Drug and Chemical Toxicology, 45(1), 435–450. [DOI] [PubMed] [Google Scholar]
- Prasad, M. , Ghosh, M. , Kumar, R. , Brar, B. , Surjith, K. , Lambe, U. P. , Ranjan, K. , Banerjee, S. , Prasad, G. , & Kumar Khurana, S. (2021). The importance of nanomedicine in prophylactic and theranostic intervention of bacterial zoonoses and reverse zoonoses in the era of microbial resistance. Journal of Nanoscience and Nanotechnology, 21(6), 3404–3452. [DOI] [PubMed] [Google Scholar]
- Prasad, R. D. , Charmode, N. , Shrivastav, O. P. , Prasad, S. R. , Moghe, A. , Sarvalkar, P. D. , & Prasad, N. R. (2021). A review on concept of nanotechnology in veterinary medicine. ES Food & Agroforestry, 4, 28–60. [Google Scholar]
- Prasanna, P. , Kumar, P. , Kumar, S. , Rajana, V. K. , Kant, V. , Prasad, S. R. , Mohan, U. , Ravichandiran, V. , & Mandal, D. (2021). Current status of nanoscale drug delivery and the future of nano‐vaccine development for leishmaniasis – A review. Biomed Pharmacother, 141, 111920. 10.1016/j.biopha.2021.111920 [DOI] [PubMed] [Google Scholar]
- Qin, L. , Zhang, H. , Zhou, Y. , Umeshappa, C. S. , & Gao, H. (2021). Nanovaccine‐based strategies to overcome challenges in the whole vaccination cascade for tumor immunotherapy. Small, 17(28), 2006000. [DOI] [PubMed] [Google Scholar]
- Rajput, M. K. , Kesharwani, S. S. , Kumar, S. , Muley, P. , Narisetty, S. , & Tummala, H. (2018). Dendritic cell‐targeted nanovaccine delivery system prepared with an immune‐active polymer. ACS Applied Materials & Interfaces, 10(33), 27589–27602. [DOI] [PubMed] [Google Scholar]
- Rawal, M. , Singh, A. , & Amiji, M. M. (2019). Quality‐by‐design concepts to improve nanotechnology‐based drug development. Pharmaceutical Research, 36(11), 1–20. [DOI] [PubMed] [Google Scholar]
- Reddy, P. R. K. , Yasaswini, D. , Reddy, P. P. R. , Zeineldin, M. , Adegbeye, M. J. , & Hyder, I. (2020). Applications, challenges, and strategies in the use of nanoparticles as feed additives in equine nutrition. Veterinary World, 13(8), 1685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rehman, A. , Jafari, S. M. , Tong, Q. , Riaz, T. , Assadpour, E. , Aadil, R. M. , Niazi, S. , Khan, I. M. , Shehzad, Q. , Ali, A. , & Khan, S. (2020). Drug nanodelivery systems based on natural polysaccharides against different diseases. Advances in Colloid and Interface Science, 284, 102251. [DOI] [PubMed] [Google Scholar]
- Renu, S. , Han, Y. , Dhakal, S. , Lakshmanappa, Y. S. , Ghimire, S. , Feliciano‐Ruiz, N. , Senapati, S. , Narasimhan, B. , Selvaraj, R. , & Renukaradhya, G. J. (2020). Chitosan‐adjuvanted Salmonella subunit nanoparticle vaccine for poultry delivered through drinking water and feed. Carbohydrate Polymers, 243, 116434. [DOI] [PubMed] [Google Scholar]
- Renu, S. , Markazi, A. D. , Dhakal, S. , Lakshmanappa, Y. S. , Shanmugasundaram, R. , Selvaraj, R. K. , & Renukaradhya, G. J. (2020). Oral deliverable mucoadhesive Chitosan‐Salmonella subunit nanovaccine for layer chickens. International Journal of Nanomedicine, 15, 761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rodríguez‐León, E. , Rodríguez‐Vázquez, B. E. , Martínez‐Higuera, A. , Rodríguez‐Beas, C. , Larios‐Rodríguez, E. , Navarro, R. E. , López‐Esparza, R. , & Iñiguez‐Palomares, R. A. (2019). Synthesis of gold nanoparticles using Mimosa tenuiflora extract, assessments of cytotoxicity, cellular uptake, and catalysis. Nanoscale Research Letters, 14(1), 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rosales‐Mendoza, S. , & González‐Ortega, O. (2019). Nanovaccines. Springer. [Google Scholar]
- Ross, K. , Senapati, S. , Alley, J. , Darling, R. , Goodman, J. , Jefferson, M. , Uz, M. , Guo, B. , Yoon, K.‐J. , & Verhoeven, D. (2019). Single dose combination nanovaccine provides protection against influenza A virus in young and aged mice. Biomaterials Science, 7(3), 809–821. [DOI] [PubMed] [Google Scholar]
- Safari Zanjani, L. , Shapoury, R. , Dezfulian, M. , Mahdavi, M. , & Shafieeardestani, M. (2018). Preparation of PLGA (poly lactic‐co‐glycolic acid) nanoparticles containing Pseudomonas aeruginosa alginate, LPS and exotoxin A as a nano‐vaccine. Biological Journal of Microorganism, 7(26), 11–27. [Google Scholar]
- Sahoo, M. , Vishwakarma, S. , Panigrahi, C. , & Kumar, J. (2021). Nanotechnology: Current applications and future scope in food. Food Frontiers, 2(1), 3–22. [Google Scholar]
- Sahu, R. , Verma, R. , Dixit, S. , Igietseme, J. U. , Black, C. M. , Duncan, S. , Singh, S. R. , & Dennis, V. A. (2018). Future of human Chlamydia vaccine: Potential of self‐adjuvanting biodegradable nanoparticles as safe vaccine delivery vehicles. Expert Review of Vaccines, 17(3), 217–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salatin, S. , Maleki Dizaj, S. , & Yari Khosroushahi, A. (2015). Effect of the surface modification, size, and shape on cellular uptake of nanoparticles. Cell Biology International, 39(8), 881–890. [DOI] [PubMed] [Google Scholar]
- Samimi, S. , Maghsoudnia, N. , Eftekhari, R. B. , & Dorkoosh, F. (2019). Lipid‐based nanoparticles for drug delivery systems. In Characterization and biology of nanomaterials for drug delivery: Nanoscience and nanotechnology in drug delivery micro and nano technologies (pp. 47–76). Elsevier. [Google Scholar]
- Sanabria, R. (2021). Nanotechnological improvement of veterinary anthelmintics. Pharmaceutical Nanotechnology, 9(1), 5–14. 10.2174/2211738508666200524233724 [DOI] [PubMed] [Google Scholar]
- Sercombe, L. , Veerati, T. , Moheimani, F. , Wu, S. Y. , Sood, A. K. , & Hua, S. (2015). Advances and challenges of liposome assisted drug delivery. Frontiers in Pharmacology, 6, 286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seyfoori, A. , Shokrollahi Barough, M. , Mokarram, P. , Ahmadi, M. , Mehrbod, P. , Sheidary, A. , Madrakian, T. , Kiumarsi, M. , Walsh, T. , & McAlinden, K. D. (2021). Emerging advances of nanotechnology in drug and vaccine delivery against viral associated respiratory infectious diseases (VARID). International Journal of Molecular Sciences, 22(13), 6937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shah, R. R. , O'Hagan, D. T. , Amiji, M. M. , & Brito, L. A. (2014). The impact of size on particulate vaccine adjuvants. Nanomedicine, 9(17), 2671–2681. [DOI] [PubMed] [Google Scholar]
- Shan, S. , Poinern, E. , Ellis, T. , Fenwick, S. , Le, X. , Edwards, J. , & Jiang, Z.‐T. (2010). Development of a nano‐vaccine against a wild bird h6n2 avian influenza virus. Procedia in Vaccinology, 2(1), 40–43. [Google Scholar]
- Shen, Y. , Hao, T. , Ou, S. , Hu, C. , & Chen, L. (2018). Applications and perspectives of nanomaterials in novel vaccine development. MedChemComm, 9(2), 226–238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sherje, A. P. , Jadhav, M. , Dravyakar, B. R. , & Kadam, D. (2018). Dendrimers: A versatile nanocarrier for drug delivery and targeting. International Journal of Pharmaceutics, 548(1), 707–720. [DOI] [PubMed] [Google Scholar]
- Shi, G. N. , Zhang, C. N. , Xu, R. , Niu, J. F. , Song, H. J. , Zhang, X. Y. , Wang, W.‐W. , Wang, Y.‐M. , Li, C. , Wei, X.‐Q. , & Kong, D.‐L. (2017). Enhanced antitumor immunity by targeting dendritic cells with tumor cell lysate‐loaded chitosan nanoparticles vaccine. Biomaterials, 113, 191–202. [DOI] [PubMed] [Google Scholar]
- Singh, A. (2021). Eliciting B cell immunity against infectious diseases using nanovaccines. Nature Nanotechnology, 16(1), 16–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soliman, W. S. , Shaapan, R. M. , Mohamed, L. A. , & Gayed, S. S. (2019). Recent biocontrol measures for fish bacterial diseases, in particular to probiotics, bio‐encapsulated vaccines, and phage therapy. Open Veterinary Journal, 9(3), 190–195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spadavecchia, J. , Movia, D. , Moore, C. , Maguire, C. M. , Moustaoui, H. , Casale, S. , Volkov, Y. , & Prina‐Mello, A. (2016). Targeted polyethylene glycol gold nanoparticles for the treatment of pancreatic cancer: From synthesis to proof‐of‐concept in vitro studies. International Journal of Nanomedicine, 11, 791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Su, T. , Feng, X. , Yang, J. , Xu, W. , Liu, T. , Zhang, M. , Ding, J. , & Chen, X. (2022). Polymer nanotherapeutics to correct autoimmunity. Journal of Controlled Release, 343, 152–174. [DOI] [PubMed] [Google Scholar]
- Sun, B. , Yu, S. , Zhao, D. , Guo, S. , Wang, X. , & Zhao, K. (2018). Polysaccharides as vaccine adjuvants. Vaccine, 36(35), 5226–5234. [DOI] [PubMed] [Google Scholar]
- Tabatabaie, F. , Samarghandi, N. , Zarrati, S. , Maleki, F. , Ardestani, M. S. , Elmi, T. , & Mosawi, S. H. (2018). Induction of immune responses by DNA vaccines formulated with dendrimer and poly (methyl methacrylate)(PMMA) nano‐adjuvants in BALB/c mice infected with Leishmania major . Open Access Macedonian Journal of Medical Sciences, 6(2), 229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tang, W.‐L. , Tang, W.‐H. , & Li, S.‐D. (2018). Cancer theranostic applications of lipid‐based nanoparticles. Drug Discovery Today, 23(5), 1159–1166. [DOI] [PubMed] [Google Scholar]
- Tatli Seven, P. , Seven, I. , Gul Baykalir, B. , Iflazoglu Mutlu, S. , & Salem, A. Z. (2018). Nanotechnology and nano‐propolis in animal production and health: An overview. Italian Journal of Animal Science, 17(4), 921–930. [Google Scholar]
- Thi, T. T. H. , Suys, E. J. , Lee, J. S. , Nguyen, D. H. , Park, K. D. , & Truong, N. P. (2021). Lipid‐based nanoparticles in the clinic and clinical trials: From cancer nanomedicine to COVID‐19 vaccines. Vaccines, 9(4), 359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thukral, A. , Ross, K. , Hansen, C. , Phanse, Y. , Narasimhan, B. , Steinberg, H. , & Talaat, A. M. (2020). A single dose polyanhydride‐based nanovaccine against paratuberculosis infection. Npj Vaccines, 5(1), 1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Usman, M. , Farooq, M. , Wakeel, A. , Nawaz, A. , Cheema, S. A. , ur Rehman, H. , Ashraf, I. , & Sanaullah, M. (2020). Nanotechnology in agriculture: Current status, challenges and future opportunities. Science of the Total Environment, 721, 137778. [DOI] [PubMed] [Google Scholar]
- Vinay, T. N. , Bhat, S. , Gon Choudhury, T. , Paria, A. , Jung, M.‐H. , Shivani Kallappa, G. , & Jung, S.‐J. (2018). Recent advances in application of nanoparticles in fish vaccine delivery. Reviews in Fisheries Science & Aquaculture, 26(1), 29–41. [Google Scholar]
- Wagner, D. A. , Kelly, S. M. , Petersen, A. C. , Peroutka‐Bigus, N. , Darling, R. J. , Bellaire, B. H. , Wannemuehler, M. J. , & Narasimhan, B. (2019). Single‐dose combination nanovaccine induces both rapid and long‐lived protection against pneumonic plague. Acta Biomaterialia, 100, 326–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang, K. , Wen, S. , He, L. , Li, A. , Li, Y. , Dong, H. , Li, W. , Ren, T. , Shi, D. , & Li, Y. (2018). “Minimalist” nanovaccine constituted from near whole antigen for cancer immunotherapy. ACS Nano, 12(7), 6398–6409. [DOI] [PubMed] [Google Scholar]
- Wang, Q. , Muhammad, T. A. , Muhammad, W. H. , Muhammad, A. M. , Muhammad, H. , Yan, R. , Xu, L. , Song, X. , & Li, X. (2021). Haemonchus contortus hepatocellular carcinoma‐associated antigen 59 with poly (lactic‐co‐glycolic acid): A promising nanovaccine candidate against Haemonchus contortus infection. Veterinary Parasitology, 292, 109398. [DOI] [PubMed] [Google Scholar]
- Wang, T. , & Luo, Y. (2019). Biological fate of ingested lipid‐based nanoparticles: Current understanding and future directions. Nanoscale, 11(23), 11048–11063. [DOI] [PubMed] [Google Scholar]
- Wang, T. , Suita, Y. , Miriyala, S. , Dean, J. , Tapinos, N. , & Shen, J. (2021). Advances in lipid‐based nanoparticles for cancer chemoimmunotherapy. Pharmaceutics, 13(4), 520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woldeamanuel, K. M. , Kurra, F. A. , & Roba, Y. T. (2021). A review on nanotechnology and its application in modern veterinary science. International Journal of Nanomaterials, Nanotechnology and Nanomedicine, 7(1), 026–031. [Google Scholar]
- Xiang, S. D. , Scholzen, A. , Minigo, G. , David, C. , Apostolopoulos, V. , Mottram, P. L. , & Plebanski, M. (2006). Pathogen recognition and development of particulate vaccines: Does size matter? Methods (San Diego, California), 40(1), 1–9. [DOI] [PubMed] [Google Scholar]
- Yadav, H. K. , Dibi, M. , Mohammad, A. , & Srouji, A. E. (2018). Nanovaccines formulation and applications—A review. Journal of Drug Delivery Science and Technology, 44, 380–387. [Google Scholar]
- Yang, M. , Li, J. , Gu, P. , & Fan, X. (2021). The application of nanoparticles in cancer immunotherapy: Targeting tumor microenvironment. Bioactive Materials, 6(7), 1973–1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang, R. , Xu, J. , Xu, L. , Sun, X. , Chen, Q. , Zhao, Y. , Peng, R. , & Liu, Z. (2018). Cancer cell membrane‐coated adjuvant nanoparticles with mannose modification for effective anticancer vaccination. ACS Nano, 12(6), 5121–5129. [DOI] [PubMed] [Google Scholar]
- Youssef, F. S. , El‐Banna, H. A. , Elzorba, H. Y. , & Galal, A. M. (2019). Application of some nanoparticles in the field of veterinary medicine. International Journal of Veterinary Science and Medicine, 7(1), 78–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yun, C.‐H. , & Cho, C.‐S. (2020). Nanoparticles to improve the efficacy of vaccines. Pharmaceutics, 12, 418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zaheer, T. , Pal, K. , & Zaheer, I. (2021). Topical review on nano‐vaccinology: Biochemical promises and key challenges. Process Biochemistry, 100, 237–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zarrati, S. , Maleki, F. , Mahdavi, M. , Khabaz zadeh Tehrani, N. , Abrehdari Tafreshi, Z. , Asadi, A. , & Tabatabaie, F. (2014). Humoral immune responses in DNA vaccine formulated with poly [methyl methacrylate] against Leishmania major . India Journal of Entomology and Zoology Studies, 2(5), 201–206. [Google Scholar]
- Zhang, C. , Wang, G.‐X. , & Zhu, B. (2020). Application of antigen presenting cell‐targeted nanovaccine delivery system in rhabdovirus disease prophylactics using fish as a model organism. Journal of Nanobiotechnology, 18(1), 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang, L. , Zhao, W. , Huang, J. , Li, F. , Sheng, J. , Song, H. , & Chen, Y. (2022). Development of a dendritic cell/tumor cell fusion cell membrane nano‐vaccine for the treatment of ovarian cancer. Frontiers in Immunology, 13, 828263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang, Y. , Lin, S. , Wang, X. Y. , & Zhu, G. (2019). Nanovaccines for cancer immunotherapy. Wiley Interdisciplinary Reviews: Nanomedicine and Nanobiotechnology, 11(5), e1559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao, K. , Chen, G. , Shi, X.‐M. , Gao, T.‐T. , Li, W. , Zhao, Y. , Zhang, F.‐Q. , Wu, J. , Cui, X. , & Wang, Y.‐F. (2012). Preparation and efficacy of a live Newcastle disease virus vaccine encapsulated in chitosan nanoparticles. PLoS One, 7(12), e53314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao, L. , Mahony, D. , Cavallaro, A. S. , Zhang, B. , Zhang, J. , Deringer, J. R. , Zhao, C.‐X. , Brown, W. C. , Yu, C. , & Mitter, N. (2016). Immunogenicity of outer membrane proteins VirB9‐1 and VirB9‐2, a novel nanovaccine against Anaplasma marginale . PLoS One, 11(4), e0154295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao, Z. , Chen, X. , Chen, Y. , Li, H. , Fang, K. , Chen, H. , Li, X. , & Qian, P. (2021). A self‐assembling ferritin nanoplatform for designing classical swine fever vaccine: Elicitation of potent neutralizing antibody. Vaccines, 9(1), 45. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The data sets generated during and/or analysed during the current study are available from the corresponding author on reasonable request.
