5,271
Views
90
CrossRef citations to date
0
Altmetric
Reviews

Current prospects and future challenges for nasal vaccine delivery

&
Pages 34-45 | Received 07 Jul 2016, Accepted 18 Sep 2016, Published online: 09 Dec 2016

ABSTRACT

Nasal delivery offers many benefits over traditional approaches to vaccine administration. These include ease of administration without needles that reduces issues associated with needlestick injuries and disposal. Additionally, this route offers easy access to a key part of the immune system that can stimulate other mucosal sites throughout the body. Increased acceptance of nasal vaccine products in both adults and children has led to a burgeoning pipeline of nasal delivery technology. Key challenges and opportunities for the future will include translating in vivo data to clinical outcomes. Particular focus should be brought to designing delivery strategies that take into account the broad range of diseases, populations and healthcare delivery settings that stand to benefit from this unique mucosal route.

In this review the current state of the art in nasal vaccine delivery will be described along with future prospects. A brief introduction to the anatomy and physiology of the nasal cavity will highlight the advantages and disadvantages of the route. Encapsulation and presentation methods along with particular formulation considerations for the nasal route will also be discussed.

There are many mucosal routes which have been regarded as potential sites for vaccine delivery such as oral, nasal, pulmonary, conjunctival, rectal and vaginal mucosa. However, for practical and cultural reasons researchers have tended to focus only on oral, nasal, and pulmonary administration.Citation1 Needle-free vaccines offer many advantages over traditional vaccination approaches including convenience, cost, ease of administration and disposal.

There are several needle free methods of vaccination such as transdermal delivery and mucosal delivery.Citation2,3 Mucosal immunization has been successfully used in human vaccination. The human mucosal immune system is large and specialized in performing inspection for foreign antigens to protect the surfaces themselves and of course human body interior. Since most infections affect or start from mucosal surfaces, using a mucosal route of vaccination is of great interest and provides a rational reason to induce a protective immune response.Citation3 Nasal delivery of vaccine offers an easily accessible route to the immune system.

The nose has the function of olfactory detection (sense of smell) and also filtration, humidification and temperature control of air as it enters the respiratory system. Moving from front to back the areas of the nasal cavity are the nasal vestibule, the respiratory region, and the olfactory region. The nasal cavity is divided by the septum to form the left and right nares, which lead into the left and right choana before opening onto the nasopharynx at the top of the throat. The turbinates bound the nasal walls and are responsible for air conditioning and the large mucosal surface area of the nasal cavity. The nose is also the main port of entry for many pathogens. The first barrier to foreign bodies is hair at the entrance to the nares, the nostrils, which successfully keeps out larger particles. The entire surface of the nasal cavity is covered in a mucus layer, which traps smaller particles. Mucus is an aqueous, viscoelastic and adhesive gelCitation4 that contains several types of mucins (abbreviated to MUC) MUC1, MUC4, MUC5A and MUC5B, MUC16, that are produced by either goblet cells or mucus subglands.Citation5,6 Cilia perform a mechanical clearing role termed mucociliary clearance by beating and thus transporting the mucus blanket with entrapped pathogens to the back of the throat at a rate of 5–6 mm per minute, either to be destroyed in the stomach or expectorated via sneezing and/or coughing. This function minimises the amount of particles able to enter the body through the mucosal surface.Citation7 The nasal route has been used to deliver vaccines for respiratory infections and sexually transmitted infections.Citation8 The rationale for targeting mucosal tissue in the genital tracts can be attributed to the mucosal immune system.

The mucosal immune system

The mucosal immune system provides local protection against pathogens that enter the body through the mucosal membranes. The mucosal immune activities are associated with lymphoid tissues, i.e. mucosa-associated lymphoid tissue (MALT), which is present in mucosal tissue in the nose, lungs, gastrointestinal tract and vaginal/rectal surfaces.Citation9 The MALT is classified into specific subcompartments, depending on the location, including the gut-associated lymphoid tissue (GALT), nasopharynx-associated lymphoid tissue (NALT),Citation10 bronchus-associated lymphoid tissue (BALT). The mucosal routes commonly used for vaccination strategies are depicted in . The mucosal immune systems are protected by immune cells that populate the region along the mucosal surfaces, and also epithelial cells and mucus that acts as physical barrier before the pathogen gain access to the underlying tissues.

Figure 1. Routes of mucosal vaccination within the mucosa-associated lymphoid tissue (MALT), with several subcompartments including: the nasopharynx-associated lymphoid tissue (NALT), bronchus-associated lymphoid tissue (BALT), gut-associated lymphoid tissue (GALT) and genital tract-associated lymphoid tissue, reproduced from Lycke et al, 2012.Citation125

Figure 1. Routes of mucosal vaccination within the mucosa-associated lymphoid tissue (MALT), with several subcompartments including: the nasopharynx-associated lymphoid tissue (NALT), bronchus-associated lymphoid tissue (BALT), gut-associated lymphoid tissue (GALT) and genital tract-associated lymphoid tissue, reproduced from Lycke et al, 2012.Citation125

Respiratory epithelial cells

The epithelial cell layers cover the mucosal surfaces including the respiratory, gastrointestinal and urogenital tracts exposed to the outer environments. The epithelial cell layer acts as a barrier that is equipped with some supporting elements such as the mucus and cilia in preventing penetration of pathogens ().

Figure 2. Structure and function of respiratory epithelial cells; equipped with mucus layer (not shown) and ciliated cells, reproduced from Grassin-Delyle (2012)Citation143.

Figure 2. Structure and function of respiratory epithelial cells; equipped with mucus layer (not shown) and ciliated cells, reproduced from Grassin-Delyle (2012)Citation143.

Furthermore, the epithelial cells can detect and uptake pathogenic organisms and/or antigenic components by performing nonspecific endocytosis or interacting with pattern recognition receptors such as Toll-like receptors (TLRs).Citation11-14 The epithelial cells together with lymphocytes and underlying antigen presenting cells (e.g. dendritic cells (DCs) and macrophages), cytokines and chemokines perform an innate, non-specific and adaptive immune response to encounter the invasion of pathogenic organisms or immunogenic substances.Citation14,15

Nasopharynx-associated lymphoid tissue (NALT)

The NALT can be simply defined as organized mucosal immune system in the nasal mucosa that consist of lymphoid tissue, B cells, T cells and antigen presenting cells (APCs) and are covered by an epithelial layer containing memory (M) cells.Citation16 M cells are present in the epithelial cell layers and have specialization in transporting antigen across the epithelium.Citation17,18

Whenever the nasal mucosa is exposed to pathogens or antigenic substances, the intruder will interact with the mucosal immune system. The type of interaction is highly dependent on the characteristics of the antigen. The pathogen or immunogenic substances may be able to pass through the nasal epithelium and interact with the APCs such as macrophages and DCs. These APCs will process the antigen and migrate to the lymph node where the immunogenic portion will be presented to the T cells. This marks the activation of the immune response cascade. A soluble antigen might be recognized by the APCs,Citation19 while particulate antigen is generally taken up by the M cells and transported to the NALT.Citation20 The NALT is also drained to the lymph node where further antigen processing will occur. A schematic representation of this process in more detail mechanisms is presented in .Citation21

Figure 3. Pathways demonstrating how particulate antigen triggers local immune response in the nasal mucosa and systemic immune response via the NALT, adapted from Csaba (2009)Citation21.

Figure 3. Pathways demonstrating how particulate antigen triggers local immune response in the nasal mucosa and systemic immune response via the NALT, adapted from Csaba (2009)Citation21.

Immunoglobulin A (IgA)

In addition to the MALT, the mucosal immune system also produces the antibody immunoglobulin A (IgA), that plays an important role in mucosal immunity at mucosal surfaces.Citation22 IgA constitutes up to 15 % of the total immunoglobulin, which is predominantly present in external secretions including the mucus in the bronchial, urogenital and digestive tracts, saliva and tears.Citation23 It was found that the production of IgA in humans could be over 1 mg/ml in secretions associated with the mucosal surfaces.Citation18 A small amount of IgA can be found in the serum while most of the IgA is located in external secretions known as secretory IgA (sIgA).Citation24 IgA consist of a dimer or tetramer, a joining J-chain polypeptide and a polypeptide chain called the secretory component.Citation24,25 IgA has several functions in mucosal defense including the entrapment of antigens or pathogens in mucus to prevent them from direct contact with the mucosal surface.Citation15,26 In addition, sIgA may also block or provide steric hindrance to surfaces of pathogenic molecules that may inhibit their attachment to the epithelium.Citation27

The predominance of IgA in mucosal areas is a result of mutual collaboration between plasma cells and epithelial cells. The activated plasma cells in the lamina propria, adjacent to mucosal surfaces produce polymeric IgA (pIgA), while the epithelial cells in the mucosal surfaces express an Ig receptor called the polymeric Ig receptor (pIgR). The released pIgA from activated plasma cells binds to pIgR, and is then taken up into the cell via endocytosis. IgA is transported across mucosal epithelial cells before being released onto the luminal surface of the epithelial cells. Proteolysis cleavage of the pIgR allows IgA to be secreted into mucosal secretions.Citation15,25,28

Mucosal vaccines

New vaccine formulations should be able to induce innate and adaptive immune response; involving antigen-specific memory T and B cells that will respond effectively to the invading pathogens.Citation29,30 Interaction with pathogens or antigens can produce the IgA secretion as an antibody response.Citation31 Intracellular antigens, can be produced by invading viruses that replicate within the host cell, or derive from cytoplasmic bacteria, while the extracellular antigens include bacteria, parasites, and toxins in the tissues. Intracellular antigens are generally processed in the host cells, coupled to a major histocompatibility complex-I (MHC-I), a cell surface molecule, and transported to the cell surface.Citation32,32 The presence of MHC-I on the cell surface will lead to activation of CD8+ T-cells to become cytotoxic T-lymphocytes (CTLs). Extracellular antigens are endocytosed and presented on MHC-II molecules for activation of CD4+ T-helper (Th) cells.Citation32-34

The activation of Th cells will release a specific set of cytokines that modulate the B cell and CD8+ CTL immune response, depending on the nature of the stimulant.Citation35 Th cell types Th-1, Th-2 or Th-17 will be induced accordingly. A Th-1 response develops in the presence of interleukin 12 (IL-12), which is in turn synthesized primarily by DCs and/or natural killer (NK) cells in the presence of bacteria or virus. The Th-1 response is marked by the production of the Th-1 cytokines e.g., interferon-gamma (IFN-γ) and tumor necrosis factor-β (TNF-β). A Th-2 response is driven by the presence of IL-4 and results in the production of specific cytokines IL-4, IL-5, IL-9 and IL-13.Citation36 It can be seen that the production of IL-4 generates a feedback loop that results in increased generation of a Th-2 response at the local site.

Nasal vaccination can also result in stimulation of Th-17 CD4+ cells. Th-17 cells are responsible for the secretion of the proinflammatory interleukins IL-17A and IL-22, as well as IL-17F and IL-21. It Is known that the Th-17 family of cytokines respond to extracellular bacterial and fungal pathogens, and Th-17 cells enhance generation of Th-1 cells through an increased IFN-γ activation giving rise to a Th-1/Th-17 immune response that activates macrophages and other innate responses.Citation36-38 Stimulation of epithelial cells by the Th-17 family of cytokines can aid tissue repair and secretion of antimicrobial peptides, which can exert a protective effect in pulmonary infection.Citation39 There is contradictory evidence, however, regarding the role of Th-17 response in nasal immunization. Early work on the role of Th polarization in nasal immunization indicated that this route always promotes a Th-17 response.Citation40 Later research has indicated that the response is more nuanced, with some contradictory evidence regarding advantages and disadvantages of IL-17A induction.Citation41,42,43

Predominance of one set of cytokines over the other is generally indicative of polarization of Th responses, for example the presence of IL-4 and absence of IFN-γ indicate a classical Th-2 polarized immune reactionCitation44 although these cytokines can also be released at the same time.Citation45,46,47 The varying cytokine profiles related to CTL and antibody production are fundamental in affording protection against a specific pathogen. Specific macrophage activation was found to play a crucial role in the eradication of Mycobacterium tuberculosis bacterial infections,Citation48 showing that the induction of specific immune responses may play a key role in determining whether a given vaccine product is effective.

The recently discovered innate lymphoid cells (ILCs) act as an early source of cytokines to regulate and direct mucosal immune responses.Citation49 Unlike B or T cells, however, they do not exhibit antigen specificity. Group 1 ILCs (ILC1s) include NK cells and produce Th-1 type cytokines IFN-γ and tumor necrosis factor-α (TNF-α ); group 2 ILCs (ILC2s) produce Th-2 type cytokines IL4, IL-5 and/or IL-13, while group 3 ILCs (ILC3s) include lymphoid tissue inducer cells that produce Th-17 type cytokines IL-17 and/or IL-22. Both ILC1s and ILC3s have been implicated in type 1 and Th17 cell-mediated immunity and disease.Citation50 Because they are involved in early release of cytokines at mucosal sites, ILCs have been implicated in directing immune response at the mucosal surface, as shown by a number of recent studies.Citation51,52 NK cells and ILC1-like cells damped the immune response after vaginal administration of ovalbumin and cholera toxin to mice.Citation53 NK cells have been shown to enhance Th proliferation through IFN-γ production,Citation54 while ILC2s play a role in directing Th-2 response.Citation55 There is also evidence that ILCs can act as APCs, although this may be specific to the lymphoid tissue site involved and is thought to occur to a lesser extent than through the professional APCs.Citation55 Finally the regulatory T-cells (Tregs) play a role in ILC and Th communication,Citation54 as well as helping to directly control Th response, which is particularly important in autoimmune dysfunction discussed later.Citation56

Advantages of nasal vaccine delivery

The nasal route has great potential for vaccination due to the organized immune systems of the nasal mucosa. The nasal epithelium encloses follicle-associated lymphoid tissues that are important in inducing mucosal immune response. The immune cells such as nearby B-cells can produce IgA at the mucosal sites where the respiratory pathogens invade.Citation57 Many published studies have shown that nasally administered vaccines induce serum IgG and mucosal IgA that are important for deliberating enhanced efficacy of vaccine.Citation57,58 The enhanced induction of mucosal IgA antibodies has been shown to play a significant role in neutralizing pathogens such as Streptococcus pneumoniaCitation59 and measles virusesCitation60 and preventing further infection. Moreover, intranasal immunization has also been reported to induce cross-reactive antibodies that might be indicative of cross-protection.Citation61,62 This effect can make vaccines more efficient by reducing the number of vaccinations required since cross-protective vaccines may produce cross-reactive antibodies that recognize more than one antigen. Given the high cost of many antigen production systems this offers a distinct advantage over other routes.

Therapeutic vaccines

While much of the work on nasal vaccine delivery is currently focused on prophylactic vaccines, the access that the nasal route provides to the mucosal immune system also has relevance for therapeutic vaccines used to treat rather than prevent disease. Nasal immunotherapy for treatment of various cancers and Alzheimer are currently generating much interest.Citation63,64 A particular focus is the use of therapeutic vaccines for the treatment of autoimmune diseases such as type I diabetes, atherosclerosis, multiple sclerosis, rheumatoid arthritis, lupus and Crohn's disease. These are caused by unchecked immune response to molecules, termed self-antigens, that are capable of inducing an immune response in a host but should not induce an immune response in a healthy individual that produces them, whereas undesirable response to innocuous environmental antigens gives rise to allergy. The autoimmune and inflammatory response is governed by regulatory T-cells (Tregs), with poor function or reduced numbers of Tregs being associated with autoimmune disease. Treatments for this family of diseases are often non-specific, or use immune suppressants that increase susceptibility to infection. Development of effective therapeutic vaccine would correct the inappropriate immune response through generation of tolerance to the self-antigen(s).Citation65 Treg cells that express the forkhead box P3 transcription factor are known as FoxP3+T-cells, with dysfunction of this subset of Tregs being implicated in a range of chronic inflammatory disorders.Citation66 It has long been known that oral delivery is effective in generating antigen tolerance, through deliberate introduction of the antigen to food.Citation67 More recently it has been shown that a similar tolerance induction can be achieved via nasal delivery through activation of the DCs in the draining lymph nodes to enhance induction of FoxP3+T-cells.Citation68 Examples of successful nasal delivery include immunization to suppress atherosclerosisCitation69,70 and arthritis.Citation71 The effect of adjuvant on tolerance is discussed in a later section.

Formulation approaches

Current nasal formulations include, solutions (drops or sprays), powders, gels and solid inserts.Citation72 Solutions are often described in the literature as they are both the easiest way of formulating a vaccine for an in vivo study or clinical trial, and are the easiest to administer for example in mice where the liquid is often pipetted directly into the nostril. In humans this often means that the subject either has to remain laying down or with their head held back for a period of time after administration, which is not realistic in a mass vaccination setting. Sprays are easier to administer and deliver vaccine further into the nasal cavity, but may still leak out of the nostril or drip into the oral cavity. Including a gelling agent in the formulation that is either mucoadhesive or able to penetrate through mucus offers increased residence time, while advantages of solid formats such as powders or solid inserts include ease of manufacture and stability, while liquids are more prone to degradation. Taste may also be a factor as formulations may travel into the oral cavity, although given that vaccines tend to be administered once or twice only, this is less of an issue than for medicines that are taken on a regular basis.

A range of naturally-occurring, synthetic and semi-synthetic polymers have been investigated as gelling agents in nasal delivery of vaccine. Administering as a gel should improve retention, although there is ongoing debate as to whether positively charged or anionic polymers offer better uptake. Those that have the ability to adhere to mucosal surfaces and selectively target M cells or APCs, should be the most effective.Citation18,26 Chitosan has been much investigated, and is a polysaccharide manufactured from chitin found in crustacean shells or fungi by a deacetylation process. Because of the range of sources this polymer is available in a range of molecular weights, but all are made up of repeating units of glucosamine and N-acetylglucosamine and bear a positive charge making it mucoadhesive. Varying the degree of deacetylation affects the charge, as does methylation. Methylating chitosan offers some advantages for mucosal delivery.

Powder formats have the advantage of increased stability over their liquid counterparts and ability to target further into the nasal cavity. An example of this is the Anthrax spray-dried powder formulation suitable for mass vaccination in developed and developing world settings.Citation73 Possible disadvantages of powders include the ease and cost of administration if specialist applicators are required. Solid inserts are tablets designed to dissolve when in contact with mucus and have been investigated for vaginal delivery in humans and nasal delivery in livestock animals,Citation74,75 and have many similarities with sublingual formulations.

Soluble antigens tend to be less immunogenic than particulate formulations, additionally encapsulating antigen into particles may improve the transport of the antigens across the nasal mucosa. For this reason there has been a great interest in developing particulate systems as carriers for vaccine products.Citation76-78 Aspects such as vaccine formulations and delivery strategies are important in designing new vaccines so that efficient induction of the innate and adaptive immune response can be obtained according to the target pathogen.Citation18,26 Particulate delivery systems that can imitate pathogens such as polymeric nanoparticles and liposomes are considered a promising approach for nasal vaccine delivery.

Nanoparticles are particles in the nanometer 1×10−9 m size range and can be made of polymers such as chitosan, alginate or synthetic co-polymers such as poly(lactic-co-glycolic acid (PLGA). Varying the molecular weight and/or ratio of lactic to glycolic acid affects the rate of degradation enabling rate of release to be controlled. But PLGA nanoparticles bear a negative charge, which is not compatible with mucosal delivery, hence the plethora of papers investigating various coatings or modifications to adjust this. Those with positive charge and enhanced residence have tended to give the best immunological responses with high serum antibody titers and sIgA levels.Citation79 Poly(lactic acid) (PLA) and polyethylene glycol (PEG) can also be combined to form co-block polymers able to incorporate antigen,Citation80 varying the molecular weight of the PEG and/or ratio of PEG to PLA alters physicochemical characteristics, release and hence efficacy.Citation81

Other polymers investigated include pullulan, a naturally occurring polysaccharide copolymer made up of maltotriose subunits from fungus;Citation82 pectin, a naturally occurring polysaccharide found in fruits; and the biodegradable synthetic polymer polycaprolactone.Citation83

Liposomes are nano- or micrometre sized particles made up of one or more lipid bilayers, which have the ability to incorporate antigen at their surface or inside the aqueous core. There are numerous examples of coated and un-coated liposomal formulations used to deliver vaccine intranasally in a range of formats.Citation84-90 Chen showed that trimethylchitosan-coated liposome powders offered improved uptake in ex vivo nasal penetration studies when compared with the same liposomes coated in chitosan.Citation91 Liposomes that also comprise lipid or other material derived from virus are known as virosomes, with material from influenza virus such as hemagglutinin (HA) and neuraminidase being commonly used.Citation92-102

Currently there is more evidence to support the hypothesis that particles smaller than 300nm are the most effective at crossing mucus,Citation103 but there is also evidence to suggest that larger particles are also able to penetrate. Results from intranasal administration of mucoadhesive microparticles suggest that penetration of the entire particle may not be necessary to induce an immune response.Citation104 It is likely that the overall combination of size and charge are key to achieving maximum immunological effect. Some examples of particulate delivery systems investigated for nasal delivery of vaccine are shown in .

Table 1. Examples of particulate formulations with published in vivo data.

Adjuvants

Some materials added to form gels or particles may act as adjuvants as well as delivery vehicles. Alternatively, adjuvants may be added as a separate component to a vaccine product. Adjuvants are materials added to a vaccine to boost the immune response and may also reduce the amount of antigen required to elicit an immune response. Alum is often used in traditional vaccines but is not effective when administered mucosally. Judicious choice of adjuvant can direct the arm of the immune system, as described previously. Often particulate delivery systems are believed to confer both the benefits of optimised delivery across mucus/mucosal tissue and inherent adjuvanting effects. Many studies have investigated these abilities and ascribed immune boosting response to one, other or both qualities.Citation26

Mucosal adjuvants that have been tested for intranasal vaccine delivery including: MF59 emulsion (containing squalene oil, the surfactants Span 85 and Tween 80 and citrate buffer),Citation105,106 lipopolysaccharide,Citation84,107 TLR agonists,Citation41,108,109 chitosan,Citation110 trimethylchitosan,Citation91,Citation110 bacterial outer membrane proteinCitation111 and cholera toxinCitation112 or heat-labile enterotoxin (LT) from E.coli.Citation113 Some side effects have been found with the use of bacterial toxin when given intranasally, including Bell's palsy (Facial paralysis) and other adverse events related to disorders of the facial nerves.Citation114-116 It has been suggested that the central nervous system was involved in the palsy as the bacterial toxin was re-directed into the brain.Citation115,117 Thus, the use of LT as vaccine adjuvant is no longer recommended. Mast cell activators such as compound 48/80 (C48/80) have shown promise in Anthrax vaccine.Citation73 As described previously, adjuvants can help to polarize immune response and this effect should be taken into account when considering adjuvant for a particular vaccine type. Mice immunized with an influenza vaccine adjuvanted with a synthetic TLR-4 agonist via the nasal route, exhibited a transient, enhanced IL-17A pathology, characterized by weight loss and morbidity, which was significantly greater than observed in mice given no-adjuvanted antigen.Citation41 The effect of adjuvants on induction of tolerance has also been noted; an intranasal co-administration of hen egg lysozyme with a TLR2 ligand enhanced Th1-type antibodies in one case,Citation118 while another TLG2 ligand, Pam3Cys, was shown to increase the risk of developing autoimmune diseaseCitation119 PLGA nanoparticles have been shown to boost tolerance in suppression of arthritisCitation120 and further research by the same group has shown that they are responsible for generation of enhanced Treg cell induction.Citation68

Current nasal vaccine products

Licensed intranasal vaccines for humans include the influenza vaccines FluMist/Fluenz™ (MedImmune, MD, USA)Citation121 and the Nasovac™ live attenuated influenza nasal spray manufactured by the Serum Institute of India, which was developed alongside its live attenuated A(H1N1), more commonly known as swine flu.Citation122 No serious side effects have been reported associated with the administration of Nasovac indicating its safety,Citation123 although its efficacy data are not sufficiently available yet.Citation124 Until recently FluMist was considered one of the most successful intranasal vaccines, it is well tolerated and had exhibited good efficacy.Citation125 A runny nose/nasal congestion has been reported as the most common adverse events of Flumist, with mild to moderate in severity.Citation121 However The US CDC (Centre for Disease Control) Advisory Committee on Immunization Practices (ACIP) recently voted that the Flumist nasal spray live attenuated influenza vaccine (LAIV) (sic), should not be used during the 2016–2017 flu season, based on “data showing poor or relatively lower effectiveness of LAIV from 2013 through 2016.”Citation126 At the time of writing no further detail was available. It should be noted that a nasal Live Attenuated Influenza Virus (LAIV) influenza vaccine has been used for over 50 y in Russia and previously the USSR. Data published from a study using the Russian intranasal vaccine showed better herd immunity for intranasal LAIV than inactivated vaccine.Citation127 Herd immunity is a crucial impact of mas vaccination programs; it is the immunity given to the whole population, even those who have not received a vaccine, because enough of the population (the herd) have received the vaccine that the infection cannot effectively spread. However, it should be noted that the Russian LAIV is administered in 2 doses 3 weeks apart, which increases cost and has the possibility of reducing compliance.

Targeting school age children for influenza has 2 benefits, first this age group tend to have the highest rates of influenza infection. Secondly targeting children reduces infection rates in through transmission from this group, although transmission rates can vary.Citation128 In the European Union an intranasal influenza vaccine was licensed in 2011. Damm et al explored the possible effect of introducing this product in Germany and concluded that introducing the vaccine to German schoolchildren would lead to a “substantial reduction in influenza-associated disease at a reasonable cost to the German statutory health insurance system.”Citation129 Researchers looking into the same question for Thailand reached similar conclusions with provisos based on willingness to pay and contact between age groups.Citation130 This study raised the issue of effectiveness across countries where healthcare systems are either new or emerging and differences in rates and timing of seasonal outbreaks. These findings highlight the differences between high and low- to middle-income countries and demonstrate the need to carefully evaluate the target population and seasonal factors before designing or selecting a vaccine product.

A recent review describes most of the commonly encountered nasal delivery devices currently on the market.Citation72 Additionally, there is a range of nasal delivery strategies at various stages along the pre-clinical-clinical pipe-line, some of these may be suitable for vaccine delivery either in their current formats or with some adaptation. A selection of these is shown in and will be described briefly. Criticalsorb is a penetration enhancing formulation based on PLGA and PLA, developed by a spin-out from University of Nottingham, UK, currently there are no details for vaccine application. The web-site of μco™ System (Muco System) shows data for a nasal flu vaccine in a non-human primate immunogenicity study, stating that more sIgA was produced in the mucosal membrane compared to injection and nasal liquid spray. and 4-times greater sIgA than a nasal liquid spray.Citation131 Optinose is a breath-actuated device for delivering powder or liquid, a schematic of the device has been published in the literature,Citation132 as has data on the use of sumitriptan delivered via the Optinose device.Citation133,134 Kurve is a device for delivering liquid formulations “via a controlled, turbulent flow,”Citation135 the makers have published results of a pilot clinical trial detailing their intranasal insulin therapy for Alzheimer disease and amnestic mild cognitive impairment A,Citation136 while Archimedes Pharma developed a chitosan-based formulation, ChiSys®, that achieved good success in a clinical trial for a Norovirus vaccine.Citation137 Because of the proprietary and often pre-approval nature of the devices described (with the exception of Flumist/Fluenz and MAD Nasal), there is a paucity of information regarding design of some of the devices described in this section. The interested reader is referred to the relevant company websites (), which will offer more current information than is possible in this review.

Table 2. Currently marketed technology for nasal delivery.

Conclusion

Safety profiles are yet to be established in humans for many of the formulation approaches described in this review. However, the ever-increasing range of clinical trials indicates the accepted need for nasal vaccines that are easy to administer and offer improved benefits over other mucosal routes in terms of cost of formulation and need for skilled personnel to administer. The obvious benefits of directly stimulating the mucosal immune response are clear, but as yet have not been fully realized with the exception of those for influenza, which demonstrate the efficiency of this route. The recent US CDC press release will no doubt impact on the pharmaceutical industry view of riskiness of nasal formats. But with increased need to immunize large populations, potentially in swift response to pandemics such as avian, swine flu and Ebola there is a clear need to have strategies in place. The interplay between formulation or carrier and adjuvant in directing immune response should be investigated. Unfortunately, the high cost of clinical trials and issues with correlating immune responses in animal models with humans have created a bottleneck. There is a growing body of evidence to suggest that genetic material can be successfully delivered via this route, while recent studies have also demonstrated the advantages associated with combining the nasal with other routes of delivery or even combining vaccine with microbicide.Citation138 This review has focused primarily on prophylactic vaccines but there is encouraging evidence that nasal delivery will have a role to play in the design of therapeutic vaccines for e.g. cancers Alzheimer and autoimmune diseases. The role of presentation is also important when designing pre-clinical studies – instillation of drops is relatively facile even in mice, while more advanced formulations require more careful consideration than those administered via pipette. The design of ex vivo, cell culture or tissue models that provide better prediction of response in humans is extremely desirable. A “one size fits all” approach is not appropriate for vaccine design where factors relating to target population, disease type and mode of infection, will all impact on both formulation and antigen optimization.

Disclosure of potential conflicts of interest

In accordance with Taylor & Francis policy and my ethical obligation as a researcher, I am reporting that both authors are named inventors of the patent covering the technology in the VaccineTab product described herein. VK has a financial interest in the VaccineTab company that may be affected by the research reported in the enclosed paper. I have disclosed those interests fully to Taylor & Francis, and I have in place an approved plan for managing any potential conflicts arising from that involvement.

References

  • Giudice EL, Campbell JD. Needle-free vaccine delivery. Adv Drug Deliv Rev 2006; 58:68-89; PMID:16564111; http://dx.doi.org/10.1016/j.addr.2005.12.003
  • Chen D, Endres RL, Erickson CA, Weis KF, McGregor MW, Kawaoka Y, Payne LG. Epidermal immunization by a needle-free powder delivery technology: immunogenicity of influenza vaccine and protection in mice. Nat Med 2000; 6:1187-90; PMID:11017153; http://dx.doi.org/10.1038/80538
  • Holmgren J, Czerkinsky C. Mucosal immunity and vaccines. Nat Med 2005; 11:S45-S53; PMID:15812489; http://dx.doi.org/10.1038/nm1213
  • Quraishi MS, Jones NS, Mason J. The rheology of nasal mucus: a review. Clin Otolaryngol 1998; 23:403-13; PMID:9800075; http://dx.doi.org/10.1046/j.1365-2273.1998.00172.x
  • Voynow JA, Rubin BK. Mucins, mucus, and sputum. Chest 2009; 135:505-12; PMID:19201713; http://dx.doi.org/10.1378/chest.08-0412
  • Thornton DJ, Rousseau K, McGuckin MA. Structure and function of the polymeric mucins in airways mucus. Annu Rev Physiol 2008; 70:459-86; PMID:17850213; http://dx.doi.org/10.1146/annurev.physiol.70.113006.100702
  • Walker WT, Liew A, Harris A, Cole J, Lucas JS. Upper and lower airway nitric oxide levels in primary ciliary dyskinesia, cystic fibrosis and asthma. Respir Med 2013; 107:380-6; PMID:23290188; http://dx.doi.org/10.1016/j.rmed.2012.11.021
  • Thomann-Harwood LJ, Kaeuper P, Rossi N, Milona P, Herrmann B, McCullough KC. Nanogel vaccines targeting dendritic cells: Contributions of the surface decoration and vaccine cargo on cell targeting and activation. J Controlled Release 2013; 166:95-105; http://dx.doi.org/10.1016/j.jconrel.2012.11.015
  • Brandtzaeg P. Function of Mucosa-Associated Lymphoid Tissue in Antibody Formation. Immunol Invest 2010; 39:303-55; PMID:20450282; http://dx.doi.org/10.3109/08820131003680369
  • van de Pavert SA, Mebius RE. New insights into the development of lymphoid tissues. Nat Rev Immunol 2010; 10:664-74; PMID:20706277; http://dx.doi.org/10.1038/nri2832
  • Hargreaves DC, Medzhitov R. Innate Sensors of Microbial Infection. J Clin Immunol; 25:503-10; PMID:16380814; http://dx.doi.org/10.1007/s10875-005-8065-4
  • J Philpott D, E Girardin S, J Sansonetti P. Innate immune responses of epithelial cells following infection with bacterial pathogens. Curr Opin Immunol 2001; 13:410-6; PMID:11498296; http://dx.doi.org/10.1016/S0952-7915(00)00235-1
  • López-Boado YS, Wilson CL, Hooper LV, Gordon JI, Hultgren SJ, Parks WC. Bacterial Exposure Induces and Activates Matrilysin in Mucosal Epithelial Cells. The Journal of Cell Biology 2000; 148:1305-15; PMID:Can't; http://dx.doi.org/10.1083/jcb.148.6.1305
  • Kagnoff MF, Eckmann L. Epithelial cells as sensors for microbial infection. J Clin Invest 1997; 100:6-10; PMID:9202050; http://dx.doi.org/10.1172/JCI119522
  • Neutra MR, Mantis NJ, Kraehenbuhl J-P. Collaboration of epithelial cells with organized mucosal lymphoid tissues. Nat Immunol 2001; 2:1004-9; PMID:11685223; http://dx.doi.org/10.1038/ni1101-1004
  • Kiyono H, Fukuyama S. NALT- versus PEYER'S-patch-mediated mucosal immunity. Nat Rev Immunol 2004; 4:699-710; PMID:15343369
  • Corr SC, Gahan CC, Hill C. M-cells: origin, morphology and role in mucosal immunity and microbial pathogenesis. FEMS Immunol Med Microbiol 2008; 52:2-12; PMID:18081850
  • Neutra MR, Kozlowski PA. Mucosal vaccines: the promise and the challenge. Nat Rev Immunol 2006; 6:148-58; PMID:16491139
  • Sharma S, Mukkur TK, Benson HA, Chen Y. Pharmaceutical aspects of intranasal delivery of vaccines using particulate systems. J Pharm Sci 2009; 98:812-43; PMID:18661544
  • Illum L. Nanoparticulate systems for nasal delivery of drugs: A real improvement over simple systems? J Pharm Sci 2007; 96:473-83; PMID:17117404
  • Csaba N, Garcia-Fuentes M, Alonso MJ. Nanoparticles for nasal vaccination. Adv Drug Deliv Rev 2009; 61:140-57; PMID:19121350
  • Fagarasan S, Honjo T. Intestinal IgA synthesis: regulation of front-line body defences. Nat Rev Immunol 2003; 3:63-72; PMID:12511876
  • Macpherson AJ, Slack E. The functional interactions of commensal bacteria with intestinal secretory IgA. Current opinion in gastroenterology 2007; 23:673-8; PMID:17906446
  • Yel L. Selective IgA Deficiency. J Clin Immunol 2010; 30:10-6; PMID:20101521
  • Snoeck V, Peters IR, Cox E. The IgA system: a comparison of structure and function in different species. Vet Res 2006; 37:455-67; PMID:16611558
  • Borges O, Lebre F, Bento D, Borchard G, Junginger HE. Mucosal Vaccines: Recent Progress in Understanding the Natural Barriers. Pharm Res 2010; 27:211-23; PMID:19953309
  • Hutchings AB, Helander A, Silvey KJ, Chandran K, Lucas WT, Nibert ML, Neutra MR. Secretory Immunoglobulin A Antibodies against the σ1 Outer Capsid Protein of Reovirus Type 1 Lang Prevent Infection of Mouse Peyer's Patches. J Virol 2004; 78:947-57; PMID:14694126
  • Macpherson AJ, McCoy KD, Johansen FE, Brandtzaeg P. The immune geography of IgA induction and function. Mucosal Immunol 0000; 1:11-22
  • van Ginkel FW, Nguyen HH, McGhee JR. Vaccines for mucosal immunity to combat emerging infectious diseases. Emerg Infect Dis 2000; 6:123-32; PMID:10756145
  • Talsma SS, Babensee JE, Murthy N, Williams IR. Development and in vitro validation of a targeted delivery vehicle for DNA vaccines. J Controlled Release 2006; 112:271-9
  • Russell-Jones GJ. Oral vaccine delivery. J Controlled Release 2000; 65:49-54; http://dx.doi.org/10.1016/S0168-3659(99)00231-X
  • Burgdorf S, Kautz A, Böhnert V, Knolle PA, Kurts C. Distinct pathways of antigen uptake and intracellular routing in CD4 and CD8 T cell activation. Science (New York, NY) 2007; 316:612-6; http://dx.doi.org/10.1126/science.1137971
  • Brandtzaeg P. Nature and function of gastrointestinal antigen-presenting cells. Allergy 2001; 56:16-20; PMID:11298000; http://dx.doi.org/10.1034/j.1398-9995.2001.00903.x
  • Diebold SS, Cotten M, Koch N, Zenke M. MHC class II presentation of endogenously expressed antigens by transfected dendritic cells. Gene Ther 2001; 8:487-93; PMID:11313828; http://dx.doi.org/10.1038/sj.gt.3301433
  • Guy B. The perfect mix: recent progress in adjuvant research. Nat Rev Micro 2007; 5:505-17; http://dx.doi.org/10.1038/nrmicro1681
  • Sansonetti PJ, Di Santo JP. Debugging how Bacteria Manipulate the Immune Response. Immunity 2007; 26:149-61; PMID:17307704; http://dx.doi.org/10.1016/j.immuni.2007.02.004
  • Khader SA, Bell GK, Pearl JE, Fountain JJ, Rangel-Moreno J, Cilley GE, Shen F, Eaton SM, Gaffen SL, Swain SL, et al. IL-23 and IL-17 in the establishment of protective pulmonary CD4(+) T cell responses after vaccination and during Mycobacterium tuberculosis challenge. Nat Immunol 2007; 8:369-77; PMID:17351619; http://dx.doi.org/10.1038/ni1449
  • Keijzer C, Haijema BJ, Meijerhof T, Voorn P, de Haan A, Leenhouts K, van Roosmalen ML, van Eden W, Broere F. Inactivated influenza vaccine adjuvanted with Bacterium-like particles induce systemic and mucosal influenza A virus specific T-cell and B-cell responses after nasal administration in a TLR2 dependent fashion. Vaccine 2014; 32:2904-10; PMID:24598720; http://dx.doi.org/10.1016/j.vaccine.2014.02.019
  • Rathore JS, Wang Y. Protective role of Th17 cells in pulmonary infection. Vaccine 2016; 34:1504-14; PMID:26878294; http://dx.doi.org/10.1016/j.vaccine.2016.02.021
  • Zygmunt BM, Rharbaoui F, Groebe L, Guzman CA. Intranasal Immunization Promotes Th17 Immune Responses. J Immunol 2009; 183:6933-8; PMID:19890060; http://dx.doi.org/10.4049/jimmunol.0901144
  • Maroof A, Yorgensen YM, Li YF, Evans JT. Intranasal Vaccination Promotes Detrimental Th17Mediated Immunity against Influenza Infection. PLoS Path 2014; 10:e1003875
  • McKinstry KK, Strutt TM, Buck A, Curtis JD, Dibble JP, Huston G, Tighe M, Hamada H, Sell S, Dutton RW, et al. IL-10 Deficiency Unleashes an Influenza-Specific Th17 Response and Enhances Survival against High-Dose Challenge. J Immunol 2009; 182:7353-63; PMID:19494257; http://dx.doi.org/10.4049/jimmunol.0900657
  • Hamada H, Garcia-Hernandez MD, Reome JB, Misra SK, Strutt TM, McKinstry KK, Cooper AM, Swain SL, Dutton RW. Tc17, a Unique Subset of CD8 T Cells That Can Protect against Lethal Influenza Challenge. J Immunol 2009; 182:3469-81; PMID:19265125; http://dx.doi.org/10.4049/jimmunol.0801814
  • Wang R, Epstein J, Baraceros FM, Gorak EJ, Charoenvit Y, Carucci DJ, Hedstrom RC, Rahardjo N, Gay T, Hobart P, et al. Induction of CD4(+) T cell-dependent CD8(+) type 1 responses in humans by a malaria DNA vaccine. Proc Natl Acad Sci U S A 2001; 98:10817-22; PMID:11526203; http://dx.doi.org/10.1073/pnas.181123498
  • Srikiatkhachorn A, Chang W, Braciale TJ. Induction of Th-1 and Th-2 Responses by Respiratory Syncytial Virus Attachment Glycoprotein Is Epitope and Major Histocompatibility Complex Independent. J Virol 1999; 73:6590-7; PMID:10400756
  • Haglund K, Leiner I, Kerksiek K, Buonocore L, Pamer E, Rose JK. High-level primary CD8(+) T-cell response to human immunodeficiency virus type 1 gag and env generated by vaccination with recombinant vesicular stomatitis viruses. J Virol 2002; 76:2730-8; PMID:11861840; http://dx.doi.org/10.1128/JVI.76.6.2730-2738.2002
  • Boyer JD, Cohen AD, Vogt S, Schumann K, Nath B, Ahn L, Lacy K, Bagarazzi ML, Higgins TJ, Baine Y, et al. Vaccination of seronegative volunteers with a human immunodeficiency virus type 1 env/rev DNA vaccine induces antigen-specific proliferation and lymphocyte production of beta-chemokines. J Infect Dis 2000; 181:476-83; PMID:10669329; http://dx.doi.org/10.1086/315229
  • D'Souza S, Romano M, Korf J, Wang X-M, Adnet P-Y, Huygen K. Partial Reconstitution of the CD4(+)-T-Cell Compartment in CD4 Gene Knockout Mice Restores Responses to Tuberculosis DNA Vaccines. Infect Immun 2006; 74:2751-9; PMID:16622212; http://dx.doi.org/10.1128/IAI.74.5.2751-2759.2006
  • Walsh KP, Mills KHG. Dendritic cells and other innate determinants of T helper cell polarisation. Trends Immunol; 34:521-30; PMID:23973621; http://dx.doi.org/10.1016/j.it.2013.07.006
  • McKenzie Andrew NJ, Spits H, Eberl G. Innate Lymphoid Cells in Inflammation and Immunity. Immunity 2014; 41:366-74; PMID:25238094; http://dx.doi.org/10.1016/j.immuni.2014.09.006
  • Artis D, Spits H. The biology of innate lymphoid cells. Nature 2015; 517:293-301; PMID:25592534; http://dx.doi.org/10.1038/nature14189
  • von Burg N, Turchinovich G, Finke D. Maintenance of Immune Homeostasis through ILC/T Cell Interactions. Front Immunol 2015; 6:416; PMID:26322047; http://dx.doi.org/10.3389/fimmu.2015.00416
  • Luci C, Bekri S, Bihl F, Pini J, Bourdely P, Nouhen K, Malgogne A, Walzer T, Braud VM, Anjuère F. NKp46+ Innate Lymphoid Cells Dampen Vaginal CD8 T Cell Responses following Local Immunization with a Cholera Toxin-Based Vaccine. PLoS ONE 2015; 10:e0143224; PMID:26630176; http://dx.doi.org/10.1371/journal.pone.0143224
  • Zingoni A, Sornasse T, Cocks BG, Tanaka Y, Santoni A, Lanier LL. Cross-talk between activated human NK cells and CD4(+) T cells via OX40-OX40 ligand interactions. J Immunol 2004; 173:3716-24; PMID:15356117; http://dx.doi.org/10.4049/jimmunol.173.6.3716
  • Mirchandani AS, Besnard AG, Yip E, Scott C, Bain CC, Cerovic V, Salmond RJ, Liew FY. Type 2 Innate Lymphoid Cells Drive CD4(+) Th2 Cell Responses. J Immunol 2014; 192:2442-8; PMID:24470502; http://dx.doi.org/10.4049/jimmunol.1300974
  • Sakaguchi S, Wing K, Miyara M. Regulatory T cells - a brief history and perspective. Eur J Immunol 2007; 37:S116-S23; PMID:17972355; http://dx.doi.org/10.1002/eji.200737593
  • Ramirez K, Wahid R, Richardson C, Bargatze RF, El-Kamary SS, Sztein MB, Pasetti MF. Intranasal vaccination with an adjuvanted Norwalk virus-like particle vaccine elicits antigen-specific B memory responses in human adult volunteers. Clin Immunol 2012; 144:98-108; PMID:22710446; http://dx.doi.org/10.1016/j.clim.2012.05.006
  • Sealy R, Jones BG, Surman SL, Hurwitz JL. Robust IgA and IgG-producing antibody forming cells in the diffuse NALT and lungs of Sendai virus-vaccinated cotton rats associate with rapid protection against human parainfluenza virus-type 1. Vaccine 2010; 28:6749-56; PMID:20682364; http://dx.doi.org/10.1016/j.vaccine.2010.07.068
  • Fujkuyama Y, Tokuhara D, Kataoka K, Gilbert RS, McGhee JR, Yuki Y, Kiyono H, Fujihashi K. Novel vaccine development strategies for inducing mucosal immunity. Expert Rev Vaccines 2012; 11:367-79; PMID:22380827; http://dx.doi.org/10.1586/erv.11.196
  • Simon JK, Ramirez K, Cuberos L, Campbell JD, Viret JF, Muñoz A, Lagos R, Levine MM, Pasetti MF. Mucosal IgA Responses in Healthy Adult Volunteers following Intranasal Spray Delivery of a Live Attenuated Measles Vaccine. Clin Vaccine Immunol 2011; 18:355-61; PMID:21228137; http://dx.doi.org/10.1128/CVI.00354-10
  • Lijek RS, Luque SL, Liu Q, Parker D, Bae T, Weiser JN. Protection from the acquisition of Staphylococcus aureus nasal carriage by cross-reactive antibody to a pneumococcal dehydrogenase. Proc Natl Acad Sci U S A 2012; 109:13823-8; PMID:22869727; http://dx.doi.org/10.1073/pnas.1208075109
  • Jang YH, Byun YH, Lee YJ, Lee YH, Lee K-H, Seong BL. Cold-adapted pandemic 2009 H1N1 influenza virus live vaccine elicits cross-reactive immune responses against seasonal and H5 influenza a viruses. J Virol 2012; 86:5953-8; PMID:22438541; http://dx.doi.org/10.1128/JVI.07149-11
  • Motohashi S, Okamoto Y, Yoshino I, Nakayama T. Anti-tumor immune responses induced by iNKT cell-based immunotherapy for lung cancer and head and neck cancer. Clin Immunol 2011; 140:167-76; PMID:21349771; http://dx.doi.org/10.1016/j.clim.2011.01.009
  • Xiao C, Davis FJ, Chauhan BC, Viola KL, Lacor PN, Velasco PT, Klein WL, Chauhan NB. Brain transit and ameliorative effects of intranasally delivered anti-amyloid-beta oligomer antibody in 5XFAD mice. J Alzheimers Dis 2013; 35:777-88; PMID:23542865
  • Keijzer C, van der Zee R, van Eden W, Broere F. Treg inducing adjuvants for therapeutic vaccination against chronic inflammatory diseases. Front Immunol 2013; 4:245; PMID:23970886; http://dx.doi.org/10.3389/fimmu.2013.00245
  • Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3+ regulatory T cells in the human immune system. Nat Rev Immunol 2010; 10:490-500; PMID:20559327; http://dx.doi.org/10.1038/nri2785
  • Weiner HL, da Cunha AP, Quintana F, Wu H. Oral tolerance. Immunol Rev 2011; 241:241-59; PMID:21488901; http://dx.doi.org/10.1111/j.1600-065X.2011.01017.x
  • Keijzer C, Spiering R, Silva AL, van Eden W, Jiskoot W, Vervelde L, Broere F. PLGA nanoparticles enhance the expression of retinaldehyde dehydrogenase enzymes in dendritic cells and induce FoxP3+ T-cells in vitro. J Controlled Release 2013; 168:35-40; http://dx.doi.org/10.1016/j.jconrel.2013.02.027
  • Li H, Ding Y, Yi G, Zeng Q, Yang W. Establishment of nasal tolerance to heat shock protein-60 alleviates atherosclerosis by inducing TGF-β-dependent regulatory T cells. J Huazhong Univ Sci Technol [Medical Sciences] 2012; 32:24-30; PMID:22282240; http://dx.doi.org/10.1007/s11596-012-0004-z
  • Klingenberg R, Lebens M, Hermansson A, Fredrikson GN, Strodthoff D, Rudling M, Ketelhuth DFJ, Gerdes N, Holmgren J, Nilsson J, et al. Intranasal Immunization With an Apolipoprotein B-100 Fusion Protein Induces Antigen-Specific Regulatory T Cells and Reduces Atherosclerosis. Arterioscler Thromb Vasc Biol 2010; 30:946-U148; http://dx.doi.org/10.1161/ATVBAHA.109.202671
  • Broere F, Wieten L, Koerkamp EIK, van Roon JAG, Guichelaar T, Lafeber F, van Eden W. Oral or nasal antigen induces regulatory T cells that suppress arthritis and proliferation of arthritogenic T cells in joint draining lymph nodes. J Immunol 2008; 181:899-906; PMID:18606641; http://dx.doi.org/10.4049/jimmunol.181.2.899
  • Djupesland PG. Nasal drug delivery devices: characteristics and performance in a clinical perspective—a review. Drug Delivery and Translational Research 2013; 3:42-62; PMID:23316447; http://dx.doi.org/10.1007/s13346-012-0108-9
  • Wang SH, Kirwan SM, Abraham SN, Staats HF, Hickey AJ. Stable Dry Powder Formulation for Nasal Delivery of Anthrax Vaccine. J Pharm Sci 2012; 101:31-47; PMID:21905034; http://dx.doi.org/10.1002/jps.22742
  • McInnes FJ, Thapa P, Baillie AJ, Welling PG, Watson DG, Gibson I, Nolan A, Stevens HNE. In vivo evaluation of nicotine lyophilised nasal insert in sheep. Int J Pharm 2005; 304:72-82; PMID:16183221; http://dx.doi.org/10.1016/j.ijpharm.2005.07.025
  • Pattani A, McKay PF, Curran RM, McCaffrey J, Gupta PN, Lowry D, Kett VL, Shattock RJ, McCarthy HO, Malcolm RK. Molecular investigations into vaginal immunization with HIV gp41 antigenic construct H4A in a quick release solid dosage form. Vaccine 2012; 30:2778-85; PMID:22361120; http://dx.doi.org/10.1016/j.vaccine.2012.02.004
  • Bachmann MF, Jennings GT. Vaccine delivery: a matter of size, geometry, kinetics and molecular patterns. Nat Rev Immunol 2010; 10:787-96; PMID:20948547; http://dx.doi.org/10.1038/nri2868
  • Peek LJ, Middaugh CR, Berkland C. Nanotechnology in vaccine delivery. Adv Drug Deliv Rev 2008; 60:915-28; PMID:18325628; http://dx.doi.org/10.1016/j.addr.2007.05.017
  • Koping-Hoggard M, Sanchez A, Alonso MJ. Nanoparticles as carriers for nasal vaccine delivery. Exp Rev Vaccines 2005; 4:185-96; PMID:15889992; http://dx.doi.org/10.1586/14760584.4.2.185
  • Slütter B, Bal S, Keijzer C, Mallants R, Hagenaars N, Que I, Kaijzel E, van Eden W, Augustijns P, Löwik C, et al. Nasal vaccination with N-trimethyl chitosan and PLGA based nanoparticles: Nanoparticle characteristics determine quality and strength of the antibody response in mice against the encapsulated antigen. Vaccine 2010; 28:6282-91; PMID:20638455; http://dx.doi.org/10.1016/j.vaccine.2010.06.121
  • Jain AK, Goyal AK, Gupta PN, Khatri K, Mishra N, Mehta A, Mangal S, Vyas SP. Synthesis, characterization and evaluation of novel triblock copolymer based nanoparticles for vaccine delivery against hepatitis B. J Controlled Release 2009; 136:161-9; http://dx.doi.org/10.1016/j.jconrel.2009.02.010
  • Jain A, Massey AS, Yusuf H, McDonald DM, McCarthy H, Kett V. Development of polymeric–cationic peptide composite nanoparticles, a nanoparticle-in- nanoparticle system for controlled gene delivery. Int J Nanomed 2015; 7183-96.
  • Nochi T, Yuki Y, Takahashi H, Sawada S-I, Mejima M, Kohda T, Harada N, Kong IG, Sato A, Kataoka N, et al. Nanogel antigenic protein-delivery system for adjuvant-free intranasal vaccines. Nat Mater 2010; 9:572-8; PMID:20562880; http://dx.doi.org/10.1038/nmat2784
  • Jesus S, Soares E, Costa J, Borchard G, Borges O. Immune response elicited by an intranasally delivered HBsAg low-dose adsorbed to poly-epsilon-caprolactone based nanoparticles. Int J Pharm 2016; 504:59-69; PMID:26976502; http://dx.doi.org/10.1016/j.ijpharm.2016.03.013
  • de Jonge MI, Hamstra HJ, Jiskoot W, Roholl P, Williams NA, Dankert J, Alphen Lv, van der Ley P. Intranasal immunisation of mice with liposomes containing recombinant meningococcal OpaB and OpaJ proteins. Vaccine 2004; 22:4021-8; PMID:15364452; http://dx.doi.org/10.1016/j.vaccine.2004.03.047
  • Wang D, Christopher ME, Nagata LP, Zabielski MA, Li H, Wong JP, Samuel J. Intranasal immunization with liposome-encapsulated plasmid DNA encoding influenza virus hemagglutinin elicits mucosal, cellular and humoral immune responses. J Clin Virol 2004; 31, Supplement 1:99-106; http://dx.doi.org/10.1016/j.jcv.2004.09.013
  • Khatri K, Goya AK, Gupta PN, Mishra N, Mehta A, Vyas SP. Surface modified liposomes for nasal delivery of DNA vaccine. Vaccine 2008; 26:2225-33; PMID:18396362; http://dx.doi.org/10.1016/j.vaccine.2008.02.058
  • Amin M, Jaafari MR, Tafaghodi M. Impact of chitosan coating of anionic liposomes on clearance rate, mucosal and systemic immune responses following nasal administration in rabbits. Colloids and Surfaces B-Biointerfaces 2009; 74:225-9; http://dx.doi.org/10.1016/j.colsurfb.2009.07.024
  • Heurtault B, Frisch B, Pons F. Liposomes as delivery systems for nasal vaccination: strategies and outcomes. Exp Opin Drug Delivery 2010; 7:829-44; PMID:20459361; http://dx.doi.org/10.1517/17425247.2010.488687
  • Wang HW, Jiang PL, Lin SF, Lin HJ, Ou KL, Deng WP, Lee LW, Huang YY, Liang PH, Liu DZ. Application of galactose-modified liposomes as a potent antigen presenting cell targeted carrier for intranasal immunization. Acta Biomater 2013; 9:5681-8; PMID:23159567; http://dx.doi.org/10.1016/j.actbio.2012.11.007
  • Fan Y, Sahdev P, Ochyl LJ, J. Akerberg J, Moon JJ. Cationic liposome–hyaluronic acid hybrid nanoparticles for intranasal vaccination with subunit antigens. J Controlled Release 2015; 208:121-9; http://dx.doi.org/10.1016/j.jconrel.2015.04.010
  • Chen KH, Di Sabatino M, Albertini B, Passerini N, Kett VL. The effect of polymer coatings on physicochemical properties of spray-dried liposomes for nasal delivery of BSA. Eur J Pharm Sci 2013; 50:312-22; PMID:23876823; http://dx.doi.org/10.1016/j.ejps.2013.07.006
  • Glück U, Gebbers J-O, Glück R. Phase 1 Evaluation of Intranasal Virosomal Influenza Vaccine with and without Escherichia coli Heat-Labile Toxin in Adult Volunteers. J Virol 1999; 73:7780-6; PMID:10438868
  • Gluck R. Pre-clinical and clinical investigation of the safety of a novel adjuvant for intranasal immunization. Vaccine 2001; 20, Supplement 1:S42-S4.
  • Cusi MG, Zurbriggen R, Valassina M, Bianchi S, Durrer P, Valensin PE, Donati M, Gluck R. Intranasal immunization with mumps virus DNA vaccine delivered by influenza virosomes elicits mucosal and systemic immunity. Virology 2000; 277:111-8; PMID:11062041; http://dx.doi.org/10.1006/viro.2000.0605
  • Durrer P, Gluck U, Spyr C, Lang AB, Zurbriggen R, Herzog C, Gluck R. Mucosal antibody response induced with a nasal virosome-based influenza vaccine. Vaccine 2003; 21:4328-34; PMID:14505915; http://dx.doi.org/10.1016/S0264-410X(03)00457-2
  • Salleras L, Dominguez A, Pumarola T, Prat A, Marcos MA, Garrido P, Artigas R, Bau A, Brotons J, Bruna X, et al. Effectiveness of virosomal subunit influenza vaccine in preventing influenza-related illnesses and its social and economic consequences in children aged 3-14 years: A prospective cohort study. Vaccine 2006; 24:6638-42; PMID:16842892; http://dx.doi.org/10.1016/j.vaccine.2006.05.034
  • Lambkin R, Oxford JS, Bossuyt S, Mann A, Metcalfe IC, Herzog C, Viret JF, Gluck R. Strong local and systemic protective immunity induced in the ferret model by an intranasal virosome-formulated influenza subunit vaccine. Vaccine 2004; 22:4390-6; PMID:15474733; http://dx.doi.org/10.1016/j.vaccine.2003.10.054
  • Hossain MJ, Bourgeois M, Quan F-S, Lipatov AS, Song J-M, Chen L-M, Compans RW, York I, Kang S-M, Donis RO. Virus-Like Particle Vaccine Containing Hemagglutinin Confers Protection against 2009 H1N1 Pandemic Influenza. Clin Vaccine Immunol 2011; 18:2010-7; PMID:22030367; http://dx.doi.org/10.1128/CVI.05206-11
  • Herbst-Kralovetz M, Mason HS, Chen Q. Norwalk virus-like particles as vaccines. Exp Rev Vaccines 2010; 9:299-307; PMID:20218858; http://dx.doi.org/10.1586/erv.09.163
  • Hagenaars N, Mastrobattista E, Glansbeek H, Heldens J, van den Bosch H, Schijns V, Betbeder D, Vromans H, Jiskoot W. Head-to-head comparison of four nonadjuvanted inactivated cell culture-derived influenza vaccines: Effect of composition, spatial organization and immunization route on the immunogenicity in a murine challenge model. Vaccine 2008; 26:6555-63; PMID:18848856; http://dx.doi.org/10.1016/j.vaccine.2008.09.057
  • de Jonge J, Leenhouts JM, Holtrop M, Schoen P, Scherrer P, Cullis PR, Wilschut J, Huckriede A. Cellular gene transfer mediated by influenza virosomes with encapsulated plasmid DNA. Biochem J 2007; 405:41-9; PMID:17355227; http://dx.doi.org/10.1042/BJ20061756
  • Cusi MG. Applications of influenza virosomes as a delivery system. Human Vaccines 2006; 2:1-7; PMID:17012895; http://dx.doi.org/10.4161/hv.2.1.2494
  • Lai SK, Wang Y-Y, Hanes J. Mucus-penetrating nanoparticles for drug and gene delivery to mucosal tissues. Adv Drug Del Rev 2009; 61:158-71; http://dx.doi.org/10.1016/j.addr.2008.11.002
  • Singh M, Briones M, O'Hagan DT. A novel bioadhesive intranasal delivery system for inactivated influenza vaccines. J Controlled Release 2001; 70:267-76; http://dx.doi.org/10.1016/S0168-3659(00)00330-8
  • Stephenson I, Nicholson KG, Hoschler K, Zambon MC, Hancock K, DeVos J, Katz JM, Praus M, Banzhoff A. Antigenically distinct MF59-adjuvanted vaccine to boost immunity to H5N1. N Engl J Med 2008; 359:1631-3; PMID:18843132; http://dx.doi.org/10.1056/NEJMc0805274
  • Schultze V, D'Agosto V, Wack A, Novicki D, Zorn J, Hennig R. Safety of MF59™ adjuvant. Vaccine 2008; 26:3209-22; PMID:18462843; http://dx.doi.org/10.1016/j.vaccine.2008.03.093
  • McAleer JP, Vella AT. Educating CD4 T cells with vaccine adjuvants: lessons from lipopolysaccharide. Trends Immunol 2010; 31:429-35; PMID:20880743; http://dx.doi.org/10.1016/j.it.2010.08.005
  • Zonneveld-Huijssoon E, van Wijk F, Roord S, Delemarre E, Meerding J, de Jager W, Klein M, Raz E, Albani S, Kuis W, et al. TLR9 agonist CpG enhances protective nasal HSP60 peptide vaccine efficacy in experimental autoimmune arthritis. Ann Rheum Dis 2012; 71:1706-15; PMID:22562976; http://dx.doi.org/10.1136/annrheumdis-2011-201131
  • Velasquez LS, Hjelm BE, Arntzen CJ, Herbst-Kralovetz MM. An intranasally delivered Toll-like receptor 7 agonist elicits robust systemic and mucosal responses to Norwalk virus-like particles. Clin Vaccine Immunol 2010; 17:1850-8; PMID:20962211; http://dx.doi.org/10.1128/CVI.00230-10
  • Sui Z, Chen Q, Wu R, Zhang H, Zheng M, Wang H, Chen Z. Cross-protection against influenza virus infection by intranasal administration of M2-based vaccine with chitosan as an adjuvant. Arch Virol 2010; 155:535-44; PMID:20195654; http://dx.doi.org/10.1007/s00705-010-0621-4
  • Noda K, Kodama S, Umemoto S, Abe N, Hirano T, Suzuki M. Nasal vaccination with P6 outer membrane protein and alpha-galactosylceramide induces nontypeable Haemophilus influenzae-specific protective immunity associated with NKT cell activation and dendritic cell expansion in nasopharynx. Vaccine 2010; 28:5068-74; PMID:20478344; http://dx.doi.org/10.1016/j.vaccine.2010.05.005
  • Miyata T, Harakuni T, Tsuboi T, Sattabongkot J, Kohama H, Tachibana M, Matsuzaki G, Torii M, Arakawa T. Plasmodium vivax ookinete surface protein Pvs25 linked to cholera toxin B subunit induces potent transmission-blocking immunity by intranasal as well as subcutaneous immunization. Infect Immun 2010; 78:3773-82; PMID:20584978; http://dx.doi.org/10.1128/IAI.00306-10
  • Freytag LC, Clements JD. Mucosal adjuvants. Vaccine 2005; 23:1804-13; PMID:15734046; http://dx.doi.org/10.1016/j.vaccine.2004.11.010
  • Mutsch M, Zhou W, Rhodes P, Bopp M, Chen RT, Linder T, Spyr C, Steffen R. Use of the Inactivated Intranasal Influenza Vaccine and the Risk of Bell's Palsy in Switzerland. N Engl J Med 2004; 350:896-903; PMID:14985487; http://dx.doi.org/10.1056/NEJMoa030595
  • van Ginkel FW, Jackson RJ, Yuki Y, McGhee JR. Cutting edge: the mucosal adjuvant cholera toxin redirects vaccine proteins into olfactory tissues. J Immunol 2000; 165:4778-82; PMID:11045998; http://dx.doi.org/10.4049/jimmunol.165.9.4778
  • Gluck R, Mischler R, Durrer P, Furer E, Lang AB, Herzog C, Cryz SJ, Jr. Safety and immunogenicity of intranasally administered inactivated trivalent virosome-formulated influenza vaccine containing Escherichia coli heat-labile toxin as a mucosal adjuvant. J Infect Dis 2000; 181:1129-32; PMID:10720540; http://dx.doi.org/10.1086/315337
  • Kanerva M, Mannonen L, Pliparinen H, Peltomaa M, Vaheri A, Pitkaranta A. Search for herpesviruses in cerebrospinal fluid of facial palsy patients by PCR. Acta Otolaryngol (Stockh) 2007; 127:775-9; http://dx.doi.org/10.1080/00016480601011444
  • Kiura K, Kataoka H, Yasuda M, Inoue N, Shibata K. The diacylated lipopeptide FSL-1 induces TLR2-mediated Th2 responses. FEMS Immunol Med Microbiol 2006; 48:44-55; PMID:16965351; http://dx.doi.org/10.1111/j.1574-695X.2006.00119.x
  • Nyirenda MH, Sanvito L, Darlington PJ, O'Brien K, Zhang GX, Constantinescu CS, Bar-Or A, Gran B. TLR2 Stimulation Drives Human Naive and Effector Regulatory T Cells into a Th17-Like Phenotype with Reduced Suppressive Function. J Immunol 2011; 187:2278-90; PMID:21775683; http://dx.doi.org/10.4049/jimmunol.1003715
  • Keijzer C, Slutter B, van der Zee R, Jiskoot W, van Eden W, Broere F. PLGA, PLGA-TMC and TMC-TPP Nanoparticles Differentially Modulate the Outcome of Nasal Vaccination by Inducing Tolerance or Enhancing Humoral Immunity. Plos One 2011; 6:10
  • Carter NJ, Curran MP. Live attenuated influenza vaccine (FluMist(R); Fluenz): a review of its use in the prevention of seasonal influenza in children and adults. Drugs 2011; 71:1591-622; PMID:21861544; http://dx.doi.org/10.2165/11206860-000000000-00000
  • Watts PJ, Smith A. Re-formulating drugs and vaccines for intranasal delivery: maximum benefits for minimum risks? Drug Discov Today 2011; 16:4-7; PMID:21074635; http://dx.doi.org/10.1016/j.drudis.2010.11.001
  • Kulkarni PS, Raut SK, Dhere RM. A post-marketing surveillance study of a human live-virus pandemic influenza A (H1N1) vaccine (Nasovac ((R)) ) in India. Human vaccines & immunotherapeutics 2013; 9:122-4; PMID:23442586; http://dx.doi.org/10.4161/hv.22317
  • Dhere R, Yeolekar L, Kulkarni P, Menon R, Vaidya V, Ganguly M, Tyagi P, Barde P, Jadhav S. A pandemic influenza vaccine in India: from strain to sale within 12 months. Vaccine 2011; 29 Suppl 1:A16-21; http://dx.doi.org/10.1016/j.vaccine.2011.04.119
  • Lycke N. Recent progress in mucosal vaccine development: potential and limitations. Nature Reviews Immunology 2012; 12:592-605; PMID:22828912; http://dx.doi.org/10.1038/nri3251
  • Control CfD. ACIP votes down use of LAIV for 2016–2017 flu season CDC Press Release 2016.
  • Rudenko LG, Slepushkin AN, Monto AS, Kendal AP, Grigorieva EP, Burtseva EP, Rekstin AR, Beljaev AL, Bragina VE, Cox N, et al. Efficacy Of Live Attenuated And Inactivated Influenza Vaccines In Schoolchildren And Their Unvaccinated Contacts In Novgorod, Russia. J Infect Dis 1993; 168:881-7; PMID:8376833; http://dx.doi.org/10.1093/infdis/168.4.881
  • Elveback LR, Fox JP, Ackerman E, Langworthy A, Boyd M, Gatewood L. An Influenza Simulation Model For Immunization Studies. Am J Epidemiol 1976; 103:152-65; PMID:814808
  • Damm O, Eichner M, Rose MA, Knuf M, Wutzler P, Liese JG, Kruger H, Greiner W. Public health impact and cost-effectiveness of intranasal live attenuated influenza vaccination of children in Germany. Eur J Health Econ 2015; 16:471-88; PMID:24859492; http://dx.doi.org/10.1007/s10198-014-0586-4
  • Meeyai A, Praditsitthikorn N, Kotirum S, Kulpeng W, Putthasri W, Cooper BS, Teerawattananon Y. Seasonal Influenza Vaccination for Children in Thailand: A Cost-Effectiveness Analysis. Plos Medicine 2015; 12:e1001829; PMID:26011712; http://dx.doi.org/10.1371/journal.pmed.1001829
  • SNBL. Nasal Flu vaccine using μco™ System. 2015.
  • Tepper SJ, Cady RK, Silberstein S, Messina J, Mahmoud RA, Djupesland PG, Shin P, Siffert J. AVP-825 Breath-Powered Intranasal Delivery System Containing 22 mg Sumatriptan Powder vs 100 mg Oral Sumatriptan in the Acute Treatment of Migraines (The COMPASS Study): A Comparative Randomized Clinical Trial Across Multiple Attacks. Headache 2015; 55:621-35; PMID:25941016; http://dx.doi.org/10.1111/head.12583
  • Cady R. A novel intranasal breath-powered delivery system for sumatriptan: a review of technology and clinical application of the investigational product AVP-825 in the treatment of migraine. Expert Opinion on Drug Delivery 2015; 12:1565-77; PMID:26119828; http://dx.doi.org/10.1517/17425247.2015.1060959
  • Tepper SJ. Clinical Implications for Breath-Powered Powder Sumatriptan Intranasal Treatment. Headache 2013; 53:1341-9; PMID:23809006; http://dx.doi.org/10.1111/head.12166
  • Kurve. KUrve ViaNase Elecetronic Nebulizer. Kurve Technology Inc., www.kurvetech.com. [accessed on August 2016]
  • Craft S, Baker LD, Montine TJ, Minoshima S, Watson GS, Claxton A, Arbuckle M, Callaghan M, Tsai E, Plymate SR, et al. Intranasal Insulin Therapy for Alzheimer Disease and Amnestic Mild Cognitive Impairment A Pilot Clinical Trial. Arch Neurol 2012; 69:29-38; PMID:21911655; http://dx.doi.org/10.1001/archneurol.2011.233
  • Smith A, Perelman M, Hinchcliffe M. Chitosan: A promising safe and immune-enhancing adjuvant for intranasal vaccines. Human vaccines & immunotherapeutics 2014; 10:797-807; PMID:24346613; http://dx.doi.org/10.4161/hv.27449
  • Le Grand R, Dereuddre-Bosquet N, Dispinseri S, Gosse L, Desjardins D, Shen XY, Tolazzi M, Ochsenbauer C, Saidi H, Tomaras G, et al. Superior Efficacy of a Human Immunodeficiency Virus Vaccine Combined with Antiretroviral Prevention in Simian-Human Immunodeficiency Virus-Challenged Nonhuman Primates. J Virol 2016; 90:5315-28; PMID:27009957; http://dx.doi.org/10.1128/JVI.00230-16
  • Lebre F, Borchard G, Faneca H, Pedroso de Lima MC, Borges O. Intranasal Administration of Novel Chitosan Nanoparticle/DNA Complexes Induces Antibody Response to Hepatitis B Surface Antigen in Mice. Mol Pharm 2016; 13:472-82; PMID:26651533; http://dx.doi.org/10.1021/acs.molpharmaceut.5b00707
  • Pawar D, Mangal S, Goswami R, Jaganathan KS. Development and characterization of surface modified PLGA nanoparticles for nasal vaccine delivery: Effect of mucoadhesive coating on antigen uptake and immune adjuvant activity. Eur J Pharm Biopharm 2013; 85:550-9; PMID:23831265; http://dx.doi.org/10.1016/j.ejpb.2013.06.017
  • Khatri K, Goyal AK, Gupta PN, Mishra N, Mehta A, Vyas SP. Surface modified liposomes for nasal delivery of DNA vaccine. Vaccine 2008; 26:2225-33; PMID:18396362; http://dx.doi.org/10.1016/j.vaccine.2008.02.058
  • Pan L, Zhang Z, Lv J, Zhou P, Hu W, Fang Y, Chen H, Liu X, Shao J, Zhao F, et al. Induction of mucosal immune responses and protection of cattle against direct-contact challenge by intranasal delivery with foot-and-mouth disease virus antigen mediated by nanoparticles. Int J Nanomedicine 2014; 9:5603-18; PMID:25506214; http://dx.doi.org/10.2147/IJN.S72318
  • Grassin-Delyle S, Buenestado A, Naline E, Faisy C, Blouquit-Laye S, Couderc LJ, Le Guen M, Fischler M, Devillier P. Intranasal drug delivery: an efficient and non-invasive route for systemic administration: focus on opioids. Pharmacol Ther 2012; 134:366-79; PMID:22465159; http://dx.doi.org/10.1016/j.pharmthera.2012.03.003

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.