Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

Review

Therapeutic
Delivery
For reprint orders, please contact: reprints@futuremedicine.com

Intranasal delivery of nanoparticle-based


vaccines

Most pathogens gain access to the human body and initiate systemic infections through Nirmal Marasini1,2, Mariusz
mucosal sites. A large number of currently marketed licensed vaccines are parenterally Skwarczynski**,1 & Istvan
administered; they generate strong systemic immunity but not mucosal immunity. Toth*,1,3,4
1
School of Chemistry & Molecular
Nasal vaccination is an appealing strategy for the induction of mucosal-specific
Biosciences, The University of
immunity; however, its development is mostly challenged by several factors, such as Queensland, St Lucia, QLD, 4072,
inefficient antigen uptake, its rapid mucociliary clearance, size-restricted permeation Australia
across epithelial barriers and absence of safe human mucosal adjuvants. Therefore, 2
School of Biomedical Sciences, The
a safer mucosal-adjuvanting strategy or efficient mucosal delivery platform is much University of Queensland, St Lucia, QLD,
4072, Australia
warranted. This review summarizes challenges and the rationale for nasal vaccine 3
Institute for Molecular Biosciences, The
development with a special focus on the use of nanoparticles based on polymers and University of Queensland, St Lucia, QLD,
lipids for mucosal vaccine delivery. 4072, Australia
4
School of Pharmacy, The University of
First draft submitted: 28 September 2016; Accepted for publication: 18 November Queensland, Woolloongabba, QLD 4102,
Australia
2016; Published online: 31 January 2017
**Author for correspondence:
m.skwarczynski@ uq.edu.au
Keywords: lipids • mucosal immunity • nanoparticles • nasal vaccines • polymers *Author for correspondence:
Tel.: +61 7 3346 9892
Fax: +61 7 3365 4273
Since the introduction of the first vaccine mucosal routes such as oral, nasal, pulmo- i.toth@ uq.edu.au
in the 19th century, mass vaccination pro- nary, ocular, rectal, vaginal, etc. Among
grams have saved millions of lives. Although mucosal routes, the simplest, convenient and
the incidence of vaccine-preventable diseases preferred routes for vaccine administration
is less common in developed countries, as are the oral and intranasal. However, the
many as 15 million people are still dying oral route is the most challenging for vaccine
yearly due to infectious diseases [1] . Most administration due to antigen degradation by
pathogens gain access to the human body proteolytic enzymes and acidic pH in the GI
through mucosal sites and initiate systemic tract [2] . In contrast, the intranasal admin-
infections. Currently, marketed licensed vac- istration route offers fairly low enzymatic
cines are parenterally administered but these degradation of antigens compared with other
only generate strong systemic immunity and mucosal routes, houses a large number of
almost no mucosal immunity. The lack of immune cells, and mucosal epithelial mem-
mucosal vaccines against existing and emerg- branes are relatively easily permeable [3] . In
ing pathogens has created the need for the addition, nasal vaccines possess several advan-
development of a novel and effective muco- tages: for example, they are needle-free and
sal vaccination strategy, which can induce noninvasive in nature, offer the possibility of
both antigen-specific systemic and mucosal mass immunization and require less trained
immunity. Unfortunately, the development manpower for immunization [4] . The differ-
is partly limited due to the absence of safe ences between the intranasal and oral routes
human-approved mucosal adjuvants. of vaccine administration are summarized in
Mucosal immune responses are typically Table 1. Despite several attractive features of
part of
induced when antigens are delivered through intranasal vaccination, only a few intranasal

10.4155/tde-2016-0068 © 2017 Future Science Ltd Ther. Deliv. (2017) 8(3), 151–167 ISSN 2041-5990 151
Review Marasini, Skwarczynski & Toth

Table 1. Differences between intranasal and oral route of vaccinations.


Intranasal Oral
Nasal mucosal surface Highly vascularized Less vascularized than nasal mucosal
surface
Absorption Rapid absorption of antigens to reach Inconsistent absorption profile/longer time
systemic circulation to reach systemic circulation
First-pass metabolism (presystemic Avoids first-pass effect Extensive first-pass metabolism
metabolism)
Influence of enzymes and pH Low influence of proteolytic enzymes and High influence of enzymes and varying pH
pH across GI tract
Required dose Relatively lower than oral/less frequent High-dose/more frequent immunization
immunizations
Immune responses Rapid local immune responses Slower or unresponsive to soluble antigens
Protective immune responses in lungs, Protective immune responses at GI tract,
upper respiratory tract and genital tract upper respiratory tract (tonsils), salivary
mucosae and mammary glands
Poor intestinal immune response

vaccines (Flumist®, Fluenz® and Nasovac®) are avail- Nasal immune system
able for human use [5–7] . Nasal vaccine development is Nasal mucosa is an important part of the immune sys-
mostly challenged by the inefficient uptake of soluble tem since it is a first site of contact for inhaled patho-
antigens by nasal immune cells, rapid mucociliary gens or vaccine-associated antigens. The nasal mucosa
clearance or short resident time of antigens in the nasal consists of epithelial cells, which play a crucial role in
tract, size-restricted permeation across epithelial bar- maintaining immune homeostasis by providing a con-
riers, absence of safe human mucosal adjuvants, poor tinuous physical barrier against exogenous substances.
antigen stability and restricted delivery volume. The predominant immunoglobulin found in nasal
Vaccines usually contain killed or live-attenuated mucosa is secretory IgA (sIgA). High levels of local
organisms or their subunit fragments: for example, car- mucosal antibodies are usually produced at the site of
bohydrate, proteins and peptides. Historically, whole antigen administration and comparatively low IgA is
pathogen-based vaccines were the most popular, mostly produced at distant mucosal sites.
because of the simplicity and relatively low cost of their The primary site for the induction of nasal immu-
production. The success of such vaccines was also related nity against administered antigens is the nasal-associ-
to their capability of presentation of multiple antigens, ated lymphoid tissues (NALT). In rodents, NALT con-
along with natural adjuvants, to the immune system that tain aggregates of organized lymphoid follicles located
ultimately led to a potent antibody and cell-mediated at the nasopharyngeal cavity [9] . This structure is func-
immunity. However, the use of whole pathogen-based tionally similar to human’s Waldeyer’s ring (tonsils
vaccines comes with some demerits such as chances and adenoids) and primarily responsible for producing
of autoimmune or strong allergic responses, produc- local immune reactions upon intranasal vaccination in
tion difficulties (not all pathogens can be cultured), the rodents [10] . Therefore, the rodent-based models are
biological contamination, etc. [8] . Therefore, alterna- well suited for early vaccine development. NALT com-
tive methods using subunit antigens such as toxins or prises B cells, T cells, macrophages and dendritic cells
small-pathogen fragments (proteins, carbohydrates (DCs)-enriched areas, which are overlayed by epithe-
and peptides) provide a rational approach toward safe lial cells, and a small number of specialized microfold
immunizations. Unfortunately, induction of the desired cells (M cells) (Figure 1) . M cells are primarily respon-
immune responses with subunit-based antigens is often sible for transporting antigens from the apical side of
difficult to achieve even after parenteral immunization, the lumen to DCs or B cells [11] . The particulate-based
as they are poorly immunogenic due to the lack of dan- antigens are actively transported by M cells, whereas
ger signals. Therefore, an adjuvant or delivery system is soluble antigens are passively absorbed from the apical
required to enhance the immunogenic efficacy of sub- side of the epithelial membrane toward the basolateral
unit vaccines. Recently, nanoparticles have been pro- region. Following antigens’ transport via M cells or
posed as a promising self-adjuvanting delivery system to epithelial barriers, resident DCs capture and process
overcome the lack of effective mucosal adjuvant. antigens and present them locally to adjacent T cells

152 Ther. Deliv. (2017) 8(3) future science group


Intranasal delivery of nanoparticle-based vaccines Review

in the subepithelial region. Alternatively, subepithelial sites in the nasal epithelial cells that express CCL28,
DCs may extend their dendrites from transepithelial a mucosa-associated epithelial chemokine [13] . Finally,
junctions to capture antigens in the luminal region. IgA+ B cells undergo final differentiation and matura-
This special antigen capture behavior by DCs is exclu- tion to generate IgA+ plasma cells (process enhanced
sive to the areas in the nasal mucosa where there is an by cytokines such as IL-5 and IL-6, a subset of Th2
absence of organized lymphoid follicles and M cells. cells) and finally produce dimeric (or polymeric) forms
Once antigens are captured by DCs’ extensions, they of IgA. The dimeric IgA binds to polymeric Ig recep-
are actively transported to the nearest draining lymph tors (expressed by the basolateral surface of nasal epi-
nodes for antigen presentation to the CD4 + T cells [12] . thelial cells) to form sIgA. sIgA is then further trans-
In the lymph nodes or organized lymphoid follicles, cytosed toward the apical luminal surface in the nasal
DCs-stimulated CD4 + T cells activate naive B cells passage. sIgA antibodies are able to recognize, bind or
and these B cells undergo isotype class switching to neutralize the toxins or pathogenic micro-organisms,
form antigen-specific IgA+ committed B cells. These thus preventing their access to internal organs and
committed IgA+ secreting B cells express high levels of forming the first line of defense against pathogens.
CCR10 (mucosal homing receptors) and migrate from Unlike other antibodies, sIgA antibodies are special
lymph nodes to the blood circulation. The circulating and unique in nature as they are resistant to protease
IgA+ B cells exit the blood and the expressed CCR10 degradation at the mucosal surface [12] . They remove
in IgA+ B cells eventually guide their entry to effector antigens by blocking access to the epithelial receptor,

Effector site pIgR G


H

sIg
gA
sIgA Dimeric IgA

Nanoparticles
ticles
F
C
Dendritic
D
Deendritic
d i i cells
ce
ells
ll

IgA+ plasma cells


M cells CD4
4+ T cell
cells
ls
s
A

IgA+ B cells
D

B
E
Mucosa

IgA+ B cells
Nasal epithelium

CD4+ T cellsh nod


p
Lym
Systemic circulation
Lymph node

Figure 1. Mechanism of immune induction in nasal-associated lymphoid tissues. The particulate antigens are
actively taken up by M cells (A) or permeate passively through epithelial junctions (B). Alternatively, antigens
are taken up by DCs extensions into the lumen (C). Once antigens are captured by DCs, they migrate to nearest
lymphoid follicles or lymph node to present antigens to CD4 + T cells. DCs-stimulated CD4 + T cells activate naive
B cells and undergo isotype switching to form antigen-specific IgA + committed B cells (D). These IgA + B cells
migrate from lymph node to the blood circulations (E). Finally, IgA + B cells exit the blood and enter toward the
effector site. They undergo final differentiation and maturations producing IgA + plasma cells (process enhanced
by IL-5 and IL-6, a subset of Th2 cells) and ultimately form dimeric or polymeric IgA (F). The dimeric or polymeric
IgA binds with pIgR in the basolateral region to form sIgA (G) and further transcytosed toward the apical side of
luminal surface (H).
DC: Dendritic cell; IL: Interleukin; M-cell: Microfold cell; pIgR: Polymeric immunoglobulin receptor; sIgA: Secretory
IgA; Th: T-helper cell.

future science group www.future-science.com 153


Review Marasini, Skwarczynski & Toth

trapping them in mucus and preventing their adher- better enhance the immunogenicity of antigens com-
ence to mucus by mucociliary-mediated exclusions [13] . pared with their soluble antigen counterparts due to
Apart from mucosal sIgA, nasal immunization can the increased stability and lipophilicity of the vaccine
also induce systemic antibodies (IgG) in the periph- compounds [21–23] . However, the optimum number of
eral lymphoid tissues as observed in parenteral vaccines lipids or carbohydrate moieties required to enhance
that can clear pathogens from the blood circulation. the appropriate immune responses are often difficult
This feature makes the intranasal route highly efficient to predict. It was observed that increasing the length of
for the delivery of vaccines. lipid moieties conjugated to peptide epitopes allowed
them to self-assemble into nanoparticles (Figure 2A)
Nanoparticles as a mucosal adjuvanting and the systemic and mucosal immune responses
system were increased upon intranasal immunization [24–26] .
Adjuvants are usually used in modern vaccine formu- However, the involvement of complex synthetic and
lations to enhance the immunogenicity of the deliv- conjugation chemistries often gives a low yield, thus
ered antigens [14] . A significant progress in the devel- limiting their mass production. Alternatively, vaccines
opment of adjuvants for parenteral vaccine delivery can be encapsulated, adsorbed, simply conjugated or
has been achieved in the last few decades. The most physically mixed with biomaterials such as polymers
popular adjuvant used in human vaccine formulations and lipid-based nano-/microparticles (Figure 2B–F) . A
is alum, due to its favorable safety profile. Although large number of synthetic/natural polymers and lipids
alum triggers the induction of an antigen-specific sys- are widely used in intranasal drug or vaccine delivery.
temic immune response, it does not induce cellular and Codelivery of vaccine components into a single par-
mucosal responses [15] . Cholera toxins and heat-labile ticulate system mimics the characteristics of natural
toxin (LT) are highly effective mucosal adjuvants. pathogens and preserves the native structure of vac-
However, their use in humans has been constrained by cines in the extracellular and intracellular fluids. The
their toxicity and the possibility of antigens redirecting particulate nature of polymers or lipids, depending
to olfactory tissues or the CNS due to the capacity of upon their physicochemical properties (e.g., hydrophi-
B subunits of cholera toxin to bind with ganglioside- licity/lipophilicity, charge, morphology, composition,
receptors presenting on all nucleated cells [16,17] . These etc.) allows them to be selectively taken up by M cells
constraints have led to an interest in the development or nonselectively by epithelial cells before reaching the
of a safer and nontoxic alternative adjuvanting strategy lymph nodes and additionally, to control the release of
for nasally administered vaccines. antigens over the period of time.
The application of nanoparticles in mucosal vac-
cine delivery has the potential to overcome obstacles Polymer-based nanoparticles for nasal
associated with the classical adjuvants [8] . Vaccines vaccination
formulated into nano- or micro-particles showed more Natural and synthetic polymers are one of the most
efficient antigen-presenting cells (APCs) internaliza- well-established carriers of proteins, peptides and DNA-
tions, cross-presentation and B-/T-cell responses [18– based vaccines. The most commonly used p­olymers in
20] . In mucosal vaccination, the adjuvanting effects nasal vaccine delivery are polyesters derivatives such
of the nanoparticles are related to: improved ability as poly(lactic-co-glycolic acid; PLGA), polylactic acid
to cross mucosal barriers, enhanced uptake by APCs, (PLA) and their copolymers, polysaccharides (such
prolonged availability to interact with APCs and the as chitosan, alginate and dextran), poly(amino acids)
provision of additional danger signals due to their size [such as poly(γ-glutamic acid) (γ-PGA), poly( l -lysine),
mimicry of pathogens. This mode of delivery system is poly( l -arginine)] and polyacrylates.
becoming increasingly popular owing to their tunable
physicochemical characteristics, capacity to protect the Polyesters
native structural integrity of the antigens, presentation PLGA and PLA are the best examples of polyester
of antigens into multimeric form and delivery of anti- polymers with safe, biodegradable and biocompat-
gens to the targeted sites, particularly the lymph node. ible properties. The well-documented safety history
The limited permeation and rapid exclusion of of PLGA has resulted in a number of applications for
soluble antigens from the mucosal passage are often drug delivery. The polymer has been examined in sev-
the primary bottlenecks for the nasal delivery of vac- eral clinical trials and marketed in drug formulations
cines. Thus, the solubility or hydrophilicity of anti- but has yet to be successfully applied in commercial
gens (e.g., peptides or proteins) can be altered by vaccines [27,28] . Both PLGA and PLA nanoparticles are
conjugation with lipids or carbohydrates. Addition- usually prepared by solvent-evaporation/emulsification
ally, these modifications of antigens were shown to and nanoprecipitation methods that allow isolating

154 Ther. Deliv. (2017) 8(3) future science group


Intranasal delivery of nanoparticle-based vaccines Review

A B C

D E F

Figure 2. Different methods of antigen incorporation to nanoparticles. (A) Amphiphilic antigens can self-
assembled itself to form nanoparticles, (B) antigens are encapsulated into the nanoparticles, (C) antigens are
adsorbed on the surface and encapsulated into the nanoparticles, (D) antigens are adsorbed on the surface
of nanoparticles, (E) antigens are conjugated onto the surface of the nanoparticles and (F) physical mixture of
antigens and nanoparticles.
particles of ultrasmall size up to a minimum of 70 and due to poor mucoadhesiveness and immune-activating
20 nm, respectively [29,30] . Both the particle prepara- properties [33,34] . It was previously shown that positive-
tion processes require antigens or polymers to be dis- charged particles are more favored for enhanced muco-
solved into organic solvents that may cause denatur- adhesiveness over negative-charged particles [33,35,36] .
ation of antigens (especially proteins). Additionally, The carboxylic group (–COOH) of PLGA imparts a
these techniques often require the use of surfactants to negative charge to PLGA that disfavors interaction of
stabilize the particles. Therefore, the major difficulty PLGA with the mucosal surface leading to rapid expul-
during the production is to remove extra traces of the sions of particles from the nasal mucosal surface, thus
remaining surfactants in order to avoid cytotoxic side making fewer particles available for activation of the
affects [28] . immune system. Slütter et al. demonstrated that the
Polyesters polymers are able to encapsulate/adsorb in vivo nasal residence time of PLGA nanoparticles
large amounts of both hydrophilic and lipophilic anti- encapsulating ovalbumin (OVA) protein as antigens
gens and immunomodulatory compounds as a cargo was similar to the soluble OVA protein [37] . Polyesters
into the core/surface of a single particulate system [31] . were coated with polyethylene glycol (PEG) or polox-
These polymers are eroded slowly by the hydrolysis of amer 188 to increase hydrophilicity, circulation time
ester linkages into nontoxic metabolites (e.g., lactic and biological stability of the nanoparticles [38,39] .
acid and glycolic acids for PLGA) before their elimi- Vila et al. prepared PLA nanoparticles with or without
nation by the kidney [32] . While PLGA is extensively PEG-encapsulating tetanus toxoid (TT) antigens to
used parenterally, its use in nasal vaccination is limited compare their biological stability and i­mmunological

future science group www.future-science.com 155


Review Marasini, Skwarczynski & Toth

fate upon intranasal administration. While PLA Chitosan & their derivatives
nanoparticles without PEG aggregated in the pres- Chitosan is linear polysaccharide predominantly com-
ence of lysozyme, PEG-coated PLA nanoparticles were posed of β-(1–4)-linked d -glucosamine and N-ace-
stable [39] . Furthermore, in vivo evaluation of both tyl- d -glucosamine. The N-acetyl- d -glucosamine moi-
the nanoparticles showed that PEG–PLA nanopar- ety of chitosan is recognized by the mannose receptors
ticles penetrated nasal mucosae efficiently and elicited on the DCs [43] . Chitosan is produced by deacetylation
higher and more long-lasting immune responses com- of naturally occurring chitin (component of exoskel-
pared with PLA-only nanoparticles. Cu et al. showed etons obtained from crustaceans such as shrimps and
that PEG coating of the PLGA nanoparticles improved crabs) using a hot alkaline solution (aqueous sodium
particle diffusion across the mucosal barrier up to ten- hydroxides) [44] . Chitosan is biodegradable and showed
fold in comparison to uncoated particles [40] . Thus, an excellent safety profile with low toxicity [45] . Due to
PEG coating on the surface of polyesters may provide their mucoadhesive and immunostimulating proper-
an effective approach to deliver antigens by enhancing ties, chitosan is a promising alternative to poly(esters)
their stability and in vivo efficiency. for a nasal vaccine carrier [46] . The enhanced mucoad-
The low efficacy of vaccines due to reduced muco- hesive property of chitosan is mainly attributed to the
sal permeation of antigen can be overcome by spe- presence of the polycationic charge (amine moieties)
cifically targeting nanoparticles to nasal M cells in the chitosan that binds with anionic sialic acids (a
that may allow the rapid transport of nanoparticles principle component in mucus) [47] . Additionally, chi-
to the underlying lymphoid tissues. There are only tosan promotes the opening of the tight junctions in
a few reports on the use of M-cell-targeting ligands the epithelial membrane for efficient permeation of
employed with polyester-based nanoparticles for particles [48] . However, recent study showed that such
nasal vaccine delivery. Rajapaksa et al. encapsulated opening of tight epithelial junctions in the presence of
fusion protein into PLGA nanoparticles incorporat- chitosan might be not efficient for antigen transporta-
ing a mixture of protein antigens hemagglutinin- tion as it did not help ovalbumin protein to cross the
histidine tagged (HA–HT) protein antigens and a epithelial barrier [49] .
C-terminal targeting peptide derived from Clostrid- Vila et al. used chitosan nanoparticles for the intra-
ium perfringens enterotoxin (CPE) to bind Claudin nasal delivery of TT as model antigens in mice [50] .
4 receptors, which are expressed in both human and High- and long-lasting antigen-specific serum IgG
mouse M cells [41] . Nanoparticles prepared with tar- and mucosal IgA antibody titers were detected upon
geting peptide were taken up significantly better by immunization with chitosan nanoparticles. The effi-
NALT M cells compared with nontargeted nanopar- ciency of chitosan nanoparticles in generating humoral
ticles. The same group also reported the influence of immune responses was independent of the molecular
dispersion of PLGA nanoparticles formulated with or weight of the chitosan. Chitosan nanoparticles have
without M cells targeting ligand or poloxamer 188 also been successfully applied as a nonviral carrier
at various ionic strengths of buffers on antigen in for DNA vaccines [51,52] . Lebre et al. used chitosan
vivo uptake by NALT M cells following intranasal nanoparticles, human serum albumin and plasmid
immunizations [38] . The nanoparticles were dispersed DNA (encoding hepatitis B surface antigens [HBsAg])
into buffers with different ionic strengths ranging to develop a vaccine against the hepatitis B virus [52] .
from as high as 1.5 × 10 -1 M to a low ionic strength The nanoparticles were able to show high transfection
1 × 10 -7 M at pH 7, while osmolarity was controlled efficiency and induce significant humoral and mucosal
by addition of mannitol to bypass any influence of responses against hepatitis B virus.
osmolarity on uptake. Upon intranasal immuniza- Although chitosan nanoparticles possess inherent
tion, nanoparticles were highly taken up by NALT adjuvant properties, they are also used in conjunc-
at low ionic strength of phosphate buffer regardless tion with other immunomodulatory compounds
of the presence of targeting peptides or poloxamer. such as CpG oligodeoxynucleotide, Quillaja saponins
Thus, the capacity of long-term electrostatic interac- (QS), monophosphoryl lipid A and mast cell-activat-
tions between the particles and mucosa (mucoadhe- ing adjuvant compound 48/80 (C48/80) for nasal
siveness) was the more important factor influencing i­mmunizations [53–56] .
antigen uptake in NALT than the use of a targeting The amine group of chitosan has a dissociation con-
moiety. Therefore, several efforts were undertaken to stant (pK a) 6.2, thus at pH > 6.2 chitosan is deprot-
modify the surface of PLGA nanoparticles by using onated, making them insoluble in water. This could
mucoadhesive polymers (such as chitosan and their prevent encapsulation of antigens that are only soluble
derivatives) to improve their mucus-binding efficacy at a neutral or higher pH. Therefore, chitosan is often
and nasal residence time [35,37,42] . modified to improve its aqueous solubility in a wider

156 Ther. Deliv. (2017) 8(3) future science group


Intranasal delivery of nanoparticle-based vaccines Review

range of pH [57] . For example, the chitosan amine were less efficiently taken up by APCs, showed low
group can be methylated to form trimethyl chitosan nasal residence time and induced weak systemic and
(TMC), which bears a permanently ionized quater- mucosal antibody titers. This further signifies that
nary amine group [58–60] . The TMC nanoparticles can incorporation of alginate into the delivery system neg-
be simply prepared at ambient temperature by ionic atively affects vaccine responses, indicating that algi-
gelation of cationic TMC and anionic tripolyphos- nate in an unfavorable polymer for intranasal delivery
phate under stirring, without the requirement of soni- of vaccines.
cation and presence of organic solvents, thus providing
a lower possibility of antigen denaturation [59] . Dextran
The mode of antigen delivery could play an impor- Dextran is a polysaccharide consisting of a linear
tant role in determining the immunogenicity of anti- repeating unit of α-1, 6-linked glucopyranose. It is
gens. Antigens can be delivered with chitosan or its used in drug delivery as mucosal absorption enhanc-
derivatives as physical mixtures or as a conjugate with ers. Dextran is efficiently recognized by the mannose
antigen [61–64] . Covalent conjugation of an antigen receptor present on the DCs [68] .
with chitosan-based polymers shows better systemic Dextran is a hydrophilic polymer and its derivative
and mucosal antibody responses than antigens encap- (dextran sodium) is most commonly used in drug or vac-
sulated or mixed with chitosan [61–63] . The conjugate cine delivery. Dextran sulfate is polyanionic due to the
approach is most beneficial for the delivery of low presence of a large number of sulfate groups [69] . Chen
immunogenic protein and peptide antigens. and colleagues developed dextran/chitosan nanopar-
Chitosan has also been used to modify the sur- ticles with IgA as targeting ligand using the polyelec-
face properties of anionic polyesters. Coating anionic trolyte complexation/ionic gelation method [70] . These
PLGA particle surfaces with chitosan-based polymers nanoparticles were able to incorporate pertussis toxin
led to the formation of the cationic nanoparticles with (PTX) and targeting ligand (IgA) with high entrap-
slightly increased size, improved nasal residence and ment efficiency (>90%). Nanoparticles were easily
enhanced humoral immunity against administered uptaken in vivo in the NALT area. However, no in vivo
antigens [35,37] . Taken together, in contrast to the immunization was conducted to show the efficacy of
majority of classical adjuvants which are associated particles in inducing antibody responses. Jaafari and
with some degree of side-effects or toxicity, the use of colleagues prepared cross-linked dextran microspheres
highly immunogenic chitosan-based nanoparticles is a (CDM) loaded with TT. CDM loaded with TT were
promising intranasal vaccine delivery strategy. able to induce potent antigen-specific serum IgG and
IgA antibody titers in rabbits after intranasal immuni-
Alginates zation [71] . Additionally, high-serum anti-TT antitoxin
Alginate is a hydrophilic anionic polysaccharide titers (175-times higher than required to achieve a pro-
obtained from brown algae and mostly used in drug tective level) were induced by dextran-based micro-
and vaccine delivery [2,65] . It is nontoxic, biodegradable spheres. Similarly, CDM physically mixed with alginate
and biocompatible. Alginates can be used to encapsu- microspheres loaded with QS and TT induced a high
late an antigen, protect it from enzymatic degradation mucosal and systemic antibody response while QS and
and facilitate its suitable release. Although the appli- TT co-encapsulated into alginate were less effective [72] .
cation of alginate as a carrier in an intranasal vaccine Despite being negatively charged, dextran can enhance
delivery system was reported, the polymer is mainly the uptake of particles in the nasal mucosa. Although
used for the oral delivery of antigens. the exact mechanism is poorly understood, it could be
Borges et al. demonstrated that alginate coating of hypothesized that solid dextran microsphere powder
the HBsAg/chitosan complex induced a detectable adsorbs water from the mucus layer that may shrink the
mucosal response in nasal washes but did not pro- mucosa and allow the penetration of the particles across
duce sufficient systemic responses upon intranasal the mucosal barrier [72] .
immunization [66] . The authors suggested that the
poor immune responses might be related to muco- Poly(amino acids)
sally induced tolerance against the antigens. Addition- Recently, poly(amino acids) have become increasingly
ally, the low mucoadhesive property of alginate could popular for delivering vaccines, genes and proteins [73] .
explain the poor efficacy of nanoparticles. In a similar Poly(amino acids) contain the repetition of a small
study, antigen encapsulated into chitosan and chito- number of the same amino acid. The poly(amino acids)
san/alginate nanoparticles was compared for nasal can be conjugated between each other (hydrophobic and
immunization [67] . In contrast to chitosan-only bear- hydrophilic poly(amino acids)) to form an amphiphi-
ing nanoparticles, chitosan/alginate nanoparticles lic block copolymer-like structure. In an aqueous solu-

future science group www.future-science.com 157


Review Marasini, Skwarczynski & Toth

tion, an amphiphilic poly(amino acids)-based system vaccines. Polyanhydride nanoparticles were prepared
can form nanoparticles by spontaneous self-assembly of by the copolymerization of methyl vinyl ether and
their hydrophilic and hydrophobic moieties [74] . maleic anhydride (PVM/MA). PVM/MA polymers
γ-PGA is a safe, biodegradable natural material that are considered safe and have been licensed for human
was examined for the delivery of vaccines against vari- use [84,85] . Polyanhydrides showed a great potential in
ous diseases such as influenza, anthrax and noninva- drug or vaccine delivery attributed to their mucoadhe-
sive cancers [75–77] . Matsuo et al. conjugated hydropho- sive, controlled release and adjuvanting properties (as
bic l -phenylalanine ethyl ester ( l -PAE) into α-position they are Toll-like receptor (TLR)-2, TLR4 and TLR5
of carboxyl functional group of γ-PGA and self-assem- agonists) [84,85] . Gamazo and colleagues used outer
bled into nanoparticles (γ-PGA- l -PAE) that effectively membrane vesicles (OMV) derived from Shigella flex-
entrapped tumor-associated antigen (OVA) [77] . Upon neri as the antigen component and encapsulated it into
intranasal vaccination with γ-PGA- l -PAE nanopar- polyanhydride nanoparticles [86] . When tested in mice
ticles, highly potent tumor-specific long-term cellular intranasally, OMV-loaded polyanhydride nanopar-
immune responses were observed. Additionally, this ticles enhanced antibody titers, IFN-γ and IL-10 pro-
vaccine induced strong systemic IgG titers, whereas duction. Upon challenge in mice with S. flexneri 2a,
very low mucosal IgA titers were detected. In contrast, nanoparticle-vaccinated groups showed 100% survival
when influenza virus hemagglutinin (HA)-loaded rate for up to 15 days indicating the enhanced protec-
γ-PGA nanoparticles were administered nasally, they tion capacity of OMV-encapsulated nanoparticles.
elicited high mucosal antibody titers and cross-protec- Polyacrylates were used as a vaccine delivery system
tive immune responses sufficient for mice survival [75] . due to their bioadhesive property and mucosal-adju-
Interestingly, unmodified γ-PGA is more popular in vanting effects [87–89] . Recently, Zaman et al. prepared
drug and parenteral vaccine delivery but it has not a polyacrylate-based delivery system by conjugating
been used on its own in nasal vaccine delivery. polyacrylate dendrimers with peptide antigens [87] .
Poly(l-lysine) is widely used alone or grafted with Upon intranasal immunization, polyacrylate-based
other polymers to deliver DNA vaccines. The cat- nanoparticles induced systemic antigen-specific IgG
ionic functional group (-NH3 +) of lysine moiety effi- responses; however, mucosal IgA antibodies were not
ciently condenses anionic plasmid DNA into com- observed.
pact nanoparticles [78] . Wu et al. reported the use of
poly(l-lysine) to deliver DNA vaccines to NALT. M Lipid-based nanoparticles
cells targeting recombinant protein sigma 1 (σ1) were Antigens can be delivered using varieties of nontoxic
conjugated to polylysine and complexed with DNA vac- and biodegradable lipids. Lipid-based nanoparticles
cines [79] . Upon intranasal vaccination with this delivery are popular vaccine delivery systems and some of them
system in mice, higher levels of both humoral and cel- have already found application in the commercialized
lular immune responses were observed in comparison to vaccines. Most of these particles showed mucoadhesive
naked DNA vaccines. properties. However, in contrast to chitosan, they were
Poly(l-arginine) is a safe, biodegradable cationic unable to open tight epithelial cell junctions in NALT
poly(amino acids) and successfully applied in Phase for antigen transportation across the membrane [90] .
II clinical trials for hepatitis C virus-based peptide
vaccines [80] . Poly(l-arginine) along with dextran sul- Liposomes
fate form polyelectrolyte capsules, which encapsulate Liposomes were first reported in the mid-1960s. Cur-
a large amount of antigens [81,82] . The cationic nature rently, they are one of the most successful drug and
of poly(l-arginine) nanoparticles enhances mucosal vaccine delivery systems. Several liposome-based prod-
membrane permeation of antigens [83] . De Koker et al. ucts are already in the market and under clinical tri-
showed that vaccination with poly(l-arginine)-based als [91] . Liposomes are spherical self-assembled lipid
capsule-encapsulating OVA antigens induced local bilayer vesicles with an aqueous core [92] . This unique
mucosal and cellular responses against adminis- feature of liposomes enables encapsulation of both the
tered antigens [82] . While mucosal immunization of hydrophilic or lipophilic agents into their aqueous core
poly(l-arginine) nanoparticles by the pulmonary route or lipid bilayer surface, respectively. Liposomes are usu-
has been reported, their use as a nasal vaccine delivery ally prepared from natural or synthetic phospholipids
system remains to be investigated [82] . with or without the addition of cholesterols. Cholester-
ols stabilize the liposomes’ bilayer by enhancing their
Other polymers rigidity. Classical neutral liposomes possess several
Other polymers such as polyanhydride and polyac- advantages when delivered parenterally; however, the
rylates were also examined for intranasal delivery of mucosal delivery of liposomes is often problematic due

158 Ther. Deliv. (2017) 8(3) future science group


Intranasal delivery of nanoparticle-based vaccines Review

to their low mucoadhesiveness or the reduced capac- interactions between particles and immune cells. In
ity of liposomes to permeate the epithelial membrane. addition, coated liposomes are usually more stable
These shortcomings have been overcome by using cat- under storage and physiological conditions. Lipo-
ionic liposomes or modulating the surface properties of some–polymer hybrid nanoparticles were used to
liposomes with cationic polymers. deliver antigens or mixtures of antigens and immu-
Cationic liposomes are known as a safe and potent nomodulatory agents [64,100] . Recently, liposomes-
mucosal delivery system [93] . Recently, Tada et al. bearing cationic moiety, 1,2-dioleoyl-3-trimethyl-
showed that the intranasal administration of OVA- ammonium propane (DOTAP), were prepared for
loaded cationic liposomes promotes antigen uptake encapsulation of F1-V antigen and MPLA [100] . These
by NALT DCs [94] . Upon intranasal immunization in cationic liposomes were electrostatically complexed
mice, cationic liposomes induced an enhanced immune with anionic-thiolated hyaluronic acid and stabilized
response (IgA and IgG antibody titers) against OVA with thiolated-PEG-forming DOTAP/HA core–PEG
compared with soluble antigen, anionic and neutral shell nanostructures. These lipid–polymer hybrid
liposomes. Slütter et al. reported contradictory find- delivery systems (DOTAP/HA core–PEG shell nano-
ings on the benefit of using cationic liposomes as a structures), co-encapsulating adjuvant and antigens,
nasal vaccine delivery system [95] . Efficiency of three induced a high level of mucosal, systemic and cellu-
different formulations: OVA antigen, OVA+CpG and lar immune response upon intranasal immunization
OVA/CpG/liposomes was compared upon intranasal in mice compared with soluble OVA and mixture of
immunization in mice. OVA/CpG-loaded liposomes monophosphoryl lipid A (MPLA) + OVA.
had the highest uptake by DCs in vitro, while they
showed poor uptake in vivo in mice in comparison Emulsions
to OVA+CpG. Moreover, the serum IgG responses Emulsions are primarily isotropic mixtures of two
in mice upon intranasal immunization for liposomes immiscible liquids (oil and water) stabilized by the
loaded with OVA+CpG were lower than OVA+CpG. surfactants, and are capable of producing particle sizes
Mucosal antibody responses were not reported. How- below 200 nm [101] . Nanoemulsion-encapsulating anti-
ever, due to the use of very strong adjuvant (CpG) gens increase the stability of antigens against enzy-
and lack of proper control (OVA-loaded liposomes), it matic degradations and control their release. The most
could not be concluded that cationic liposomes lack well-known emulsions used for vaccine delivery are
adjuvanting effects. In addition, except where men- squalene-based o/w emulsions (MF 59 and AS03) that
tioned above, OVA/CPG/liposomes’ as alternative have been applied in marketed vaccines [102] . How-
delivery system utilizing cationic liposomes showed ever, their application is essentially limited to only the
clear benefits of this formulation [96,97] . p­arenteral route.
The efficiency of a liposome-based nasal vaccine can Makidon et al. studied a novel nanoemulsion formu-
be improved by modifying its surface functionality. lation based on soybean oil-in-water (o/w) for subunit
Among various pathogen-recognizing receptors, APCs vaccine delivery [103] . The soybean-based nanoemulsions
express C-type lectin receptors that recognize the car- were effectively internalized across the epithelial mem-
bohydrates domain and efficiently promote antigen brane by the transcellular process and did not require
presentation on MHC I and MHC II molecules [98] . the opening of tight junctions or M cells’ participa-
APCs can be targeted using carbohydrates on the tion for its enhanced permeation. This nanoemulsion
surface of liposomes [98,99] . Wang et al. prepared lipo- platform was used to develop a vaccine against hepati-
somes surface-modified with carbohydrate domain- tis B [104] . HBsAg-loaded nanoemulsion was tested for
galactose [99] . The OVA-loaded galactosylated lipo- its efficacy in different animal models using intranasal
somes were efficiently endocytosed by macrophages administration. Robust systemic, mucosal and cellu-
and produced a significant amount of systemic and lar antigen-specific immune responses were observed
mucosal antibody titers upon intranasal immunization and the serum IgG antibody titers were comparable
in mice, compared with free antigens and unmodified to those induced by alum-adjuvanted HBsAg upon
liposomes. i­ntramuscular a­dministration in mice.
Liposomes can be additionally modified with poly- Nanoemulsions provide a mucoadhesive effect by
mers such as chitosan, TMC or hyaluronic acids [92] . interacting with mucin and these interactions depend
The surface modification of liposomes with mucoad- on the properties of oils and surfactants used in the
hesive polymers not only provides for the additional formulations; for example, the hydrophilic-lipophilic
protection of antigens against enzymatic degradation balance value and charge in the surfactants. The
but also allows them to retain antigens into the nasal overall charge in the nanoemulsion can be altered by
mucosa for prolonged periods of time, thus e­nhancing varying nanoemulsion compositions. By varying the

future science group www.future-science.com 159


Review Marasini, Skwarczynski & Toth

s­urfactants or altering the ratio of surfactant mixtures lipopeptides such as tripalmitoyl-S-glyceryl cysteine
used in nanoemulsions, the type of immune responses (Pam3Cys) and its derivative (Pam2Cys) are commonly
(Th1, Th2 and Th17) were altered [105] . Wong et al. used to induce potent immunity against peptide-based
observed that a highly cationic nanoemulsion had antigens [114] . The adjuvant activity of these bacterial
more affinity toward mucin than uncharged and lipopeptides is due to the recognition of lipid moiety
anionic nanoemulsion [106] . by the TLR2, a pathogen-associated molecular pattern
receptor present on the APCs [115] . A comparison of
Immunostimulating complexes efficacy between Pam2cys and Pam3Cys showed that
Immunostimulating complexes (ISCOMs) are self- Pam2Cys containing peptide epitopes were more immu-
assembled cage-like structures with icosahedral sym- nogenic than Pam3Cys containing peptide epitopes,
metry composed of antigens, QS, phospholipids and which was most likely related to the poor aqueous solu-
cholesterol [107] . They are usually 40-nm-sized parti- bility of Pam3Cys [116] . Jackson and colleagues observed
cles and incorporate hydrophobic antigens into their that upon conjugation of Pam2Cys or Pam3Cys into
structure. The efficiency of antigens’ incorporation the middle of a long peptide sequence (carrying B cells
into ISCOMs is mainly related to the strength of the and T-cell epitopes; lipopeptide 1, Figure 3), the formed
hydrophobic interactions of antigens and the phospho- lipopeptides were more water-soluble than when lipids
lipids [107,108] . ISCOMs-based vaccines incorporating were attached to the end of the N-terminal moiety of
recombinant HBsAg were shown to enhance muco- the peptide antigen (lipopeptide 2) [116] . Upon intrana-
sal, systemic and cellular immunity upon intranasal sal immunization in mice, the branched lipopeptide 1
immunization [109] . More importantly, ISCOMs gen- (Figure 3) containing Pam2Cys as a lipid moiety showed
erate both Th1 and Th2 immune responses, while significantly higher systemic antibody titers than a
the most popular cholera toxin generates only Th2 linear Pam2Cys containing lipopeptide 2. Intranasal
responses against administered antigens [108] . administration of Pam2Cys with influenza virus antigen
Classical ISCOMs also known as Quil A® derived induced secretion of IL-2, IL-6, IL-10, IFN-γ, MCP-1
from Q. saponaria contain saponins, which are toxic and and TNF-α and protected mice against the influenza A
therefore their clinical application was restricted [110]. virus challenge [117] .
Consequently, the saponins were substituted with Lipopeptides can self-assemble in water to form
less toxic analogs known as QB-90 obtained from both nano- and microsized particles [118] . The self-
Q. brasiliensis. QB-90 induced comparable or higher assembling property of lipopeptides relies on the
antigen-specific mucosal and systemic antibody titers hydrophilic-to-lipophilic balance or interactions
than Quil A [111,112] . Cibulski et al. prepared ISCOMs among peptides and lipids [118,119] . Toth and colleagues
comprising QB-90, phospholipids, cholesterol and introduced self-adjuvanting lipopeptides, also known
OVA, and studied their efficiency to induce immune as lipid core peptides (LCPs), as a vaccine candidate
responses in mice after intranasal administration [111] . against group A streptococcus infection [120,121] . The
Mice immunized with ISCOMs (QB-90) showed sig- LCP-based vaccine candidates are chemically synthe-
nificantly higher antigen-specific serum and mucosal sized in a pure and defined molecular form by standard
immunity at nasal and distal mucosal sites such as the solid-phase peptide synthesis using either Boc or Fmoc
vagina and intestines, compared with antigens alone chemistry. LCPs contain lipoamino acids conjugated
and classical Quil A-based ISCOMs. to lysine moiety(s) via an amino acids linker (usually
Particles delivered via the nasal route should pos- two serines). The lysine(s) serve as branching moiety
sess low mucociliary clearance for efficient uptake in to which multiple peptide epitopes are attached [120] .
the NALT. Pandey et al. used the gamma scintigraphy Toth et al. incorporated a B-cell epitope (J14) derived
technique to show that ISCOMs were retained in the from a highly conserved region of M-protein from
nasal cavity for at least 2 h, while the control sodium group A streptococcus infection and a universal
pertechnetate solution was rapidly cleared [113] . The T-helper cell epitope (P25) into the LCP system. The
hydrophobic property of ISCOMs was found to be cru- LCP construct improved antigen-specific mucosal and
cial for ISCOMs’ deposition in the nasal tract. Thus, systemic immune responses upon intranasal immuni-
the use of mucoadhesive ISCOMs seems an alternative zation [26,122] . They also studied the structure activity
for the intranasal delivery system for vaccines. relationship of spatial arrangements of epitopes and
lipids [26] . The lipopeptide 3 (Figure 3) elicited high
Lipopeptides antigen-specific mucosal titers. In contrast, when
Lipopeptides are amphiphilic molecules containing sin- the position of epitopes and lipids were rearranged
gle or multiple copies of peptides covalently conjugated (lipopeptides 4 and 5), the IgA antibody titers were
to one or more lipidic moieties. The bacterial-derived s­ignificantly lower.

160 Ther. Deliv. (2017) 8(3) future science group


Intranasal delivery of nanoparticle-based vaccines Review

Recently, Ghaffar et al. showed that the combina-


tion of cationic liposomes and lipopeptides provided Peptide 1 Peptide 2
a synergistic effect on immune responses upon intra-
nasal administration in the mice model [96] . Different
lipopeptides, varying in the number of lipids/peptides, Lipids 1 or 2 Lipopeptide 1
were anchored to liposomes. After three intranasal
immunizations, lipopeptides bearing two lipids in
the final construct showed the highest and most long-
lasting mucosal and systemic antibody titers compared Lipids 1 or 2 Peptide 1 Peptide 2
with free lipopeptides (containing the same number of
lipid moieties), peptides encapsulated into liposomes Lipopeptide 2
or lipopeptides-bearing single lipid moiety encapsu- Peptide 3
lated into liposomes. The immunogenic efficiency of
Lipid 3
lipopeptide-loaded liposomes upon intranasal admin-
istration was further enhanced by coating of liposomes Peptide 4 Lipopeptide 3
with chitosan-based polymer [64] . Recently, cationic
lipopeptides were incorporated either on the surface or
encapsulated into PLGA nanoparticles [123] . Nanopar- Peptide 4 Peptide 3
ticles-bearing lipopeptide antigens inside the PLGA
nanoparticles showed significant improvement in
inducing antigen-specific mucosal and systemic anti-
Lipid 3 Lipopeptide 4
bodies production compared with lipopeptide-coated
PLGA nanoparticles.

Does size of the nanoparticles influence Peptide 3 Peptide 4


nasal immune responses?
The role of size of nanoparticles in APCs’ uptake
efficacy, their transportation from injection sites to Lipid 3 Lipopeptide 5
the lymph nodes, their stability in the systemic cir-
culations and their potency to induce antigen-specific Figure 3. Schematic representations of synthetic lipopeptides with
immune responses are extensively reviewed else- different spatial arrangement of epitopes. Lipopeptides 1 and 2
where [18,19,124,125] . However, these reviews focused on contained Peptide 1: T-cell epitope (GALNNRFQIKGVELKS) obtained from
vaccines administered by the parenteral route. Unlike influenza virus hemagglutinin; Peptide 2: B-cell epitope (EHWSYGLRPG),
parenteral vaccines, intranasal nanoparticle-based vac- a leutinizing hormone-releasing hormone; and Lipid 1: Pam2Cys or Lipid
2: Pam3Cys homologous to gram-negative bacterial outer membrane lipid.
cines primarily are required to cross mucosal barriers Lipopeptides 3, 4 and 5 contained Peptide 3: universal T-helper cell epitope
before being available to APCs for further antigen (P25: KLIPNASLIENCTKAEL) derived from canine distemper virus; Peptide
processing. Alonso and colleagues prepared different 4: B-cell epitope (J14: KQAEDKVKASREAKKQVEKALEQLEDKVK) derived
sized PLA–PEG particles (200 nm, 1.5, 5 and 10 μm) from M-protein of group A Streptococcus; Lipid 3: two copies of synthetic
encapsulating radio-labeled model protein, TT [126] . lipoamino acids (2-R/S-amino hexadecanoic acid).Pam2Cys: Dipalmitoyl-S-
glyceryl cysteine; Pam3Cys: Tripalmitoyl-S-glyceryl cysteine.
The 200-nm-sized particles demonstrated stronger
mucosal permeation ability and higher bioavailability showed a diverse range of particle sizes upon self-
than microparticles. assembling into phosphate-buffered saline (5, 10 and
While there is no doubt that parenterally admin- 50–100 nm). The smallest size nanoparticles (5 and 10
istered small nanoparticles (10–50 nm) induce nm) were efficiently taken up by APCs leading to high
humoral immunity more efficiently than larger size APCs maturations in vitro. Furthermore, intranasal
nanoparticles, several contrasting reports on optimum immunization with small-size nanoparticles (5 and 10
nanoparticle size for enhanced immunogenicity have nm) induced the highest systemic IgG antibody pro-
been reported for intranasal vaccines [125] . Recently, ductions compared with larger size particles (50–100
Toth and colleagues showed that the immunogenic- nm). However, production of mucosal antibody titers
ity of lipopeptide-based vaccines relies on the size of was not reported. In another study, the role of particle
nanoparticles [20] . Three different LCP vaccine candi- sizes after intranasal administration of lipopeptide/
dates incorporating B-cell epitopes (J14 and LP-88/30) liposomes vaccines was examined [97] . Various sizes
and universal T-helper cell epitope (P25) were pre- (70, 140, 400 and 150–1000 nm) of lipopeptide-loaded
pared. These self-adjuvanting LCP vaccine candidates liposomes were prepared. The smaller sized liposomes

future science group www.future-science.com 161


Review Marasini, Skwarczynski & Toth

induced the production of higher mucosal IgA and vaccine development is the requirement to cross the
systemic IgG antibody titers than larger liposomes; mucosal membrane to be available for DCs to activate
however, the difference was not statistically significant. the immune cascade. As discussed in this review, poly-
Tada et al. formulated different sized OVA-loaded cat- meric or lipid-based nanoparticles that mimic the size
ionic liposomes (50, 100 and 1000 nm) and studied of pathogens are an attractive choice for nasal vaccina-
their influence in inducing humoral responses [94] . No tion owing to their ability to permeate transcellularly
size-based correlation on antigen-specific mucosal IgA or paracellularly across the tight epithelial junctions,
and systemic IgG responses was observed. In contrast, and their potential to target M cells in the NALT
other studies showed that larger nanoparticles induced region. While there is no clear correlation between
better humoral immunity than smaller sized counter- size of nanoparticles and their mucosal immunoge-
parts. For instance, Gutierro et al. observed that 1000- nicity, most studies agree that nanosized particles are
nm sized BSA-encapsulated PLGA nanoparticles were more efficient in inducing immune responses than
more efficient in inducing systemic immunity com- microsized particles. Several other factors such as the
pared with 500- and 200-nm particles, upon intrana- efficiency of antigen loading, surface charge, muco-
sal administration. Their effect on mucosal immunity adhesiveness, antigen release profile, nature and the
was not reported [127] . Similarly, Stano et al. compared composition of biomaterials influence overall immune
efficiency of different-sized OVA-conjugated pluronic- responses.
stabilized polypropylene sulfide nanoparticles (30 vs Polyesters (e.g., PLGA and PLA) and polysaccha-
200 nm) upon intranasal administration in mice [128] . rides (e.g., chitosan and its analogs) and self-adju-
They showed that large nanoparticles (200 nm) with a vanting lipopeptides have shown promising potential
slight anionic charge (-6.5 mV) induced significantly as mucosal vaccine delivery systems. However, deliv-
stronger mucosal and systemic antibody titers than ery of antigens in nanoparticle-based delivery systems
smaller nanoparticles (30 nm) with cationic charge intended for nasal administration need to overcome
(+8 mV). several practical hurdles such as the possibility of anti-
Apparently, many physicochemical properties of gen denaturation by the use of organic solvents during
nanoparticles, apart from the size, influence the per- nanoparticle production, low antigen entrapment into
meation of mucosal membrane (e.g., lipophilicity, nanoparticles and potential cellular toxicity. Thus,
molecular weight of polymeric nanoparticles as well as there is a manifest need to develop a nanoplatform
their stability and charges) [129] . To properly examine for nasal vaccines using particle-engineering strategies
any size-immunogenicity correlation, other properties that decrease these risks, maintain long-term antigen
of nanoparticles must be maintained constant, which is retention in the nasal mucosa and prolong antigen
very difficult to achieve in most cases. Thus, the role of availability to interact with immune cells. Some of the
particle size in mucosal and systemic immunity upon strategies include functionalization of the particle sur-
intranasal vaccination is still waiting for careful and face by conjugating with antigens or M cells target-
thorough examination. Taken together, at the current ing ligands, and modification of the physicochemical
stage, most studies report that small-sized particles properties of the particles (optimal particle composi-
were more efficient in mucosal barrier permeation and tion, charge, shape and hydro-/lipophilicity), etc.
induction of immunity against administered antigens. There are long lists of clinical trials for nasal vac-
cines, while only a few nasal vaccines such as Flu-
Conclusion & future perspective mist (USA), Fluenz (EU), Nasovac (India) have been
Owing to the large number of pathogens that infect approved for human use. However, no current clini-
through the mucosal surface, a mucosal vaccine deliv- cal trial exists for a nanoparticle-based vaccine in the
ery route is expected to be a popular nature-mimicking database of the US National Institute of Health (clini-
path for vaccine administration. While parenteral vac- caltrials.gov) [130] . In vivo studies discussed in this
cines induce only systemic immunity, mucosal vac- review are mostly based on preclinical data and cannot
cines are able to induce both the mucosal and systemic be generalized to humans. However, animal model-
immunity. This unique feature of mucosal vaccines based investigations provide some promising forecasts
allows mucosal IgA antibodies to opsonize or bind for future human studies with nanoparticles. The use
the pathogens at the primary site of pathogen expo- of safe, biodegradable, biocompatible and biologically
sure (mucosa), thus preventing further systemic inva- stable nanoparticles creates high hopes of translating
sions. Mucosal vaccines are particularly important for nasal nanoparticle vaccines from laboratory to clini-
blocking the spread of diseases from nonsymptomatic cal use. The vaccine candidates should be well-defined
patients carrying the pathogen on their mucosal tissue. chemically and produced in its purest form to avoid any
Unlike parenteral vaccines, the major hurdle for nasal toxicity issues. In the near future, novel conjugation

162 Ther. Deliv. (2017) 8(3) future science group


Intranasal delivery of nanoparticle-based vaccines Review

strategies that combine synthetic peptide-based epi- is a recipient of an international postgraduate research schol-
topes (single or multiple) with M-cell-targeting moi- arship (IPRS) and Australian postgraduate award (APA) for his
eties, TLR agonists lipids, carbohydrates, dendrimers PhD study. The contents are solely the responsibility of authors
and polymers into single nanoparticles shall provide a and do not necessarily represent the official views of NHMRC.
rational approach to successfully deliver nasal vaccines The authors have no other relevant affiliations or financial in-
and tailor the required immune responses. volvement with any organization or entity with a financial inter-
est in or financial conflict with the subject matter or materials
Financial & competing interest disclosure discussed in the manuscript apart from those disclosed.
This work was supported by the National Health and Medical No funded writing assistance was utilized in the production
Research Council (NHMRC), Australia. The author, N Marasini, of this manuscript.

Executive summary
Background
• Most pathogens gain access to the human body through mucosal sites and initiate systemic infections.
• Parenterally administered vaccines induce strong systemic immunity and almost no mucosal immunity.
• Nasal vaccination seems an appealing strategy for the induction of antigen-specific systemic and mucosal
immunity.
• A safer mucosal adjuvanting strategy or mucosal platform technology for intranasal immunization is much
warranted.
Nanoparticles as nasal adjuvanting system
• Particulate vaccines formulated into nanosize but not microsize showed more efficient APC internalizations,
cross-presentation and B- and T-cell responses.
• Adjuvanting effects of the nanoparticles are related to their improved ability to cross mucosal barriers,
enhanced uptake by APCs, prolonged availability to interact with APCs and provision of additional danger
signals due to their size mimicry of pathogens.
• Nanoparticles are functionalized by conjugating with antigens or M-cell-targeting ligands, and modification
in physicochemical properties of the particles (optimal particle composition, charge, shape and hydro/
lipophilicity, etc.) to enhance immunogenicity of administered antigens.
• The surface of nanoparticles can be modified by using mucoadhesive polymers (such as chitosan and their
derivatives) or imparting a cationic charge on the particle’s surface to improve mucus-binding efficacy and
nasal residence time.
Conclusion & future perspective
• The use of safe, biodegradable, biocompatible and biologically stable nanoparticles as nasal delivery systems
creates a high hope of translating nasal nanoparticle vaccines from laboratory to clinical use.

6 Mcentee C, Lavelle EC, O’hagan DT. Chapter 63 – Antigen


References delivery systems I: nonliving microparticles, liposomes, and
Papers of special note have been highlighted as:
immune-stimulating complexes (ISCOMs) A2 – J Mestecky.
• of interest; •• of considerable interest
In: Mucosal Immunology (4th Edition). Strober W, Russell
1 Dye C. After 2015: infectious diseases in a new era of health MW, Kelsall BL, Cheroutre H, Lambrecht BN (Eds).
and development. Philos. Trans. R. Soc. Lond. B. Biol. Sci. Academic Press, Boston, MA, USA, 1211–1231 (2015).
369(1645), 20130426 (2014).
7 Mcnaughton R, Lynn E, Osborne V, Coughtrie A, Layton D,
2 Marasini N, Skwarczynski M, Toth I. Oral delivery of Shakir S. Safety of Intranasal Quadrivalent Live Attenuated
nanoparticle-based vaccines. Expert Rev. Vaccines 13(11), Influenza Vaccine (QLAIV) in children and adolescents: a
1361–1376 (2014). pilot prospective cohort study in England. Drug Saf. 39(4),
3 Csaba N, Garcia-Fuentes M, Alonso MJ. Nanoparticles 323–333 (2016).
for nasal vaccination. Adv. Drug Del. Rev. 61(2), 140–157 8 Skwarczynski M, Toth I. Peptide-based synthetic vaccines.
(2009). Chem. Sci. 7(2), 842–854 (2016).
4 Zaman M, Chandrudu S, Toth I. Strategies for intranasal 9 Debertin AS, Tschernig T, Tönjes H, Kleemann WJ, Tröger
delivery of vaccines. Drug Deliv. Transl. Res. 3(1), 100–109 HD, Pabst R. Nasal-associated lymphoid tissue (NALT):
(2013). frequency and localization in young children. Clin. Exp.
5 Lycke N. Recent progress in mucosal vaccine development: Immunol. 134(3), 503–507 (2003).
potential and limitations. Nat. Rev. Immunol. 12(8), 10 Liang B, Hyland L, Hou S. Nasal-associated lymphoid tissue
592–605 (2012). is a site of long-term virus-specific antibody production
• Highlights recent updates on mucosal vaccinations. following respiratory virus infection of mice. J. Virol. 75(11),
5416–5420 (2001).

future science group www.future-science.com 163


Review Marasini, Skwarczynski & Toth

11 Csencsits KL, Jutila MA, Pascual DW. Nasal-associated 26 Zaman M, Abdel-Aal AB, Fujita Y et al. Structure-activity
lymphoid tissue: phenotypic and functional evidence for relationship for the development of a self-adjuvanting
the primary role of peripheral node addressin in naive mucosally active lipopeptide vaccine against Streptococcus
lymphocyte adhesion to high endothelial venules in a pyogenes. J. Med. Chem. 55(19), 8515–8523 (2012).
mucosal site. J. Immunol. 163(3), 1382–1389 (1999). •• The authors explored the structure–activity relationship
12 Neutra MR, Kozlowski PA. Mucosal vaccines: the promise based on different spital arrangement of epitopes.
and the challenge. Nat. Rev. Immunol. 6(2), 148–158 (2006).
27 Brito LA, O’hagan DT. Designing and building the next
13 Macpherson AJ, Mccoy KD, Johansen FE, Brandtzaeg P. The generation of improved vaccine adjuvants. J. Control. Rel. 190,
immune geography of IgA induction and function. Mucosal 563–579 (2014).
Immunol. 1(1), 11–22 (2008).
28 Pavot V, Berthet M, Resseguier J et al. Poly(lactic acid) and
•• Explains detailed mechanisms of immune inductions in poly(lactic-co-glycolic acid) particles as versatile carrier
mucosal tissues. platforms for vaccine delivery. Nanomedicine (Lond). 9(17),
14 Azmi F, Ahmad Fuaad AA, Skwarczynski M, Toth I. Recent 2703–2718 (2014).
progress in adjuvant discovery for peptide-based subunit 29 Reisch A, Runser A, Arntz Y, Mely Y, Klymchenko AS.
vaccines. Hum. Vaccin. Immunother. 10(3), 778–796 (2014). Charge-controlled nanoprecipitation as a modular approach
15 Kool M, Fierens K, Lambrecht BN. Alum adjuvant: some of to ultrasmall polymer nanocarriers: making bright and stable
the tricks of the oldest adjuvant. J. Med. Microbiol. 61(Pt 7), nanoparticles. ACS Nano 9(5), 5104–5116 (2015).
927–934 (2012). 30 Zhou J, Patel TR, Sirianni RW et al. Highly penetrative, drug-
16 Eriksson AM, Schon KM, Lycke NY. The cholera toxin- loaded nanocarriers improve treatment of glioblastoma. Proc.
derived CTA1-DD vaccine adjuvant administered intranasally Natl Acad. Sci. USA 110(29), 11751–11756 (2013).
does not cause inflammation or accumulate in the nervous 31 Primard C, Poecheim J, Heuking S, Sublet E, Esmaeili F,
tissues. J. Immunol. 173(5), 3310–3319 (2004). Borchard G. Multifunctional PLGA-based nanoparticles
17 Van Ginkel FW, Jackson RJ, Yuki Y, Mcghee JR. Cutting encapsulating simultaneously hydrophilic antigen and
edge: the mucosal adjuvant cholera toxin redirects vaccine hydrophobic immunomodulator for mucosal immunization.
proteins into olfactory tissues. J. Immunol. 165(9), 4778–4782 Mol. Pharm. 10(8), 2996–3004 (2013).
(2000). 32 Makadia HK, Siegel SJ. Poly lactic-co-glycolic acid (PLGA)
18 Shah RR, O’hagan DT, Amiji MM, Brito LA. The impact of as biodegradable controlled drug delivery carrier. Polymers
size on particulate vaccine adjuvants. Nanomedicine (Lond). (Basel). 3(3), 1377–1397 (2011).
9(17), 2671–2681 (2014). 33 Pawar D, Goyal AK, Mangal S et al. Evaluation of
19 Skwarczynski M, Toth I. Recent advances in peptide- mucoadhesive PLGA microparticles for nasal immunization.
based subunit nanovaccines. Nanomedicine (Lond). 9(17), AAPS J. 12(2), 130–137 (2010).
2657–2669 (2014). 34 Silva AL, Rosalia RA, Varypataki E, Sibuea S, Ossendorp F,
•• Comphrehesive review article on peptide-based Jiskoot W. Poly-(lactic-co-glycolic-acid)-based particulate
vaccines: particle uptake by dendritic cells is a key parameter
nanovaccines.
for immune activation. Vaccine 33(7), 847–854 (2015).
20 Zaman M, Chandrudu S, Giddam AK et al. Group A
35 Pawar D, Mangal S, Goswami R, Jaganathan KS.
Streptococcal vaccine candidate: contribution of epitope to
Development and characterization of surface modified PLGA
size, antigen presenting cell interaction and immunogenicity.
nanoparticles for nasal vaccine delivery: effect of mucoadhesive
Nanomedicine (Lond). 9(17), 2613–2624 (2014).
coating on antigen uptake and immune adjuvant activity. Eur.
21 Mora AL, Tam JP. Controlled lipidation and encapsulation J. Pharm. Biopharm. 85(3, Part A), 550–559 (2013).
of peptides as a useful approach to mucosal immunizations. J.
36 Fromen CA, Robbins GR, Shen TW, Kai MP, Ting JP,
Immunol. 161(7), 3616–3623 (1998).
Desimone JM. Controlled analysis of nanoparticle charge on
22 Batzloff MR, Hartas J, Zeng W, Jackson DC, Good MF. mucosal and systemic antibody responses following pulmonary
Intranasal vaccination with a lipopeptide containing a immunization. Proc. Natl Acad. Sci. USA 112(2), 488–493
conformationally constrained conserved minimal peptide, a (2015).
universal T cell epitope, and a self-adjuvanting lipid protects
37 Slütter B, Bal S, Keijzer C et al. Nasal vaccination with
mice from group A streptococcus challenge and reduces throat
N-trimethyl chitosan and PLGA based nanoparticles:
colonization. J. Infect. Dis. 194(3), 325–330 (2006).
nanoparticle characteristics determine quality and strength
23 Mcdonald DM, Byrne SN, Payne RJ. Synthetic self-adjuvanting of the antibody response in mice against the encapsulated
glycopeptide cancer vaccines. Front. Chem. 3 60 (2015). antigen. Vaccine 28(38), 6282–6291 (2010).
24 Chan A, Hussein WM, Ghaffar KA et al. Structure-activity 38 Rajapaksa TE, Bennett KM, Hamer M, Lytle C, Rodgers
relationship of lipid core peptide-based Group A Streptococcus VG, Lo DD. Intranasal M cell uptake of nanoparticles is
vaccine candidates. Bioorg. Med. Chem. 24(14), 3095–3101 independently influenced by targeting ligands and buffer
(2016). ionic strength. J. Biol. Chem. 285(31), 23739–23746 (2010).
25 Zaman M, Abdel-Aal AB, Fujita Y et al. Immunological 39 Vila A, Sanchez A, Evora C, Soriano I, Vila Jato JL, Alonso
evaluation of lipopeptide group A streptococcus (GAS) MJ. PEG-PLA nanoparticles as carriers for nasal vaccine
vaccine: structure-activity relationship. PLoS ONE 7(1), delivery. J. Aerosol. Med. 17(2), 174–185 (2004).
e30146 (2012).

164 Ther. Deliv. (2017) 8(3) future science group


Intranasal delivery of nanoparticle-based vaccines Review

40 Cu Y, Saltzman WM. Controlled surface modification chitosan nanospheres encapsulated with influenza whole
with Poly(ethylene)glycol enhances diffusion of PLGA virus and adjuvants. Int. J. Pharm. 475(1–2), 1–8 (2014).
nanoparticles in human cervical mucus. Mol. Pharm. 6(1), 55 Smith A, Perelman M, Hinchcliffe M. Chitosan: a promising
173–181 (2009). safe and immune-enhancing adjuvant for intranasal vaccines.
41 Rajapaksa TE, Stover-Hamer M, Fernandez X, Eckelhoefer Hum. Vaccin. Immunother. 10(3), 797–807 (2014).
HA, Lo DD. Claudin 4-targeted protein incorporated into 56 Bento D, Staats HF, Goncalves T, Borges O. Development
PLGA nanoparticles can mediate M cell targeted delivery. J. of a novel adjuvanted nasal vaccine: C48/80 associated
Control. Rel. 142(2), 196–205 (2010). with chitosan nanoparticles as a path to enhance mucosal
42 Dyawanapelly S, Koli U, Dharamdasani V, Jain R, Dandekar immunity. Eur. J. Pharm. Biopharm. 93, 149–164 (2015).
P. Improved mucoadhesion and cell uptake of chitosan 57 Smith A, Perelman M, Hinchcliffe M. Chitosan: a promising
and chitosan oligosaccharide surface-modified polymer safe and immune-enhancing adjuvant for intranasal vaccines.
nanoparticles for mucosal delivery of proteins. Drug Deliv. Hum. Vaccin. Immunother 10(3), 797–807 (2014).
Transl. Res. 6(4), 365–379 (2016).
58 Marasini N, Giddam AK, Ghaffar KA et al. Multilayer
43 Park J, Babensee JE. Differential functional effects of engineered nanoliposomes as a novel tool for oral delivery
biomaterials on dendritic cell maturation. Acta. Biomater. of lipopeptide-based vaccines against group A Streptococcus.
8(10), 3606–3617 (2012). Nanomedicine (Lond). 11(10), 1223–1236 (2016).
•• The authors demonstrated the role of different biomaterials 59 Subbiah R, Ramalingam P, Ramasundaram S et al. N,N,N-
for maturation of antigen-presenting cells. Trimethyl chitosan nanoparticles for controlled intranasal
44 Islam N, Ferro V. Recent advances in chitosan-based delivery of HBV surface antigen. Carbohydr. Polym. 89(4),
nanoparticulate pulmonary drug delivery. Nanoscale 8(30), 1289–1297 (2012).
14341–14358 (2016). 60 Mann AJ, Noulin N, Catchpole A et al. Intranasal H5N1
45 Kobayashi T, Fukushima K, Sannan T et al. Evaluation of the vaccines, adjuvanted with chitosan derivatives, protect
effectiveness and safety of chitosan derivatives as adjuvants for ferrets against highly pathogenic influenza intranasal and
intranasal vaccines. Viral Immunol. 26(2), 133–142 (2013). intratracheal challenge. PLoS ONE 9(5), e93761 (2014).
46 Arca HC, Gunbeyaz M, Senel S. Chitosan-based systems 61 Liu Q, Zhang C, Zheng X et al. Preparation and evaluation
for the delivery of vaccine antigens. Exp. Rev.Vaccines. 8(7), of antigen/N-trimethylaminoethylmethacrylate chitosan
937–953 (2009). conjugates for nasal immunization. Vaccine 32(22),
2582–2590 (2014).
47 Jabbal-Gill I, Watts P, Smith A. Chitosan-based delivery
systems for mucosal vaccines. Expert Opin. Drug Deliv. 9(9), 62 Liu Q, Zheng X, Zhang C et al. Conjugating influenza a
1051–1067 (2012). (H1N1) antigen to n-trimethylaminoethylmethacrylate
chitosan nanoparticles improves the immunogenicity of the
48 Benediktsdottir BE, Baldursson O, Masson M. Challenges
antigen after nasal administration. J. Med. Virol. 87(11),
in evaluation of chitosan and trimethylated chitosan
1807–1815 (2015).
(TMC) as mucosal permeation enhancers: From synthesis
to in vitro application. J. Control. Rel. 10(173), 18–31 63 Slutter B, Bal SM, Que I et al. Antigen-adjuvant
(2014). nanoconjugates for nasal vaccination: an improvement over
the use of nanoparticles? Mol. Pharm. 7(6), 2207–2215
49 Bernocchi B, Carpentier R, Lantier I, Ducournau C, Dimier-
(2010).
Poisson I, Betbeder D. Mechanisms allowing protein delivery
in nasal mucosa using NPL nanoparticles. J. Control. Rel. 64 Marasini N, Ghaffar KA, Giddam AK et al. Highly
232, 42–50 (2016). immunogenic trimethyl chitosan-based delivery system
for intranasal lipopeptide vaccines against Group A
50 Vila A, Sánchez A, Janes K et al. Low molecular weight
Streptococcus Curr. Drug Deliv. doi:10.2174/1567201813666
chitosan nanoparticles as new carriers for nasal vaccine
160721141322 (2016) (Epub ahead of print).
delivery in mice. Eur. J. Pharm. Biopharm. 57(1), 123–131
(2004). 65 Lee KY, Mooney DJ. Alginate: properties and biomedical
applications. Prog. Polym. Sci. 37(1), 106–126 (2012).
51 Ou J, Shi W, Xu Y, Tao Z. Intranasal immunization with
DNA vaccine coexpressing Der p 1 and ubiquitin in an 66 Borges O, Cordeiro-Da-Silva A, Tavares J et al. Immune
allergic rhinitis mouse model. Ann. Allergy Asthma Immunol. response by nasal delivery of hepatitis B surface antigen
113(6), 658–665.e651 (2014). and codelivery of a CpG ODN in alginate coated chitosan
nanoparticles. Eur. J. Pharm. Biopharm. 69(2), 405–416
52 Lebre F, Borchard G, Faneca H, Pedroso De Lima MC,
(2008).
Borges O. Intranasal administration of novel chitosan
nanoparticle/DNA complexes induces antibody response 67 Bento D, Staats HF, Gonçalves T, Borges O. Development
to hepatitis B surface antigen in mice. Mol. Pharm. 13(2), of a novel adjuvanted nasal vaccine: C48/80 associated
472–482 (2016). with chitosan nanoparticles as a path to enhance mucosal
immunity. Eur. J. Pharm. Biopharm. 93, 149–164 (2015).
53 Bento D, Staats HF, Borges O. Effect of particulate adjuvant
on the anthrax protective antigen dose required for effective 68 Shen L, Higuchi T, Tubbe I et al. A trifunctional dextran-
nasal vaccination. Vaccine 33(31), 3609–3613 (2015). based nanovaccine targets and activates murine dendritic
cells, and induces potent cellular and humoral immune
54 Dehghan S, Tafaghodi M, Bolourieh T et al. Rabbit nasal
responses. PLoS ONE 8(12), e80904 (2013).
immunization against influenza by dry-powder form of

future science group www.future-science.com 165


Review Marasini, Skwarczynski & Toth

69 Van Tomme SR, Hennink WE. Biodegradable dextran Eur. J. Pharm. Biopharm. 52(1), 21–30 (2001).
hydrogels for protein delivery applications. Expert Rev. Med. 84 Tamayo I, Irache JM, Mansilla C, Ochoa-Repáraz J, Lasarte
Devices 4(2), 147–164 (2007). JJ, Gamazo C. Poly(anhydride) nanoparticles act as Active
70 Sharma S, Benson HAE, Mukkur TKS, Rigby P, Chen Th1 adjuvants through Toll-like receptor exploitation. Clin.
Y. Preliminary studies on the development of IgA-loaded Vaccine Immunol. 17(9), 1356–1362 (2010).
chitosan-dextran sulphate nanoparticles as a potential nasal 85 Ojer P, De Cerain AL, Areses P, Peñuelas I, Irache JM.
delivery system for protein antigens. J. Microencapsul. 30(3), Toxicity studies of poly(anhydride) nanoparticles as carriers for
283–294 (2013). oral drug delivery. Pharm. Res. 29(9), 2615–2627 (2012).
71 Sajadi Tabassi SA, Tafaghodi M, Jaafari MR. Induction 86 Camacho AI, Irache JM, De Souza J, Sánchez-Gómez S,
of high antitoxin titers against tetanus toxoid in rabbits by Gamazo C. Nanoparticle-based vaccine for mucosal protection
intranasal immunization with dextran microspheres. Int. J. against Shigella flexneri in mice. Vaccine 31(32), 3288–3294
Pharm. 360(1–2), 12–17 (2008). (2013).
72 Tafaghodi M, Rastegar S. Preparation and in vivo study of dry 87 Zaman M, Skwarczynski M, Malcolm JM et al. Self-
powder microspheres for nasal immunization. J. Drug Target. adjuvanting polyacrylic nanoparticulate delivery system for
18(3), 235–242 (2010). group A streptococcus (GAS) vaccine. Nanomedicine 7(2),
73 Numata K. Poly(amino acid)s/polypeptides as potential 168–173 (2011).
functional and structural materials. Polym. J. 47(8), 537–545 88 Sahdev P, Ochyl LJ, Moon JJ. Biomaterials for nanoparticle
(2015). vaccine delivery systems. Pharm. Res. 31(10), 2563–2582
74 Trimaille T, Verrier B. Micelle-based adjuvants for subunit (2014).
vaccine delivery. Vaccines (Basel). 3(4), 803–813 (2015). 89 Hilgers LA, Ghenne L, Nicolas I, Fochesato M, Lejeune
75 Okamoto S, Matsuura M, Akagi T et al. Poly(gamma-glutamic G, Boon B. Alkyl-polyacrylate esters are strong mucosal
acid) nano-particles combined with mucosal influenza virus adjuvants. Vaccine 18(28), 3319–3325 (2000).
hemagglutinin vaccine protects against influenza virus infection 90 Li M, Zhao M, Fu Y et al. Enhanced intranasal delivery of
in mice. Vaccine 27(42), 5896–5905 (2009). mRNA vaccine by overcoming the nasal epithelial barrier via
76 Schneerson R, Kubler-Kielb J, Liu T-Y et al. Poly(γ-d-glutamic intra- and paracellular pathways. J. Control. Rel. 228, 9–19
acid) protein conjugates induce IgG antibodies in mice to (2016).
the capsule of Bacillus anthracis: A potential addition to 91 Chang H-I, Yeh M-K. Clinical development of liposome-based
the anthrax vaccine. Proc. Natl Acad. Sci. USA 100(15), drugs: formulation, characterization, and therapeutic efficacy.
8945–8950 (2003). Int. J. Nanomedicine. 7, 49–60 (2012).
77 Matsuo K, Koizumi H, Akashi M et al. Intranasal 92 Marasini N, Ghaffar KA, Skwarczynski M, Toth I. Liposomes
immunization with poly(γ-glutamic acid) nanoparticles as a vaccine delivery system. In Micro and Nanotechnologies for
entrapping antigenic proteins can induce potent tumor Vaccine Development.Skwarczynski M, Toth I (Eds). Elsevier
immunity. J. Control. Rel. 152(2), 310–316 (2011). Inc., Burlington, MA, USA, 221–239 (2016).
78 Mann A, Richa R, Ganguli M. DNA condensation by poly-l- 93 Ghaffar KA, Giddam AK, Zaman M, Skwarczynski M, Toth
lysine at the single molecule level: role of DNA concentration I. Liposomes as nanovaccine delivery systems. Curr. Top. Med.
and polymer length. J. Control. Rel. 125(3), 252–262 (2008). Chem. 14(9), 1194–1208 (2014).
79 Wu Y, Wang X, Csencsits KL, Haddad A, Walters N, Pascual 94 Tada R, Hidaka A, Iwase N et al. Intranasal immunization
DW. M cell-targeted DNA vaccination. Proc. Natl Acad. Sci. with DOTAP cationic liposomes combined with DC-
USA 98(16), 9318–9323 (2001). cholesterol induces potent antigen-specific mucosal and
80 Wedemeyer H, Schuller E, Schlaphoff V et al. Therapeutic systemic immune responses in mice. PLoS ONE 10(10),
vaccine IC41 as late add-on to standard treatment in patients e0139785 (2015).
with chronic hepatitis C. Vaccine 27(37), 5142–5151 (2009). 95 Slütter B, Bal SM, Ding Z, Jiskoot W, Bouwstra JA.
81 De Koker S, De Geest BG, Singh SK et al. Polyelectrolyte Adjuvant effect of cationic liposomes and CpG depends on
microcapsules as antigen delivery vehicles to dendritic cells: administration route. J. Control. Rel. 154(2), 123–130 (2011).
uptake, processing, and cross-presentation of encapsulated 96 Ghaffar KA, Marasini N, Giddam AK et al. Liposome-based
antigens. Angew. Chem. Int. Ed. Engl. 48(45), 8485–8489 intranasal delivery of lipopeptide vaccine candidates against
(2009). group A streptococcus. Acta Biomater. 41, 161–168 (2016).
•• The authors explored use of polyelectrolyte systems for •• Showed synergistic effect of liposomes and lipopeptide-based
vaccines delivery. vaccines in enhancing mucosal and systemic immunity.
82 De Koker S, Naessens T, De Geest BG et al. Biodegradable 97 Ghaffar KA, Marasini N, Giddam AK et al. The role of size
polyelectrolyte microcapsules: antigen delivery tools with in development of mucosal liposome-lipopeptide vaccine
Th17 skewing activity after pulmonary delivery. J. Immunol. candidates against Group A Streptococcus. Med. Chem. (13(1),
184(1), 203–211 (2010). 22–27, (2016).
83 Miyamoto M, Natsume H, Iwata S et al. Improved nasal 98 Jiang P-L, Lin H-J, Wang H-W et al. Galactosylated liposome
absorption of drugs using poly-l-arginine: effects of as a dendritic cell-targeted mucosal vaccine for inducing
concentration and molecular weight of poly-l-arginine on the protective anti-tumor immunity. Acta Biomater. 11, 356–367
nasal absorption of fluorescein isothiocyanate-dextran in rats. (2015).

166 Ther. Deliv. (2017) 8(3) future science group


Intranasal delivery of nanoparticle-based vaccines Review

99 Wang H-W, Jiang P-L, Lin S-F et al. Application of galactose- 115 Phillipps KS, Wykes MN, Liu XQ, Brown M, Blanchfield
modified liposomes as a potent antigen presenting cell targeted J, Toth I. A novel synthetic adjuvant enhances dendritic cell
carrier for intranasal immunization. Acta Biomater. 9(3), function. Immunology 128(1 Suppl.), e582–e588 (2009).
5681–5688 (2013). 116 Zeng W, Ghosh S, Lau YF, Brown LE, Jackson DC. Highly
100 Fan Y, Sahdev P, Ochyl LJ, J Akerberg J, Moon JJ. Cationic immunogenic and totally synthetic lipopeptides as self-
liposome-hyaluronic acid hybrid nanoparticles for intranasal adjuvanting immunocontraceptive vaccines. J. Immunol.
vaccination with subunit antigens. J. Control. Rel. 208, 169(9), 4905–4912 (2002).
121–129 (2015). 117 Tan AC, Mifsud EJ, Zeng W et al. Intranasal administration
101 Marasini N, Yan YD, Poudel BK, Choi HG, Yong CS, Kim of the TLR2 agonist Pam2Cys provides rapid protection
JO. Development and optimization of self-nanoemulsifying against influenza in mice. Mol. Pharm. 9(9), 2710–2718
drug delivery system with enhanced bioavailability by box- (2012).
behnken design and desirability function. J. Pharm. Sci. 118 Skwarczynski M, Parhiz BH, Soltani F et al. Lipid peptide
101(12), 4584–4596 (2012). core nanoparticles as multivalent vaccine candidates against
102 Fox CB. Squalene emulsions for parenteral vaccine and drug Streptococcus pyogenes. Aust. J. Chem. 65(1), 35–39 (2012).
delivery. Molecules 14(9), 3286–3312 (2009). 119 Hamley IW. Lipopeptides: from self-assembly to bioactivity.
103 Makidon PE, Belyakov IM, Blanco LP et al. Nanoemulsion Chem. Commun (Camb). 51(41), 8574–8583 (2015).
mucosal adjuvant uniquely activates cytokine production by 120 Skwarczynski M, Toth I. Lipid-core-peptide system for self-
nasal ciliated epithelium and induces dendritic cell trafficking. adjuvanting synthetic vaccine delivery. Methods Mol. Biol. 751,
Eur. J. Immunol. 42(8), 2073–2086 (2012). 297–308 (2011).
104 Makidon PE, Bielinska AU, Nigavekar SS et al. Pre-clinical 121 Skwarczynski M, Zaman M, Toth I. Chapter 78 – Lipo-
evaluation of a novel nanoemulsion-based hepatitis B mucosal peptides/saccharides for peptide vaccine delivery A2. In:
vaccine. PLoS ONE 3(8), e2954 (2008). Handbook of Biologically Active Peptides (2nd Edition). Kastin
105 Wong PT, Leroueil PR, Smith DM et al. Formulation, AJ (Ed.). Academic Press, Boston, MA, USA, 571–579 (2013).
high throughput in vitro screening and in vivo functional 122 Abdel-Aal AB, Zaman M, Fujita Y, Batzloff MR, Good MF,
characterization of nanoemulsion-based intranasal vaccine Toth I. Design of three-component vaccines against group A
adjuvants. PLoS ONE 10(5), e0126120 (2015). streptococcal infections: importance of spatial arrangement
106 Wong PT, Wang SH, Ciotti S et al. Formulation and of vaccine components. J. Med. Chem. 53(22), 8041–8046
characterization of nanoemulsion intranasal adjuvants: (2010).
effects of surfactant composition on mucoadhesion and 123 Marasini N, Khalil ZG, Giddam AK et al. Lipid core peptide/
immunogenicity. Mol. Pharm. 11(2), 531–544 (2014). poly(lactic-co-glycolic acid) as a highly potent intranasal
107 Sanders MT, Brown LE, Deliyannis G, Pearse MJ. vaccine delivery system against Group A streptococcus. Int. J.
ISCOMTM-based vaccines: the second decade. Immunol. Cell Pharm. 513(1–2), 410–420 (2016).
Biol. 83(2), 119–128 (2005). 124 Benne N, Van Duijn J, Kuiper J, Jiskoot W, Slutter B.
108 Hu K-F, Lövgren-Bengtsson K, Morein B. Immunostimulating Orchestrating immune responses: how size, shape and rigidity
complexes (ISCOMs) for nasal vaccination. Adv. Drug Deliv. affect the immunogenicity of particulate vaccines. J. Control.
Rev. 51(1–3), 149–159 (2001). Rel. 234, 124–134 (2016).
109 Pandey RS, Dixit VK. Evaluation of ISCOM vaccines for 125 Oyewumi MO, Kumar A, Cui Z. Nano-microparticles as
mucosal immunization against hepatitis B. J. Drug Target. immune adjuvants: correlating particle sizes and the resultant
18(4), 282–291 (2010). immune responses. Expert Rev. Vaccines. 9(9), 1095–1107
110 Sjolander A, Cox JC, Barr IG. ISCOMs: an adjuvant with (2010).
multiple functions. J. Leukoc. Biol. 64(6), 713–723 (1998). 126 Vila A, Sánchez A, Évora C, Soriano I, Mccallion O, Alonso
111 Cibulski SP, Mourglia-Ettlin G, Teixeira TF et al. Novel MJ. PLA-PEG particles as nasal protein carriers: the influence
ISCOMs from Quillaja brasiliensis saponins induce mucosal of the particle size. Int. J. Pharm. 292(1–2), 43–52 (2005).
and systemic antibody production, T-cell responses and 127 Gutierro I, Hernández RM, Igartua M, Gascón AR, Pedraz
improved antigen uptake. Vaccine 34(9), 1162–1171 (2016). JL. Size dependent immune response after subcutaneous, oral
112 De Costa F, Yendo ACA, Cibulski SP et al. Alternative and intranasal administration of BSA loaded nanospheres.
inactivated poliovirus vaccines adjuvanted with Quillaja Vaccine 21(1–2), 67–77 (2002).
brasiliensis or Quil-a saponins are equally effective in inducing 128 Stano A, Nembrini C, Swartz MA, Hubbell JA, Simeoni E.
specific immune responses. PLoS ONE 9(8), e105374 (2014). Nanoparticle size influences the magnitude and quality of
113 Pandey RS, Babbar AK, Kaul A, Mishra AK, Dixit VK. mucosal immune responses after intranasal immunization.
Evaluation of ISCOM matrices clearance from rabbit nasal Vaccine 30(52), 7541–7546 (2012).
cavity by gamma scintigraphy. Int. J. Pharm. 398(1–2), 129 Lai SK, Wang Y-Y, Hanes J. Mucus-penetrating nanoparticles
231–236 (2010). for drug and gene delivery to mucosal tissues. Adv. Drug
114 Benmohamed L, Wechsler SL, Nesburn AB. Lipopeptide Deliv. Rev. 61(2), 158–171 (2009).
vaccines – yesterday, today, and tomorrow. Lancet Infect. Dis. 130 US National Institute of Health. (2016).
2(7), 425–431 (2002). https://clinicaltrials.gov

future science group www.future-science.com 167

You might also like