Sei sulla pagina 1di 11

Vaccine 32 (2014) 327337

Contents lists available at ScienceDirect

Vaccine
journal homepage: www.elsevier.com/locate/vaccine

Review

Nanoparticle vaccines
Liang Zhao a,1 , Arjun Seth a,1 , Nani Wibowo a , Chun-Xia Zhao a , Neena Mitter b ,
Chengzhong Yu a , Anton P.J. Middelberg a,
a
The University of Queensland, Australian Institute for Bioengineering and Nanotechnology, St. Lucia QLD 4072, Australia
b
The University of Queensland, Queensland Alliance for Agriculture and Food Innovation, St. Lucia QLD 4072, Australia

a r t i c l e i n f o a b s t r a c t

Article history: Nanotechnology increasingly plays a signicant role in vaccine development. As vaccine development
Received 29 August 2013 orientates toward less immunogenic minimalist compositions, formulations that boost antigen effec-
Received in revised form tiveness are increasingly needed. The use of nanoparticles in vaccine formulations allows not only
11 November 2013
improved antigen stability and immunogenicity, but also targeted delivery and slow release. A number
Accepted 18 November 2013
of nanoparticle vaccines varying in composition, size, shape, and surface properties have been approved
Available online 2 December 2013
for human use and the number of candidates is increasing. However, challenges remain due to a lack
of fundamental understanding regarding the in vivo behavior of nanoparticles, which can operate as
Keywords:
Vaccine
either a delivery system to enhance antigen processing and/or as an immunostimulant adjuvant to acti-
Nanoparticle vate or enhance immunity. This review provides a broad overview of recent advances in prophylactic
Nanotechnology nanovaccinology. Types of nanoparticles used are outlined and their interaction with immune cells and
Adjuvant the biosystem are discussed. Increased knowledge and fundamental understanding of nanoparticle mech-
Nanovaccinology anism of action in both immunostimulatory and delivery modes, and better understanding of in vivo
biodistribution and fate, are urgently required, and will accelerate the rational design of nanoparticle-
containing vaccines.
2013 The Authors. Published by Elsevier Ltd. All rights reserved.

1. Introduction using the cellular endocytosis mechanism, in particular pinocy-


tosis [6]. These cutting-edge innovations underpinned a market
Vaccine development has a proud history as one of the most worth US $6.8 billion in 2006 [7] and predicted to reach US $160
successful public health interventions to date. Vaccine develop- billion by 2015 [8]. Indeed, nanoparticles are revolutionizing the
ment is historically based on Louis Pasteurs isolate, inactivate, diagnosis of diseases as well as the delivery of biologically-active
inject paradigm. As vaccine development moves increasingly compounds for disease prevention and treatment. The emergence
to draw on modern concepts of rational design, the number of of virus-like particles (VLPs) and the resurgence of nanoparticles,
candidate vaccines is increasing [1,2]. Most candidate vaccines rep- such as quantum dots and magnetic nanoparticles, marks a conver-
resent minimalist compositions [3], which typically exhibit lower gence of protein biotechnology with inorganic nanotechnology that
immunogenicity. Adjuvants and novel delivery systems that boost promises an era of signicant progress for nanomedicine [9,10]. A
immunogenicity are increasingly needed as we move toward the number of approved nano-sized vaccine and drug delivery systems
era of modern vaccines. highlight the revolution in disease prevention and treatment that
Nanotechnology offers the opportunity to design nanoparticles is occurring [4,1113].
varying in composition, size, shape, and surface properties, for The use of nanotechnology in vaccinology, in particular, has
application in the eld of medicine [4,5]. Nanoparticles, because of been increasing exponentially in the past decade (Fig. 1), lead-
their size similarity to cellular components, can enter living cells ing to the birth of nanovaccinology [3]. In both prophylactic
and therapeutic approaches, nanoparticles are used as either
a delivery system to enhance antigen processing and/or as an
immunostimulant adjuvant to activate or enhance immunity. Ther-
This is an open-access article distributed under the terms of the Creative Com- apeutic nanovaccinology is mostly applied for cancer treatment
mons Attribution-NonCommercial-No Derivative Works License, which permits [1416], and is increasingly explored to treat other diseases or
non-commercial use, distribution, and reproduction in any medium, provided the conditions, such as Alzheimers [17], hypertension [9], and nico-
original author and source are credited.
Corresponding author. Tel.: +61 7 3346 4189; fax: +61 7 3346 4197. tine addiction [11]. Prophylactic nanovaccinology, on the other
E-mail address: a.middelberg@uq.edu.au (A.P.J. Middelberg). hand, has been applied for the prevention of different diseases.
1
These authors contributed equally. A number of prophylactic nanovaccines have been approved

0264-410X/$ see front matter 2013 The Authors. Published by Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.vaccine.2013.11.069
328 L. Zhao et al. / Vaccine 32 (2014) 327337

[37], hepatitis B virus (HBV) [22], Bacillus anthracis [29], and model
antigens such as ovalbumin and tetanus toxoid [26,27]. g-PGA
nanoparticles are comprised of amphiphilic poly(amino acid)s,
which self-assemble into nano-micelles with a hydrophilic outer
shell and a hydrophobic inner core [31,32]. g-PGA nanoparticles
are generally used to encapsulate hydrophobic antigen [31,32].
Polystyrene nanoparticles can conjugate to a variety of antigens
as they can be surface-modied with various functional groups
[33,38].
Natural polymers based on polysaccharide have also been
used to prepare nanoparticle adjuvants, such as pullulan [39,40],
alginate [41], inulin [42,43], and chitosan [4449]. In particular,
chitosan-based nanoparticles have been widely studied due to their
biocompatibility, biodegradability, nontoxic nature and their abil-
ity to be easily modied into desired shapes and sizes [31,50,51].
These nanoparticles have been used in the preparation of various
Fig. 1. Number of publications returned using the search terms nanoparticle* vaccines including HBV vaccines [49], Newcastle disease vaccines
and vaccin* from Web of Science (http://apps.webofknowledge.com/; results for [48], and DNA vaccines [44,46,47]. Inulin, a well-known activator of
a search conducted on 29 July 2013). complement via the alternative pathway [52], is also a potent adju-
vant. Nanoparticle adjuvants derived from inulin, such as AdvaxTM ,
for human use and more are in clinical or pre-clinical trials have shown enhancement of immune response in vaccines against
[13,1820]. various viruses including inuenza [42] and hepatitis B [43].
In this review, we provide an overview of recent advances in Polymers, such as Poly(L-lactic acid) (PLA), PLGA, PEG, and
the broad area of nanovaccinology, but limit our review only to natural polymers such as polysaccharides [41,5355], have also
prophylactic vaccines. We rst survey advances in the types of been used to synthesize hydrogel nanoparticles, which are a type
nanoparticles, which are dened as any particulate material with of nano-sized hydrophilic three-dimensional polymer network.
size 11000 nm [21], used for prophylactic vaccine design (Fig. 2). Nanogels have favorable properties including exible mesh size,
We then discuss the interaction of nanoparticles with the antigen large surface area for multivalent conjugation, high water content,
of interest, differentiating the role of the nanoparticle as either and high loading capacity for antigens [55,56]. Chitosan nanogels
delivery system and/or immunostimulant adjuvant. The interac- have been widely used in antigen delivery, such as Clostridium
tion of nanoparticles with immune cells and the biosystem are botulinum type-A neurotoxin subunit antigen Hc for an adjuvant-
also discussed to provide understanding of antigen and nanopar- free intranasal vaccine [57], and recombinant NcPDI antigen for
ticle processing in vivo, as well as clearance. This latter aspect is Neospora caninum vaccination [58].
of particular timeliness considering that there is limited history of
safe use for non-VLP nanoparticles in humans. We then conclude 2.2. Inorganic nanoparticles
with remarks about the further potential and future prospects for
prophylactic nanovaccinology. Many inorganic nanoparticles have been studied for their
use in vaccines. Although these nanoparticles are mostly non-
2. Types of nanoparticles biodegradable, the advantage of them lies in their rigid structure
and controllable synthesis [33]. Gold nanoparticles (AuNPs) are
2.1. Polymeric nanoparticles used in vaccine delivery [35], as they can be easily fabricated into
different shapes (spherical, rod, cubic, etc.) [59] with a size range of
A great variety of synthetic polymers are used to prepare 2150 nm [60], and can be surface-modied with carbohydrates
nanoparticles, such as poly(d,l-lactide-co-glycolide) (PLG) [2224], [61]. Gold nanorods have been used as a carrier for an antigen
poly(d,l-lactic-coglycolic acid)(PLGA) [22,2530], poly(g-glutamic derived from respiratory syncytial virus by conjugating the antigen
acid) (g-PGA) [31,32], poly(ethylene glycol) (PEG) [24], and to the surface [62]. Other types of gold nanoparticles have been used
polystyrene [33,34]. PLG and PLGA nanoparticles have been the as carriers for antigens derived from other viruses such as inuenza
most extensively investigated due to their excellent biocompati- [63] and foot-and-mouth disease [64], or as a DNA vaccine adjuvant
bility and biodegradability [35,36]. These polymeric nanoparticles for human immunodeciency virus (HIV) [65].
entrap antigen for delivery to certain cells or sustain antigen release Carbon nanoparticles are another commonly-studied compo-
by virtue of their slow biodegradation rate [2729,31,36]. PLGA has sition for drug and vaccine delivery [60]. They are known for
been used to carry antigen derived from various pathogens includ- their good biocompatibility and can be synthesized into a vari-
ing Plasmodium vivax with mono-phosphoryl lipid A as adjuvant ety of nanotubes and mesoporous spheres [6668]. The diameter

Fig. 2. The size range of nanoparticles used in nanovaccinology.


L. Zhao et al. / Vaccine 32 (2014) 327337 329

of carbon nanotubes (CNTs) used as carriers is generally 0.82 nm protein antigen [35,92,99101]. These spherical particles can trap
with a length of 1001000 nm [69,70], while the size of mesoporous the antigen by apolar interactions [35]. ISCOMATRIX comprises
carbon spheres is around 500 nm [67]. Multiple copies of protein ISCOMs without antigen [35,92,100,102]. ISCOMATRIX can be
and peptide antigens can be conjugated on to CNTs for delivery and mixed with antigen, enabling a more exible application than is
have enhanced the level of IgG response [67,6971]. Mesoporous possible for ISCOMs, by removing the limitation of hydrophobic
carbon nanoparticles have been studied for application as an oral antigens [35]. Various antigens have been used to form ISCOMs,
vaccine adjuvant [67]. including antigens derived from inuenza [103,104], herpes
One of the most promising inorganic materials for nanovaccinol- simplex virus [105], HIV [106], and Newcastle disease [99].
ogy and delivery system design is silica. Silica-based nanoparticles
(SiNPs) are biocompatible and have excellent properties as 2.5. Virus-like particles
nanocarriers for various applications, such as selective tumor
targeting [72], real-time multimodal imaging [73], and vaccine Virus-like particles (VLP) are self-assembling nanoparticles,
delivery. The SiNPs can be prepared with tunable structural param- lacking infectious nucleic acid, formed by self-assembly of biocom-
eters. By controlling the solgel chemistry, the particle size and patible capsid proteins [107,108]. VLPs are the ideal nanovaccine
shape of SiNPs can be adjusted to selectively alter their inter- system as they harness the power of evolved viral structure, which
action with cells [74]. The abundant surface silanol groups are is naturally optimized for interaction with the immune system, but
benecial for further modication to introduce additional function- avoid the infectious components. VLPs take the good aspects of
ality, such as cell recognition, absorption of specic biomolecules, viruses and avoid the bad. The naturally-optimized nanoparticle
improvement of interaction with cells, and enhancement of cellu- size and repetitive structural order means that VLPs induce potent
lar uptake [7578]. In addition, porous SiNPs such as mesoporous immune responses, even in the absence of adjuvant [109]. VLP
silica nanoparticles (MSNs) and hollow SiNPs can be prepared by based vaccines are the rst nanoparticle class to reach market the
templating methods, which can be applied as a multifunctional rst VLP vaccine for hepatitis B virus was commercialized in 1986
platform to simultaneously deliver cargo molecules with various [110] and have become widely administered in healthy popu-
molecular weights [74]. MSNs with sizes in the range of 50200 nm lations. In nanovaccinology, VLP nanoparticles have the strongest
have been studied as both nano-carriers and adjuvants for delivery evidence base for safe use in healthy humans. Newer VLP vaccines
of effective antigens [7981], such as those derived from porcine for human papillomavirus [111] and hepatitis E [112] have been
circovirus [82] and HIV [83]. MSNs can be used to control the release approved for use in humans in 2006 and 2011, respectively.
of antigens by controlling the shape, pore size and surface func- VLPs can be derived from a variety of viruses (Fig. 3) [107],
tionalization [79,84]. Compared to solid SiNPs, MSNs have higher with sizes ranging from 20 nm to 800 nm [13,113], and can be
loading capacity for their larger specic surface area, and better manufactured with a variety of process technologies [114]. The
performance in delivery and controlled release due to the tun- historical approach to VLP manufacture involves an in vivo route,
able hollow and mesoporous structure. In addition, MSNs can be where the assembly of capsid proteins into VLPs occurs inside
degraded which can then be excreted in the urine [8587]. With the expression host. The assembled particle is then puried away
these properties, MSNs show potential to become high-efciency, from adherent and encapsulated contaminants. In some cases it
controlled-release nano-carriers in future vaccine formulations. becomes necessary to disassemble and then re-assemble the VLP
Calcium phosphate nanoparticles can be created by mixing cal- to improve quality [114]; recently-approved VLP vaccines typi-
cium chloride, dibasic sodium phosphate and sodium citrate under cally include some aspect of extracellular assembly within the
specic conditions [88,89]. They are non-toxic and can be formed processing regime. An emerging approach for VLP assembly is
into a size of 50100 nm [90]. These nanoparticles are useful adju- through cell-free in vitro processing [115119]. This approach
vants for DNA vaccines and mucosal immunity [79,8890], and inverts the traditional assemble-then-purify paradigm; large-scale
show excellent biocompatibility. purication of the VLP building blocks from contaminants occurs
rst, then these are assembled in vitro, avoiding the need to disas-
2.3. Liposomes semble VLP structures after assembly in a cell. Further review of VLP
manufacturing approaches is available elsewhere [13,19,120,121].
Liposomes are formed by biodegradable and nontoxic phos- VLPs commercialized to date are based on self-assembly of pro-
pholipids. Liposomes can encapsulate antigen within the core teins derived from the target virus. However, VLPs can also act as a
for delivery [91] and incorporate viral envelope glycoproteins to delivery platform where a target antigen from a virus unrelated to
form virosomes [92,93] including for inuenza [94]. Combination the VLP used is modularized on the surface of a VLP [20,122125].
of 1,2-dioleoyl-3-trimethylammonium propane (DOTAP) modied These modular VLPs exploit known benets of VLPs (optimized
cationic liposome and a cationic polymer (usually protamine) con- particle size and molecular structure) to target disease in an engi-
densed DNA are called liposome-polycation-DNA nanoparticles neered fashion. With many VLP vaccines currently in clinical or
(LPD), a commonly used adjuvant delivery system in DNA vaccine pre-clinical trials [13,19], an increase in the number of approved
studies [95,96]. The components of LPD spontaneously rearrange VLP-based vaccines can be expected.
into a nano-structure around 150 nm in size with condensed
DNA located inside the liposome [96]. Liposomes modied with 2.6. Self-assembled proteins
maleimide can be synthesized into interbilayer-crosslinked multi-
lamellar vesicles (ICMVs) by cation driven fusion and crosslinking Recognizing the power of the VLP approach, self-assembling
[97] enabling slowed release of entrapped antigen. A number of systems that attempt to drive higher levels of protein quaternary
liposome systems have been established and approved for human structuring have emerged for the preparation of nanoparticle-
use, such as Inexal V and Epaxal , which have been discussed in based vaccines. Ferritin is a protein that can self-assemble into
other reviews [91,98]. nearly-spherical 10 nm structure [126]. By genetically fusing
inuenza virus haemagglutinin (HA) to ferritin, the recombined
2.4. Immunostimulating complex (ISCOM) protein spontaneously assembled into an octahedrally-symmetric
particle and reformed 8 trimeric HA spikes [126] to give a higher
ISCOMs are cage like particles about 40 nm large in size, made immune response than trivalent inactivated inuenza vaccine,
of the saponin adjuvant Quil A, cholesterol, phospholipids, and which typically is processed to destroy rather than build viral
330 L. Zhao et al. / Vaccine 32 (2014) 327337

Fig. 3. Structure of virus-like particles. Virus-like particles can be derived from a variety of viruses. HEV, hepatitis E virus; HPV, human papillomavirus 16; SIV-HIV, hybrid
VLP between simian immunodeciency virus gag and human immunodeciency virus env; HCV, hepatitis C virus; BTV, bluetongue virus.
Reprinted from Trends in Microbiology, Vol. 11, Issue 8, Rob Noad and Polly Roy, Virus-like particles as immunogens, Pages 438444, Copyright (2003), with permission from
Elsevier [107].

structure. This example highlights the importance of driving 2.7. Emulsions


higher-order molecular structure in modern vaccines. The major
vault protein (MVP) is another kind of self-assembling protein. Another type of nanoparticles used as adjuvants in vaccines
Ninety-six units of MVP can self-assemble into a barrel-shaped delivery is nano-sized emulsions [100,128,129]. These nanopar-
vault nanoparticle, with a size of approximately 40 nm wide and ticles can exist as oil-in-water or water-in-oil forms, where the
70 nm long [127]. Antigens that are genetically fused with a mini- droplet size can vary from 50 nm to 600 nm [128]. Emulsions can
mal interaction domain can be packaged inside vault nanoparticles carry antigens inside their core for efcient vaccine delivery [128]
by self-assembling process when mixed with MVPs [127]. Vault or can also be simply mixed with the antigen. One commonly-
nanoparticles have been used to encapsulate the major outer mem- used emulsion is MF59TM , an oil-in-water emulsion which has been
brane protein of Chlamydia muridarum for studies of mucosal licensed as a safe and potent vaccine adjuvant in over 20 countries
immunity [127]. [35,130]. It has been widely studied for use in inuenza vaccines
L. Zhao et al. / Vaccine 32 (2014) 327337 331

Fig. 4. Schematic representation of PEG (white) chemically conjugated to DAMP4 protein (dark blue) being introduced to a solution containing pre-formed nanoemulsion
oil core (light yellow) stabilized by AM1 peptide (red), in aqueous buffer (light blue background). DAMP4 protein, which is chemically similar to AM1 peptide, is able to
integrate into the oil-water interface formed between the core and the aqueous bulk. Prior conjugation of PEG to DAMP4 leads to its functional display at the interface through
non-covalent molecular self-assembly. (For interpretation of the references to color in gure legend, the reader is referred to the web version of the article.)
Reprinted from Small, http://dx.doi.org/10.1002/smll.201300078, B.J. Zeng, Y.P. Chuan, B. OSullivan, I. Caminschi, M.H. Lahoud, R. Thomas, A.P.J. Middelberg, Receptor-Specic
Delivery of Protein Antigen to Dendritic Cells by a Nanoemulsion Formed Using Top-Down Non-Covalent Click Self-Assembly, Copyright (2003), with permission from John
Wiley and Sons [138].

[130132]. Another is MontanideTM , a large family of both oil-in- typically necessary. For immune potentiator approaches, nanopar-
water and water-in-oil emulsions, including ISA 50 V, 51, 201, 206 ticles activate certain immune pathways which might then enhance
and 720 [35,133]. Montanide ISA 51 and 720 have been used in antigen processing and improve immunogenicity.
Malaria vaccines [134,135], Montanide ISA 201 and 206 have been Hard material nanoparticles, such as those based on silica, gold,
used in foot-and-mouth disease vaccines [136]. and calcium phosphate, have predominantly been examined for
Recently, a tailorable nano-sized emulsion (TNE) platform tech- use as a delivery system [139] and have thus been engineered
nology has been developed using non-covalent click self-assembly to promote antigen attachment. Attachment of antigen has been
for antigen and drug delivery [137,138]. An oil-in-water nanoemul- achieved through simple physical adsorption or more complex
sion is formed using designed biosurfactant peptides and proteins. methods, such as chemical conjugation or encapsulation (Fig. 5).
Using a self-assembling peptide-protein system, immune-evading Adsorption of antigen onto a nanoparticle is generally based simply
PEG and a receptor-specic antibody can be arrayed in a selectively on charge or hydrophobic interaction [79,140,141]. Therefore, the
proportioned fashion on the aqueous interface of a nano-sized oil- interaction between nanoparticle and antigen is relatively weak,
in-water emulsion (Fig. 4). Targeted delivery of protein antigen to which may lead to rapid disassociation of antigen and nanopar-
dendritic cells was achieved [138]. This work demonstrates a new ticle in vivo. Encapsulation and chemical conjugation provide for
and simple way to make biocompatible designer nanoemulsions stronger interaction between nanoparticle and antigen. In encap-
using non-covalent click self-assembly by sequential top-down sulation, antigens are mixed with nanoparticle precursors during
reagent addition. synthesis, resulting in encapsulation of antigen when the precur-
sors particulate into a nanoparticle [88]. Antigen is released only
when the nanoparticle has been decomposed in vivo or inside
3. Nanoparticle interaction with antigen the cell. On the other hand, for chemical conjugation, antigen is
chemically cross-linked to the surface of a nanoparticle [142]. Anti-
Vaccine formulations comprising nanoparticles and antigens gen is taken up by the cell together with the nanoparticle and
can be classied by nanoparticle action into those based on delivery is then released inside the cell. In soft matter nanoparticle deliv-
system or immune potentiator approaches. As a delivery system, ery system, such as those based on VLPs, ISCOM, ISCOMATRIXTM ,
nanoparticles can deliver antigen to the cells of the immune system, or liposomes, attachment of antigen is achieved through chemi-
i.e. the antigen and nanoparticle are co-ingested by the immune cell, cal conjugation, adsorption, encapsulation, or fusion at DNA level
or act as a transient delivery system, i.e. protect the antigen and [91,94,101,102,123125].
then release it at the target location [79]. For nanoparticles to func- For nanoparticles to act as an immune potentiator, attach-
tion as a delivery system, association of antigen and nanoparticle is ment or interaction between the nanoparticle and antigen is not
332 L. Zhao et al. / Vaccine 32 (2014) 327337

Fig. 5. Interaction of nanoparticle with antigen of interest. Formulation of nanoparticle and antigen of interest can be through attachment (e.g. conjugation, encapsulation,
or adsorption) or simple mixing.

necessary, and may be undesirable in cases where modication smaller than 500 nm [163]. Similarly, 300 nm sized PLGA particles
of antigenic structure occurs at the nanoparticle interface. Soft- also showed higher internalization and activation of DCs in com-
matter nanoparticles, such as emulsion-based adjuvants MF59TM parison to 17, 7 and 1 m particles [164]. Higher uptake of smaller
and AS03TM , have been shown to adjuvant a target antigen even PLA particles (200600 nm) in comparison to larger ones (28 m)
when they are injected independently of, and before, the antigen has also been reported for uptake by macrophages [165]. Differ-
[143,144]. Building on this idea, formulation of immune potentia- ent studies however, show discrepancies in optimum nanoparticle
tor nanoparticles with a target antigen could be possible through vaccine size. Amphiphilic poly(amino acid) (PAA) nanoparticles of
simple mixing of nanoparticle and adjuvant, shortly prior to injec- 30 nm were shown to have a lower DC uptake than that of 200 nm
tion, with minimal association between nanoparticle and antigen nanoparticles [166]. Polyacrylamide hydrogel particles of 35 nm
needed. This approach has only recently been investigated for and 3.5 m in size showed no difference in macrophages uptake
hard-material nanoparticle adjuvants, with results suggesting that [167]. These discrepancies may be related to the intrinsic differ-
nanoparticles may act as a size-dependent immune potentiator ences in the material properties, with each material having an
adjuvant even when not conjugated to the antigen [145]. This new optimum size for induction of potent immune response [168].
nding is consistent with a number of other studies that have In addition to particle size, surface charge also plays a signicant
demonstrated induction of inammatory immune responses after role in the activation of immune response. Cationic nanoparticles
injection of hard material nanoparticles alone and without antigen have been shown to induce higher APC uptake due to electrostatic
[146,147]. Further studies into the use of nanoparticles as immune- interactions with anionic cell membranes [163]. In vitro stud-
potentiating adjuvants are clearly needed. As the interaction of ies suggested that a cationic surface could signicantly enhance
nanoparticles with the immune system becomes more fully under- the uptake of polystyrene particles of micron size (1 m) by
stood, we expect their impact to be broadened. macrophages and DCs in comparison with a neutral or negative sur-
face [163,169,170], but not for the smaller nanoparticles (100 nm)
4. Nanoparticle interactions with antigen presenting cells [163]. However, other in vivo studies revealed that either positively
[171] or negatively charged [172] liposomes could act as efcient
Incorporating antigenic components into nanoparticles has adjuvants to induce cell-mediated immune response. Furthermore,
attracted extensive interest with a focus on how to deliver antigen due to their electrostatic interaction with anionic cell membranes,
more efciently to antigen presenting cells (APCs) and subse- cationic particles are more likely to induce hemolysis and platelet
quently induce their maturation and cross presentation of antigen aggregation than neutral or anionic particles [173].
for activation of a potent immune response [148152]. As special- Particle shape plays an equally important role in the interaction
ized APCs which efciently uptake and process antigen, dendritic between nanoparticles and APCs. For big particles (>1 m), parti-
cells (DCs) and macrophages are often targeted in vaccine design. cle shape plays a dominant role in phagocytosis by macrophages as
Good understanding of DC and macrophage uptake mechanisms the uptake of particles is strongly dependent on the local shape at
and interactions of NPs with these cells is therefore very important the interface between particles and APCs [174]. Worm-like parti-
for developing efcacious nanoparticle vaccines [153155]. Stud- cles with high aspect ratios (>20) exhibited negligible phagocytosis
ies have reported that size, charge and shape of nanoparticles play compared to spherical particles [175]. On the other hand, spherical
signicant roles in antigen uptake. gold nanoparticles (AuNPs) (40 nm) were more effective in induc-
Generally, nanoparticles having a comparable size to pathogens ing antibody response than other shapes (cube and rod) or the
can be easily recognized and are consequently taken up ef- 20 nm-sized AuNPs, even though the rods (40 nm 10 nm) were
ciently by APCs for induction of immune response [156162]. more efcient in APC uptake than the spherical and cubic AuNPs
DCs preferentially uptake virus-sized particles (20200 nm) while [59].
macrophages preferentially uptake larger particles (0.55 m) A number of studies also reported the effect of hydrophobicity,
[156]. In an in vitro study using polystyrene particles ranging from showing higher immune response for hydrophobic particles than
0.04 m to 15 m, the optimum size for DC uptake was found to be hydrophilic ones [176,177]. A number of other factors such as
L. Zhao et al. / Vaccine 32 (2014) 327337 333

surface modication (pegylation, targeting ligands) and vaccine three-dimensional images in vivo. Superparamagnetic iron oxide
cargo [45] have been shown to affect the interaction between nanoparticles (SPION) have been extensively used as contrast
nanoparticles and APCs as well. agents for morphological imaging [199,200]. PET usually employs
an imaging device (PET scanner) and a radiotracer that is usually
intravenously injected into the bloodstream. Due to high sensitivity
5. Nanoparticle-biosystem interactions of this technique, it is used to study the biodistribution of particles
of interest. The only disadvantage of this technique is relatively
Designing safe and efcacious nanoparticle vaccines requires a low spatial resolution as compared to other techniques. PET imag-
thorough understanding of the interaction of nanoparticles with ing of 64 Cu radiolabelled shell-crosslinked nanoparticles has been
biological systems which then determines the fate of nanoparticles demonstrated [201]. Fluorescence imaging facilitates imaging of
in vivo. Physicochemical properties of nanoparticles including size, nanoparticles using uorescent tags. Dye-doped silica nanoparti-
shape, surface charge, and hydrophobicity inuence the interac- cles as contrast imaging agents for in vivo uorescence imaging in
tion of nanoparticles with plasma proteins [178,179] and immune small animals have been reported [202].
cells [176]. These interactions as well as morphology of vascular Nowadays, more attention is being paid to synergize two or
endothelium play an important role in distribution of nanoparticles more imaging techniques that complement each other and provide
in various organs and tissues of the body. an opportunity to overcome shortcomings of individual techniques
The lymph node (LN) is a target organ for vaccine delivery since in terms of resolution or sensitivity. For instance, simultaneous
cells of the immune system, in particular B and T cells, reside there. PET-MRI imaging is a new emerging hybrid imaging system that
Ensuring delivery of antigen to LNs, by direct drainage [180,181] combines the morphological imaging component of MRI with the
or by migration of well-armed peripheral APCs [182], for optimum functional imaging component of PET [203]. Multifunctionality of
induction of immune response is therefore an important aspect of nanoparticles can be utilized for such hyphenated imaging.
nanoparticle vaccine design. Distribution of nanoparticles to the
LN is mainly affected by size [183,184]. Nanoparticles with a size 6. Concluding remark
range of 10100 nm can penetrate the extracellular matrix easily
and travel to the LNs where they are taken up by resident DCs Nanoparticle-containing vaccines have attracted tremendous
for activation of immune response [184187]. Particles of larger interest in recent years, and a wide variety of nanoparticles have
size (>100 nm) linger at the administration point [181,186,188] been developed and employed as delivery vehicles or immune
and are subsequently scavenged by local APCs [181,187,189], while potentiators, allowing not only improvement of antigen stability
smaller particles (<10 nm) drain to the blood capillaries [184,189]. and the enhancement of antigen processing and immunogenicity,
The route of administration and biological environment to which but also the targeted delivery and slow release of antigens. In addi-
nanoparticles are exposed could also affect the draining of nanopar- tion, nanoparticles have been increasingly used to deliver not only
ticles to the LN. It was reported that small PEG coated liposomes antigen of interest but also co-adjuvant, such as poly(I:C), CpG and
(8090 nm) were signicantly present in larger amounts in LNs MPL [188,204]. However, the application of nanoparticles in vac-
after subcutaneous administration as compared to intravenous and cine delivery as well as in drug delivery is still at an early stage
intraperitoneal administration [190]. of development. A number of challenges remain, including dif-
In addition to targeting lymphatic organ for efcient activation culty in reproducibly synthesizing non-aggregated nanoparticles
of immune response, design of nanoparticle vaccines also needs having consistent and desirable properties, a lack of fundamen-
to consider nanoparticle clearance from the body. Adverse effects tal understanding of how the physical properties of nanoparticles
may occur when nanoparticles are not degraded or excreted from affect their biodistribution and targeting, and how these proper-
the body and hence, accumulate in different organs and tissues. ties inuence their interactions with the biological system at all
Clearance of nanoparticles could be achieved through degradation levels from cell through tissue and to whole body. Therefore, ratio-
by the immune system or by renal or biliary clearance. nal design in combination with the reproducible production of
Renal clearance through kidneys can excrete nanoparticles nanoparticles with desirable properties, functionalities and ef-
smaller than 8 nm [191,192]. Surface charge also plays an important cacy becomes increasingly important, and it is anticipated that
role in determining renal clearance of nanoparticles. Few reports the adoption of new technologies, for example microuidics, for
have suggested that for appropriate identically sized particles, the controlled synthesis of nanoparticles will accelerate the devel-
based on surface charge, ease of renal clearance follows the order opment of suitable nanoparticles for pharmaceutical applications
of positively-charged < neutral < negatively charged [193,194]. This [205]. Furthermore, by integrating some other attractive proper-
may be attributed to the presence of negatively-charged membrane ties, such as slow release, targeting and alternative administration
of glomerular capillary [195]. methods and delivery pathways, novel vaccine systems for unmet
On the other hand, biliary clearance through liver allows excre- needs including single-dose and needle-free delivery will become
tion of nanoparticles larger than 200 nm [191,196]. Surface charge practical in the near future.
also plays role in biliary clearance with increase in surface charges
showing increased distribution of nanoparticles in the liver [197]. References
Furthermore, a study reported shape dependent distribution of
nanoparticles where short rod nanoparticles were predominantly [1] Oberg AL, Kennedy RB, Li P, Ovsyannikova IG, Poland GA. Systems biology
found in liver, while long rods were found in spleen. Short rod approaches to new vaccine development. Current Opinion in Immunology
2011;23:43643.
nanoparticles were excreted at a faster rate than longer ones [198]. [2] Rappuoli R, Mandl CW, Black S, De Gregorio E. Vaccines for the twenty-rst
In order to aid understanding of interaction of nanoparticles century society. Nature Reviews Immunology 2011;11:86572.
with immune cells and the biosystem, many different in vivo molec- [3] Mamo T, Poland GA. Nanovaccinology: the next generation of vaccines meets
21st century materials science and engineering. Vaccine 2012;30:660911.
ular imaging techniques including magnetic resonance imaging [4] Couvreur P, Vauthier C. Nanotechnology: intelligent design to treat complex
(MRI), positron emission tomography (PET), uorescence imag- disease. Pharmaceutical Research 2006;23:141750.
ing, single photon emission computed tomography (SPECT), X-ray [5] Moghimi SM, Hunter AC, Murray JC. Nanomedicine: current status and future
prospects. The FASEB Journal 2005;19:31130.
computed tomography (CT) and ultrasound imaging could be
[6] Treuel L, Jiang X, Nienhaus GU. New views on cellular uptake and trafc-
employed. Owing to its excellent soft tissue contrast and non- king of manufactured nanoparticles. Journal of the Royal Society Interface
invasive nature, MRI imaging is extensively used for obtaining 2013;10:20120939.
334 L. Zhao et al. / Vaccine 32 (2014) 327337

[7] Wagner V, Dullaart A, Bock A-K, Zweck A. The emerging nanomedicine land- [37] Moon JJ, Suh H, Polhemus ME, Ockenhouse CF, Yadava A, Irvine DJ. Antigen-
scape. Nature Biotechnology 2006;24:12117. displaying lipid-enveloped PLGA nanoparticles as delivery agents for a
[8] Global Industry Analysts Inc. Nanomedicine a global market report; 2009. Plasmodium vivax malaria vaccine. PloS ONE 2012;7:e31472.
[9] Tissot AC, Maurer P, Nussberger J, Sabat R, Pster T, Ignatenko S, et al. Effect [38] Scheerlinck J-PY, Gloster S, Gamvrellis A, Mottram PL, Plebanski M. Systemic
of immunisation against angiotensin II with CYT006-AngQb on ambulatory immune responses in sheep, induced by a novel nano-bead adjuvant. Vaccine
blood pressure: a double-blind, randomized, placebo-controlled phase IIa 2006;24:112431.
study. The Lancet 2008;371:8217. [39] Uenaka A, Wada H, Isobe M, Saika T, Tsuji K, Sato E, et al. T cell
[10] Pankhurst QA, Connolly J, Jones SK, Dobson J. Applications of mag- immunomonitoring and tumor responses in patients immunized with a com-
netic nanoparticles in biomedicine. Journal of Physics D: Applied Physics plex of cholesterol-bearing hydrophobized pullulan (CHP) and NY-ESO-1
2003;36:R16781. protein. Cancer Immunity: A Journal of the Academy of Cancer Immunology
[11] Maurer P, Jennings GT, Willers J, Rohner F, Lindman Y, Roubicek K, et al. 2007;7:919.
A therapeutic vaccine for nicotine dependence: preclinical efcacy, and [40] Hasegawa K, Noguchi Y, Koizumi F, Uenaka A, Tanaka M, Shimono M, et al.
phase I safety and immunogenicity. European Journal of Immunology In vitro stimulation of CD8 and CD4T cells by dendritic cells loaded with a
2005;35:203140. complex of cholesterol-bearing hydrophobized pullulan and NY-ESO-1 pro-
[12] Dobrovolskaia MA, McNeil SE. Immunological properties of engineered nano- tein: identication of a new HLA-DR15-binding CD4 T-cell epitope. Clinical
materials. Nature Nanotechnology 2007;2:46978. Cancer Research 2006;12:19217.
[13] Roldao A, Mellado MCM, Castilho LR, Carrondo MJ, Alves PM. Virus-like par- [41] Li P, Luo Z, Liu P, Gao N, Zhang Y, Pan H, et al. Bioreducible alginate-poly
ticles in vaccine development. Expert Review of Vaccines 2010;9:114976. (ethylenimine) nanogels as an antigen-delivery system robustly enhance
[14] Bolhassani A, Safaiyan S, Rafati S. Improvement of different vaccine delivery vaccine-elicited humoral and cellular immune responses. Journal of Con-
systems for cancer therapy. Molecular Cancer 2011;10:322. trolled Release 2013;168:2719.
[15] Krishnamachari Y, Geary S, Lemke C, Salem A. Nanoparticle delivery systems [42] Honda-Okubo Y, Saade F, Petrovsky N. AdvaxTM a polysaccharide adjuvant
in cancer vaccines. Pharmaceutical Research 2011;28:21536. derived from delta inulin, provides improved inuenza vaccine protection
[16] Hamdy S, Haddadi A, Hung RW, Lavasanifar A. Targeting dendritic cells with through broad-based enhancement of adaptive immune responses. Vaccine
nano-particulate PLGA cancer vaccine formulations. Advanced Drug Delivery 2012;30:537381.
Reviews 2011;63:94355. [43] Saade F, Honda-Okubo Y, Trec S, Petrovsky N. A novel hepatitis B vaccine
[17] Chackerian B. Virus-like particle based vaccines for Alzheimer disease. Human containing AdvaxTM , a polysaccharide adjuvant derived from delta inulin,
Vaccines 2010;6:92630. induces robust humoral and cellular immunity with minimal reactogenicity
[18] Correia-Pinto JF, Csaba N, Alonso MJ. Vaccine delivery carriers: insights in preclinical testing. Vaccine 2013;31:19992007.
and future perspectives. International Journal of Pharmaceutics 2013;440: [44] Feng G, Jiang Q, Xia M, Lu Y, Qiu W, Zhao D, et al. Enhanced immune response
2738. and protective effects of nano-chitosan-based DNA vaccine encoding T cell
[19] Kushnir N, Streateld SJ, Yusibov V. Virus-like particles as a highly efcient epitopes of Esat-6 and FL against Mycobacterium tuberculosis infection. PLoS
vaccine platform: diversity of targets and production systems and advances ONE 2013;8:e61135.
in clinical development. Vaccine 2012;31:5883. [45] Thomann-Harwood LJ, Kaeuper P, Rossi N, Milona P, Herrmann B, McCul-
[20] Plummer EM, Manchester M. Viral nanoparticles and virus-like particles: lough KC. Nanogel vaccines targeting dendritic cells: contributions of the
platforms for contemporary vaccine design. Wiley Interdisciplinary Reviews: surface decoration and vaccine cargo on cell targeting and activation. Journal
Nanomedicine and Nanobiotechnology 2011;3:17496. of Controlled Release 2013;166:95105.
[21] Food and Drug Administration US. Considering whether an FDA- [46] Zhao F, Zhang X, Liu S, Zeng T, Yu J, Gu W, et al. Assessment of the immune
regulated product involves the application of nanotechnology; 2011 responses to Treponema pallidum Gpd DNA vaccine adjuvanted with IL-2
http://wwwfdagov/regulatoryinformation/guidances/ucm257698htm and chitosan nanoparticles before and after Treponema pallidum challenge
[22] Thomas C, Rawat A, Hope-Weeks L, Ahsan F. Aerosolized PLGA nanoparticles in rabbits. Science China-Life Sciences 2013;56:17480.
enhance humoral, mucosal and cytokine responses to hepatitis B vaccine. [47] Nanda RK, Edao BM, Hajam IA, Sekar SC, Ganesh K, Bhanuprakash V, et al.
Molecular Pharmaceutics 2011;8:40515. An effective mannosylated chitosan nanoparticle DNA vaccine for FMD virus.
[23] Kim SY, Doh HJ, Jang MH, Ha YJ, Chung SI, Park HJ. Oral immunization with Virologica Sinica 2012;27:3736.
Helicobacter pylori-loaded poly(d,l-lactide-co-glycolide) nanoparticles. Heli- [48] Zhao K, Chen G, Shi X-m, Gao T-t, Li W, Zhao Y, et al. Preparation and efcacy of
cobacter 1999;4:339. a live Newcastle disease virus vaccine encapsulated in chitosan nanoparticles.
[24] Vila A, Sanchez A, Evora C, Soriano I, Vila Jato J, Alonso M. PEG-PLA nanopar- PLoS ONE 2012;7:e53314.
ticles as carriers for nasal vaccine delivery. Journal of Aerosol Medicine [49] Borges O, Cordeiro-da-Silva A, Tavares J, Santarm N, de Sousa A, Borchard G,
2004;17:17485. et al. Immune response by nasal delivery of hepatitis B surface antigen and
[25] L J-M, Wang X, Marin-Muller C, Wang H, Lin PH, Yao Q, et al. Current advances codelivery of a CpG ODN in alginate coated chitosan nanoparticles. European
in research and clinical applications of PLGA-based nanotechnology. Expert Journal of Pharmaceutics and Biopharmaceutics 2008;69:40516.
Review of Molecular Diagnostics 2009;9:32541. [50] Chua BY, Al Kobaisi M, Zeng WG, Mainwaring D, Jackson DC. Chitosan
[26] Demento SL, Cui W, Criscione JM, Stern E, Tulipan J, Kaech SM, et al. Role of microparticles and nanoparticles as biocompatible delivery vehicles for
sustained antigen release from nanoparticle vaccines in shaping the T cell peptide and protein-based immunocontraceptive vaccines. Molecular Phar-
memory phenotype. Biomaterials 2012;33:495764. maceutics 2012;9:8190.
[27] Diwan M, Tafaghodi M, Samuel J. Enhancement of immune responses by co- [51] Arca HC, Gnbeyaz M, Senel S. Chitosan-based systems for the delivery of
delivery of a CpG oligodeoxynucleotide and tetanus toxoid in biodegradable vaccine antigens. Expert Review of Vaccines 2009;8:93753.
nanospheres. Journal of Controlled Release 2002;85:24762. [52] Gtze O, Mller-Eberhard HJ. The C3-activator system: an alternate
[28] Silva AL, Rosalia RA, Sazak A, Carstens MG, Ossendorp F, Oostendorp J, et al. pathway of complement activation. Journal of Experimental Medicine
Optimization of encapsulation of a synthetic long peptide in PLGA nanoparti- 1971;134:90108.
cles: low-burst release is crucial for efcient CD8(+) T cell activation. European [53] Dmoulins T, Bassi I, Thomann-Harwood L, Jandus C, Kaeuper P, Simon H-U,
Journal of Pharmaceutics and Biopharmaceutics 2013;83:33845. et al. Alginate-coated chitosan nanogel capacity to modulate the effect of TLR
[29] Manish M, Rahi A, Kaur M, Bhatnagar R, Singh S. A single-dose PLGA encap- ligands on blood dendritic cells. Nanomedicine: Nanotechnology, Biology and
sulated protective antigen domain 4 nanoformulation protects mice against Medicine 2013;9:80617.
Bacillus anthracis spore challenge. PLoS ONE 2013;8:e6188590. [54] Vinogradov S, Batrakova E, Kabanov A. Poly(ethylene glycol)-
[30] Lutsiak M, Kwon GS, Samuel J. Biodegradable nanoparticle delivery of a Th2- polyethyleneimine NanoGelTM particles: novel drug delivery systems
biased peptide for induction of Th1 immune responses. Journal of Pharmacy for antisense oligonucleotides. Colloids and Surfaces B: Biointerfaces
and Pharmacology 2006;58:73947. 1999;16:291304.
[31] Akagi T, Baba M, Akashi M. Biodegradable nanoparticles as vaccine adjuvants [55] Ferreira SA, Gama FM, Vilanova M. Polymeric nanogels as vaccine delivery sys-
and delivery systems: regulation of immune responses by nanoparticle-based tems. Nanomedicine: Nanotechnology, Biology and Medicine 2012;9:15973.
vaccine. In: Kunugi S, Yamaoka T, editors. Polymers in nanomedicine. Berlin: [56] Raemdonck K, Demeester J, De Smedt S. Advanced nanogel engineering for
Springer-Verlag Berlin; 2012. p. 3164. drug delivery. Soft Matter 2009;5:70715.
[32] Akagi T, Kaneko T, Kida T, Akashi M. Preparation and characteriza- [57] Nochi T, Yuki Y, Takahashi H, Sawada S.-i., Mejima M, Kohda T, et al.
tion of biodegradable nanoparticles based on poly (-glutamic acid) Nanogel antigenic protein-delivery system for adjuvant-free intranasal vac-
with l-phenylalanine as a protein carrier. Journal of Controlled Release cines. Nature Materials 2010;9:5728.
2005;108:22636. [58] Debache K, Kropf C, Schutz CA, Harwood LJ, Kauper P, Monney T, et al. Vacci-
[33] Kalkanidis M, Pietersz GA, Xiang SD, Mottram PL, Crimeen-Irwin B, Ardipradja nation of mice with chitosan nanogel-associated recombinant NcPDI against
K, et al. Methods for nano-particle based vaccine formulation and evaluation challenge infection with Neospora caninum tachyzoites. Parasite Immunology
of their immunogenicity. Methods 2006;40:209. 2011;33:8194.
[34] Minigo G, Scholzen A, Tang CK, Hanley JC, Kalkanidis M, Pietersz GA, et al. [59] Niikura K, Matsunaga T, Suzuki T, Kobayashi S, Yamaguchi H, Orba Y, et al.
Poly-l-lysine-coated nanoparticles: a potent delivery system to enhance DNA Gold nanoparticles as a vaccine platform: inuence of size and shape on
vaccine efcacy. Vaccine 2007;25:131627. immunological responses in vitro and in vivo. ACS Nano 2013;7:392638.
[35] Peek LJ, Middaugh CR, Berkland C. Nanotechnology in vaccine delivery. [60] Gregory AE, Titball R, Williamson D. Vaccine delivery using nanoparticles.
Advanced Drug Delivery Reviews 2008;60:91528. Frontiers in Cellular and Infection Microbiology 2013;3:13.
[36] Danhier F, Ansorena E, Silva JM, Coco R, Le Breton A, Prat V. PLGA-based [61] Marradi M, Chiodo F, Garcia I, Penades S. Glyconanoparticles as multifunc-
nanoparticles: an overview of biomedical applications. Journal of Controlled tional and multimodal carbohydrate systems. Chemical Society Reviews
Release 2012;161:50522. 2013;42:472845.
L. Zhao et al. / Vaccine 32 (2014) 327337 335

[62] Stone JW, Thornburg NJ, Blum DL, Kuhn SJ, Wright DW, Crowe Jr JE. [88] He Q, Mitchell AR, Johnson SL, Wagner-Bartak C, Morcol T, Bell SJD. Cal-
Gold nanorod vaccine for respiratory syncytial virus. Nanotechnology cium phosphate nanoparticle adjuvant. Clinical and Diagnostic Laboratory
2013;24:295102. Immunology 2000;7:899903.
[63] Tao W, Ziemer KS, Gill HS. Gold nanoparticle-M2e conjugate coformu- [89] He Q, Mitchell A, Morcol T, Bell SJ. Calcium phosphate nanoparticles induce
lated with CpG induces protective immunity against inuenza A virus. mucosal immunity and protection against herpes simplex virus type 2. Clin-
Nanomedicine 2013:116, http://dx.doi.org/10.2217/nnm.13.58 [Epub ahead ical and Diagnostic Laboratory Immunology 2002;9:10214.
of print]. [90] Joyappa DH, Ashok Kumar C, Banumathi N, Reddy GR, Suryanarayana VV.
[64] Chen Y-S, Hung Y-C, Lin W-H, Huang GS. Assessment of gold nanoparticles Calcium phosphate nanoparticle prepared with foot and mouth disease virus
as a size-dependent vaccine carrier for enhancing the antibody response P1-3CD gene construct protects mice and guinea pigs against the challenge
against synthetic foot-and-mouth disease virus peptide. Nanotechnology virus. Veterinary Microbiology 2009;139:5866.
2010;21:1951018. [91] Giddam AK, Zaman M, Skwarczynski M, Toth I. Liposome-based deliv-
[65] Xu L, Liu Y, Chen Z, Li W, Liu Y, Wang L, et al. Surface-engineered gold ery system for vaccine candidates: constructing an effective formulation.
nanorods: promising DNA vaccine adjuvant for HIV-1 treatment. Nano Letters Nanomedicine 2012;7:187793.
2012;12:200312. [92] Sharma S, Mukkur T, Benson HA, Chen Y. Pharmaceutical aspects of intranasal
[66] Bianco A, Kostarelos K, Prato M. Applications of carbon nanotubes in drug delivery of vaccines using particulate systems. Journal of Pharmaceutical Sci-
delivery. Current Opinion in Chemical Biology 2005;9:6749. ences 2009;98:81243.
[67] Wang T, Zou M, Jiang H, Ji Z, Gao P, Cheng G. Synthesis of a novel kind of [93] Khatri K, Goyal AK, Gupta PN, Mishra N, Mehta A, Vyas SP. Surface modied
carbon nanoparticle with large mesopores and macropores and its applica- liposomes for nasal delivery of DNA vaccine. Vaccine 2008;26:222533.
tion as an oral vaccine adjuvant. European Journal of Pharmaceutical Sciences [94] Glck R, Moser C, Metcalfe IC. Inuenza virosomes as an efcient sys-
2011;44:6539. tem for adjuvanted vaccine delivery. Expert Opinion on Biological Therapy
[68] Smart S, Cassady A, Lu G, Martin D. The biocompatibility of carbon nanotubes. 2004;4:113945.
Carbon 2006;44:103447. [95] Li S, Rizzo M, Bhattacharya S, Huang L. Characterization of cationic lipid-
[69] Villa CH, Dao T, Ahearn I, Fehrenbacher N, Casey E, Rey DA, et al. Single-walled protamine-DNA (LPD) complexes for intravenous gene delivery. Gene
carbon nanotubes deliver peptide antigen into dendritic cells and enhance IgG Therapy 1998;5:9307.
responses to tumor-associated antigens. ACS Nano 2011;5:530011. [96] Li S, Huang L. In vivo gene transfer via intravenous administration of cationic
[70] Parra J, Abad-Somovilla A, Mercader JV, Taton TA, Abad-Fuentes A. Carbon lipid-protamine-DNA (LPD) complexes. Gene Therapy 1997;4:891900.
nanotube-protein carriers enhance size-dependent self-adjuvant antibody [97] Moon JJ, Suh H, Bershteyn A, Stephan MT, Liu H, Huang B, et al. Interbilayer-
response to haptens. Journal of Controlled Release 2013;170:24251. crosslinked multilamellar vesicles as synthetic vaccines for potent humoral
[71] Pantarotto D, Partidos CD, Hoebeke J, Brown F, Kramer E, Briand J-P, and cellular immune responses. Nature Materials 2011;10:24351.
et al. Immunization with peptide-functionalized carbon nanotubes enhances [98] Watson DS, Endsley AN, Huang L. Design considerations for liposomal
virus-specic neutralizing antibody responses. Chemistry and Biology vaccines: inuence of formulation parameters on antibody and cell-
2003;10:9616. mediated immune responses to liposome associated antigens. Vaccine
[72] Ow H, Larson DR, Srivastava M, Baird BA, Webb WW, Wiesner U. Bright 2012;30:225672.
and stable core-shell uorescent silica nanoparticles. Nano Letters 2005;5: [99] Homhuan A, Prakongpan S, Poomvises P, Maas RA, Crommelin DJ, Kersten GF,
1137. et al. Virosome and ISCOM vaccines against Newcastle disease: preparation,
[73] Benezra M, Penate-Medina O, Zanzonico PB, Schaer D, Ow H, Burns characterization and immunogenicity. European Journal of Pharmaceutical
A, et al. Multimodal silica nanoparticles are effective cancer-targeted Sciences 2004;22:45968.
probes in a model of human melanoma. Journal of Clinical Investigation [100] Aguilar J, Rodriguez E. Vaccine adjuvants revisited. Vaccine 2007;25:375262.
2011;121:276880. [101] Morein B, Sundquist B, Hoglund S, Dalsgaard K, Osterhaus A. ISCOM a novel
[74] Niu Y, Popat A, Yu M, Karmakar S, Gu W, Yu C. Recent advances in the rational structure for antigenic presentation of membrane proteins from enveloped
design of silica-based nanoparticles for gene therapy. Therapeutic Delivery viruses. Nature 1984;308:45760.
2012;3:121737. [102] Pearse MJ, Drane D. ISCOMATRIXTM adjuvant: a potent inducer of humoral
[75] Yu M, Jambhrunkar S, Thorn P, Chen J, Gu W, Yu C. Hyaluronic acid and cellular immune responses. Vaccine 2004;22:23915.
modied mesoporous silica nanoparticles for targeted drug delivery to CD44- [103] Sambhara S, Woods S, Arpino R, Kurichh A, Tamane A, Underdown B, et al.
overexpressing cancer cells. Nanoscale 2013;5:17883. Heterotypic protection against inuenza by immunostimulating complexes
[76] Alshamsan A, Haddadi A, Incani V, Samuel J, Lavasanifar A, Uludag H. For- is associated with the induction of cross-reactive cytotoxic T lymphocytes.
mulation and delivery of siRNA by oleic acid and stearic acid modied Journal of Infectious Diseases 1998;177:126674.
polyethylenimine. Molecular Pharmaceutics 2008;6:12133. [104] Coulter A, Harris R, Davis R, Drane D, Cox J, Ryan D, et al. Intranasal vaccination
[77] Xia T, Kovochich M, Liong M, Meng H, Kabehie S, George S, et al. with ISCOMATRIX adjuvanted inuenza vaccine. Vaccine 2003;21:9469.
Polyethyleneimine coating enhances the cellular uptake of mesoporous sil- [105] Mohamedi S, Brewer J, Alexander J, Heath A, Jennings R. Antibody responses,
ica nanoparticles and allows safe delivery of siRNA and DNA constructs. ACS cytokine levels and protection of mice immunized with HSV-2 antigens for-
Nano 2009;3:327386. mulated into NISV or ISCOM delivery systems. Vaccine 2000;18:208394.
[78] He X-x, Wang K, Tan W, Liu B, Lin X, He C, et al. Bioconjugated nanoparticles [106] Agrawal L, Haq W, Hanson CV, Rao DN. Generating neutralizing antibod-
for DNA protection from cleavage. Journal of the American Chemical Society ies, Th1 response and MHC non restricted immunogenicity of HIV-I env and
2003;125:71689. gag peptides in liposomes and ISCOMs with in-built adjuvanticity. Journal of
[79] Mody KT, Popat A, Mahony D, Cavallaro AS, Yu C, Mitter N. Mesoporous Immune Based Therapies and Vaccines 2003;1:526.
silica nanoparticles as antigen carriers and adjuvants for vaccine delivery. [107] Noad R, Roy P. Virus-like particles as immunogens. Trends in Microbiology
Nanoscale 2013;5:516779. 2003;11:43844.
[80] Carvalho LV, Ruiz RdC, Scaramuzzi K, Marengo EB, Matos JR, Tambourgi DV, [108] Grgacic EV, Anderson DA. Virus-like particles: passport to immune recogni-
et al. Immunological parameters related to the adjuvant effect of the ordered tion. Methods 2006;40:605.
mesoporous silica SBA-15. Vaccine 2010;28:782936. [109] Zhang LF, Zhou J, Chen S, Cai LL, Bao QY, Zheng FY, et al. HPV6b virus
[81] Mahony D, Cavallaro AS, Stahr F, Mahony TJ, Qiao SZ, Mitter N. Mesoporous like particles are potent immunogens without adjuvant in man. Vaccine
silica nanoparticles act as a self-adjuvant for ovalbumin model antigen in 2000;18:10518.
mice. Small 2013;9:313846. [110] Andre FE. Overview of a 5-year clinical-experience with a yeast-derived
[82] Guo H-C, Feng X-M, Sun S-Q, Wei Y-Q, Sun D-H, Liu X-T, et al. Immuniza- hepatitis-B vaccine. Vaccine 1990;8:S748.
tion of mice by hollow mesoporous silica nanoparticles as carriers of porcine [111] Cutts FT, Franceschi S, Goldie S, Castellsague X, de Sanjose S, Garnett G, et al.
circovirus type 2 ORF2 protein. Virology Journal 2012;9:108. Human papillomavirus and HPV vaccines: a review. Bulletin of the World
[83] Cheng K, El-Boubbou K, Landry CC. Binding of HIV-1 gp120 glycoprotein to sil- Health Organization 2007;85:71926.
ica nanoparticles modied with CD4 glycoprotein and CD4 peptide fragments. [112] Bin Park S. Hepatitis E vaccine debuts. Nature 2012;491:212.
ACS Applied Materials & Interfaces 2011;4:23543. [113] Pushko P, Pumpens P, Grens E. Development of virus-like particle technology
[84] Manzano M, Aina V, Arean C, Balas F, Cauda V, Colilla M, et al. Studies on from small highly symmetric to large complex virus-like particle structures.
MCM-41 mesoporous silica for drug delivery: effect of particle morphol- Intervirology 2013;56:14165.
ogy and amine functionalization. Chemical Engineering Journal 2008;137: [114] Pattenden LK, Middelberg APJ, Niebert M, Lipin DI. Towards the prepara-
307. tive and large-scale precision manufacture of virus-like particles. Trends in
[85] Zhai W, He C, Wu L, Zhou Y, Chen H, Chang J, et al. Degradation of hol- Biotechnology 2005;23:5239.
low mesoporous silica nanoparticles in human umbilical vein endothelial [115] Campbell S, Vogt VM. In vitro assembly of virus-like particles with Rous
cells. Journal of Biomedical Materials Research Part B: Applied Biomaterials sarcoma virus Gag deletion mutants: identication of the p10 domain as a
2012;100B:1397403. morphological determinant in the formation of spherical particles. Journal of
[86] Yamada H, Urata C, Aoyama Y, Osada S, Yamauchi Y, Kuroda K. Preparation Virology 1997;71:442535.
of colloidal mesoporous silica nanoparticles with different diameters and [116] Snchez-Rodrguez SP, Mnch-Anguiano L, Echeverra O, Vzquez-Nin G,
their unique degradation behavior in static aqueous systems. Chemistry of Mora-Pale M, Dordick JS, et al. Human parvovirus B19 virus-like particles:
Materials 2012;24:146271. in vitro assembly and stability. Biochimie 2012;94:8708.
[87] Chen K, Zhang J, Gu H. Dissolution from inside: a unique degradation [117] Zhang W, Carmichael J, Ferguson J, Inglis S, Ashraan H, Stanley M. Expression
behaviour of core-shell magnetic mesoporous silica nanoparticles and of human papillomavirus type 16 L1 protein in Escherichia coli: denatur-
the effect of polyethyleneimine coating. Journal of Materials Chemistry ation, renaturation, and self-assembly of virus-like particles in vitro. Virology
2012;22:2200512. 1998;243:42331.
336 L. Zhao et al. / Vaccine 32 (2014) 327337

[118] Liew MWO, Chuan YP, Middelberg APJ. High-yield and scalable cell-free [145] Wibowo N. Engineering viral capsomeres as a vaccine platform. The
assembly of virus-like particles by dilution. Biochemical Engineering Journal University of Queensland: Australian Institute for Bioengineering & Nano-
2012;67:8896. technology; 2012. p. 85107 [Ph.D. thesis].
[119] Lu Y, Welsh JP, Chan W, Swartz JR. Escherichia coli-based cell free production [146] Vallhov H, Kupferschmidt N, Gabrielsson S, Paulie S, Strmme M, Garcia-
of agellin and ordered agellin display on virus-like particles. Biotechnology Bennett AE, et al. Adjuvant properties of mesoporous silica particles tune the
and Bioengineering 2013;110:207385. development of effector T cells. Small 2012;8:211624.
[120] Chuan YP, Lua LHL, Middelberg APJ. Virus-like particle bioprocessing. Wein- [147] Vallhov H, Gabrielsson S, Strmme M, Scheynius A, Garcia-Bennett AE. Meso-
heim, Germany: Biopharmaceutical Production Technology: Wiley-VCH porous silica particles induce size dependent effects on human dendritic cells.
Verlag GmbH & Co. KGaA; 2012. p. 13963. Nano Letters 2007;7:357682.
[121] Lua LHL, Connors NK, Sainsbury F, Chuan YP, Wibowo N, Middelberg APJ. [148] Reddy ST, Swartz MA, Hubbell JA. Targeting dendritic cells with biomate-
Bioengineering virus-like particles as vaccines. Biotechnology and Bioengi- rials: developing the next generation of vaccines. Trends in Immunology
neering 2013, http://dx.doi.org/10.1002/bit.25159. 2006;27:5739.
[122] Tissot AC, Renhofa R, Schmitz N, Cielens I, Meijerink E, Ose V, et al. Versa- [149] Jones KS. Biomaterials as vaccine adjuvants. Biotechnology Progress
tile virus-like particle carrier for epitope based vaccines. PLoS ONE 2010;5: 2008;24:80714.
e9809. [150] Abensee JEB. Interaction of dendritic cells with biomaterials. Seminars in
[123] Middelberg APJ, Rivera-Hernandez T, Wibowo N, Lua LHL, Fan Y, Magor G, et al. Immunology 2008;20:1018.
A microbial platform for rapid and low-cost virus-like particle and capsomere [151] Bachmann MF, Jennings GT. Vaccine delivery: a matter of size, geom-
vaccines. Vaccine 2011;29:715462. etry, kinetics and molecular patterns. Nature Reviews Immunology
[124] Neirynck S, Deroo T, Saelens X, Vanlandschoot P, Jou WM, Fiers W. A universal 2010;10:78796.
inuenza A vaccine based on the extracellular domain of the M2 protein. [152] Scheerlinck J-PY, Greenwood DLV. Virus-sized vaccine delivery systems. Drug
Nature Medicine 1999;5:115763. Discovery Today 2008;13:8827.
[125] Ionescu RM, Przysiecki CT, Liang X, Garsky VM, Fan J, Wang B, et al. [153] Zolnik BS, Gonzalez-Fernandez A, Sadrieh N, Dobrovolskaia MA. Minireview:
Pharmaceutical and immunological evaluation of human papillomavirus nanoparticles and the immune system. Endocrinology 2010;151:45865.
virus like particle as an antigen carrier. Journal of Pharmaceutical Sciences [154] Dobrovolskaia MA, Aggarwal P, Hall JB, McNeil SE. Preclinical studies
2006;95:709. to understand nanoparticle interaction with the immune system and its
[126] Kanekiyo M, Wei C-J, Yassine HM, McTamney PM, Boyington JC, Whittle JR, potential effects on nanoparticle biodistribution. Molecular Pharmaceutics
et al. Self-assembling inuenza nanoparticle vaccines elicit broadly neutral- 2008;5:48795.
izing H1N1 antibodies. Nature 2013;499:1026. [155] Kumari A, Yadav SK. Cellular interactions of therapeutically delivered
[127] Champion CI, Kickhoefer VA, Liu G, Moniz RJ, Freed AS, Bergmann LL, et al. A nanoparticles. Expert Opinion on Drug Delivery 2011;8:14151.
vault nanoparticle vaccine induces protective mucosal immunity. PLoS ONE [156] Xiang SD, Scholzen A, Minigo G, David C, Apostolopoulos V, Mottram PL,
2009;4:e5409. et al. Pathogen recognition and development of particulate vaccines: does
[128] Shah P, Bhalodia D, Shelat P. Nanoemulsion: a pharmaceutical review. Sys- size matter. Methods 2006;40:19.
tematic Reviews in Pharmacy 2010;1:2432. [157] Akagi T, Wang X, Uto T, Baba M, Akashi M. Protein direct delivery to dendritic
[129] Aucouturier J, Dupuis L, Ganne V. Adjuvants designed for veterinary and cells using nanoparticles based on amphiphilic poly(amino acid) derivatives.
human vaccines. Vaccine 2001;19:266672. Biomaterials 2007;28:342736.
[130] OHagan DT. MF59 is a safe and potent vaccine adjuvant that enhances [158] Uto T, Wang X, Sato K, Haraguchi M, Akagi T, Akashi M, et al. Targeting
protection against inuenza virus infection. Expert Review of Vaccines of antigen to dendritic cells with poly(gamma-glutamic acid) nanoparticles
2007;6:699710. induces antigen-specic humoral and cellular immunity. Journal of Immunol-
[131] De Donato S, Granoff D, Minutello M, Lecchi G, Faccini M, Agnello M, et al. ogy 2007;178:297986.
Safety and immunogenicity of MF59-adjuvanted inuenza vaccine in the [159] Copland MJ, Baird MA, Rades T, McKenzie JL, Becker B, Reck F, et al. Liposomal
elderly. Vaccine 1999;17:3094101. delivery of antigen to human dendritic cells. Vaccine 2003;21:88390.
[132] Nicholson KG, Colegate AE, Podda A, Stephenson I, Wood J, Ypma E, et al. [160] Elamanchili P, Diwan M, Cao M, Samuel J. Characterization of poly(d,l-lactic-
Safety and antigenicity of non-adjuvanted and MF59-adjuvanted inuenza co-glycolic acid) based nanoparticulate system for enhanced delivery of
A/Duck/Singapore/97 (H5N3) vaccine: a randomized trial of two potential antigens to dendritic cells. Vaccine 2004;22:240612.
vaccines against H5N1 inuenza. The Lancet 2001;357:193743. [161] Lutsiak MEC, Robinson DR, Coester C, Kwon GS, Samuel J. Analysis of poly(d,l-
[133] Aucouturier J, Dupuis L, Deville S, Ascarateil S, Ganne V. Montanide ISA 720 lactic-co-glycolic acid) nanosphere uptake by human dendritic cells and
and 51: a new generation of water in oil emulsions as adjuvants for human macrophages in vitro. Pharmaceutical Research 2002;19:14807.
vaccines. Expert Review of Vaccines 2002;1:1118. [162] Lin AY, Lunsford J, Bear AS, Young JK, Eckels P, Luo L, et al. High-density sub-
[134] Kumar S, Jones TR, Oakley MS, Zheng H, Kuppusamy SP, Taye A, et al. CpG 100-nm peptide-gold nanoparticle complexes improve vaccine presentation
oligodeoxynucleotide and Montanide ISA 51 adjuvant combination enhanced by dendritic cells in vitro. Nanoscale Research Letters 2013;8:72.
the protective efcacy of a subunit malaria vaccine. Infection and Immunity [163] Foged C, Brodin B, Frokjaer S, Sundblad A. Particle size and surface charge
2004;72:94957. affect particle uptake by human dendritic cells in an in vitro model. Interna-
[135] Oliveira GA, Wetzel K, Calvo-Calle JM, Nussenzweig R, Schmidt A, Birkett A, tional Journal of Pharmaceutics 2005;298:31522.
et al. Safety and enhanced immunogenicity of a hepatitis B core particle plas- [164] Joshi VB, Geary SM, Salem AK. Biodegradable particles as vaccine delivery
modium falciparum malaria vaccine formulated in adjuvant Montanide ISA systems: size matters. AAPS Journal 2013;15:8594.
720 in a phase I trial. Infection and Immunity 2005;73:358797. [165] Kanchan V, Panda AK. Interactions of antigen-loaded polylactide particles
[136] Dar P, Kalaivanan R, Sied N, Mamo B, Kishore S, Suryanaryana VS, et al. with macrophages and their correlation with the immune response. Bioma-
Montanide ISATM 201 adjuvanted FMD vaccine induces improved immune terials 2007;28:534457.
responses and protection in cattle. Vaccine 2013;31:332732. [166] Kim H, Uto T, Akagi T, Baba M, Akashi M. Amphiphilic poly(amino acid)
[137] Chuan YP, Zeng BY, OSullivan B, Thomas R, Middelberg APJ. Co-delivery nanoparticles induce size-dependent dendritic cell maturation. Advanced
of antigen and a lipophilic anti-inammatory drug to cells via a tai- Functional Materials 2010;20:392531.
lorable nanocarrier emulsion. Journal of Colloid and Interface Science [167] Cohen JA, Beaudette TT, Tseng WW, Bachelder EM, Mende I, Engleman EG,
2012;368:61624. et al. T-cell activation by antigen-loaded pH-sensitive hydrogel particles in
[138] Zeng BJ, Chuan YP, OSullivan B, Caminschi I, Lahoud MH, Thomas R, vivo: the effect of particle size. Bioconjugate Chemistry 2009;20:1119.
et al. Receptor-specic delivery of protein antigen to dendritic cells by [168] Yan S, Gu W, Xu ZP. Re-considering how particle size and other properties
a nanoemulsion formed using top-down non-covalent click self-assembly. of antigenadjuvant complexes impact on the immune responses. Journal of
Small 2013;9:373642. Colloid and Interface Science 2013;395:110.
[139] Oyewumi MO, Kumar A, Cui Z. Nano-microparticles as immune adjuvants: [169] Thiele L, Merkle HP, Walter E. Phagocytosis and phagosomal fate of surface-
correlating particle sizes and the resultant immune responses. Expert Review modied microparticles in dendritic cells and macrophages. Pharmaceutical
of Vaccines 2010;9:1095107. Research 2003;20:2218.
[140] Wendorf J, Singh M, Chesko J, Kazzaz J, Soewanan E, Ugozzoli M, et al. A [170] Wischke C, Borchert H-H, Zimmermann J, Siebenbrodt I, Lorenzen DR. Stable
practical approach to the use of nanoparticles for vaccine delivery. Journal cationic microparticles for enhanced model antigen delivery to dendritic cells.
of Pharmaceutical Sciences 2006;95:273850. Journal of Controlled Release 2006;114:35968.
[141] Stieneker F, Kreuter J, Lower J. High antibody-titers in mice with poly- [171] Nakanishi T, Kunisawa J, Hayashi A, Tsutsumi Y, Kubo K, Nakagawa S, et al.
methylmethacrylate nanoparticles as adjuvant for HIV vaccines. AIDS 1991;5: Positively charged liposome functions as an efcient immunoadjuvant in
4315. inducing cell-mediated immune response to soluble proteins. Journal of Con-
[142] Sltter B, Soema PC, Ding Z, Verheul R, Hennink W, Jiskoot W. Conjugation trolled Release 1999;61:23340.
of ovalbumin to trimethyl chitosan improves immunogenicity of the antigen. [172] Yotsumoto S, Aramaki Y, Kakiuchi T, Tsuchiya S. Induction of antigen-
Journal of Controlled Release 2010;143:20714. dependent interleukin-12 production by negatively charged liposomes
[143] Morel S, Didierlaurent A, Bourguignon P, Delhaye S, Baras B, Jacob V, encapsulating antigens. Vaccine 2004;22:35039.
et al. Adjuvant System AS03 containing [alpha]-tocopherol modulates innate [173] Goodman CM, McCusker CD, Yilmaz T, Rotello VM. Toxicity of gold nanopar-
immune response and leads to improved adaptive immunity. Vaccine ticles functionalized with cationic and anionic side chains. Bioconjugate
2011;29:246173. Chemistry 2004;15:897900.
[144] OHagan DT, Ott GS, Van Nest G. Recent advances in vaccine adjuvants: the [174] Champion JA, Mitragotri S. Role of target geometry in phagocytosis.
development of MF59 emulsion and polymeric microparticles. Molecular Proceedings of the National Academy of Sciences of the United States of
Medicine Today 1997;3:6975. America 2006;103:49304.
L. Zhao et al. / Vaccine 32 (2014) 327337 337

[175] Champion JA, Mitragotri S. Shape induced inhibition of phagocytosis of poly- [190] Allen T, Hansen C, Guo L. Subcutaneous administration of liposomes: a
mer particles. Pharmaceutical Research 2009;26:2449. comparison with the intravenous and intraperitoneal routes of injection.
[176] Hillaireau H, Couvreur P. Nanocarriers entry into the cell: relevance to drug Biochimica et Biophysica Acta (BBA)-Biomembranes 1993;1150:916.
delivery. Cellular and Molecular Life Sciences 2009;66:287396. [191] Longmire M, Choyke PL, Kobayashi H. Clearance properties of nano-sized
[177] Raghuvanshi RS, Katare YK, Lalwani K, Ali MM, Singh O, Panda AK. Improved particles and molecules as imaging agents: considerations and caveats.
immune response from biodegradable polymer particles entrapping tetanus Nanomedicine 2008;3:70317.
toxoid by use of different immunization protocol and adjuvants. International [192] Soo Choi H, Liu W, Misra P, Tanaka E, Zimmer JP, Itty Ipe B, et al. Renal clearance
Journal of Pharmaceutics 2002;245:10921. of quantum dots. Nature Biotechnology 2007;25:116570.
[178] Aggarwal P, Hall JB, McLeland CB, Dobrovolskaia MA, McNeil SE. Nanoparti- [193] Ohlson M, Srensson J, Haraldsson B. A gel-membrane model of glomerular
cle interaction with plasma proteins as it relates to particle biodistribution, charge and size selectivity in series. American Journal of Physiology Renal
biocompatibility and therapeutic efcacy. Advanced Drug Delivery Reviews Physiology 2001;280:F396405.
2009;61:42837. [194] William MD, Matthew JL, Bryan DM. Structural determinants of glomeru-
[179] Nel AE, Mdler L, Velegol D, Xia T, Hoek EM, Somasundaran P, et al. Under- lar permeability. American Journal of Physiology Renal Physiology
standing biophysicochemical interactions at the nano-bio interface. Nature 2001;281:57996.
Materials 2009;8:54357. [195] Almeida JPM, Chen AL, Foster A, Drezek R. In vivo biodistribution of nanopar-
[180] Moon JJ, Suh H, Li AV, Ockenhouse CF, Yadava A, Irvine DJ. Enhancing humoral ticles. Nanomedicine 2011;6:81535.
responses to a malaria antigen with nanoparticle vaccines that expand Tfh [196] Arora S, Rajwade JM, Paknikar KM. Nanotoxicology and in vitro studies:
cells and promote germinal center induction. Proceedings of the National the need of the hour. Toxicology and Applied Pharmacology 2012;258:
Academy of Sciences 2012;109:10805. 15165.
[181] Reddy ST, van der Vlies AJ, Simeoni E, Angeli V, Randolph GJ, ONeill CP, et al. [197] He C, Hu Y, Yin L, Tang C, Yin C. Effects of particle size and surface charge on
Exploiting lymphatic transport and complement activation in nanoparticle cellular uptake and biodistribution of polymeric nanoparticles. Biomaterials
vaccines. Nature Biotechnology 2007;25:115964. 2010;31:365766.
[182] Seubert A, Monaci E, Pizza M, OHagan DT, Wack A. The adjuvants aluminum [198] Huang X, Li L, Liu T, Hao N, Liu H, Chen D, et al. The shape effect of mesoporous
hydroxide and MF59 induce monocyte and granulocyte chemoattractants and silica nanoparticles on biodistribution, clearance, and biocompatibility in
enhance monocyte differentiation toward dendritic cells. Journal of Immunol- vivo. ACS Nano 2011;5:53909.
ogy 2008;180:540212. [199] Sulek S, Mammadov B, Mahcicek DI, Sozeri H, Atalar E, Tekinay AB, et al.
[183] Oussoren C, Storm G. Lymphatic uptake and biodistribution of liposomes Peptide functionalized superparamagnetic iron oxide nanoparticles as MRI
after subcutaneous injection. 3. Inuence of surface modication with contrast agents. Journal of Materials Chemistry 2011;21:1515762.
poly(ethyleneglycol). Pharmaceutical Research 1997;14:147984. [200] Lee H, Lee E, Kim DK, Jang NK, Jeong YY, Jon S. Antibiofouling polymer-coated
[184] Swartz MA. The physiology of the lymphatic system. Advanced Drug Delivery superparamagnetic iron oxide nanoparticles as potential magnetic resonance
Reviews 2001;50:320. contrast agents for in vivo cancer imaging. Journal of the American Chemical
[185] Cubas R, Zhang S, Kwon SK, Sevick-Muraca EM, Li M, Chen CY, et al. Society 2006;128:73839.
Virus-like particle (VLP) lymphatic trafcking and immune response gen- [201] Sun G, Xu J, Hagooly A, Rossin R, Li Z, Moore DA, et al. Strategies for optimized
eration after immunization by different routes. Journal of Immunotherapy radiolabeling of nanoparticles for in vivo PET imaging. Advanced Materials
2009;32:11828. 2007;19:315762.
[186] Dane KY, Nembrini C, Tomei AA, Eby JK, ONeil CP, Velluto D, et al. Nano- [202] Wang K, He X, Yang X, Shi H. Functionalized silica nanoparticles: a platform for
sized drug-loaded micelles deliver payload to lymph node immune cells and uorescence imaging at the cell and small animal levels. Accounts of Chemical
prolong allograft survival. Journal of Controlled Release 2011;156:15460. Research 2013;46:136776.
[187] Fis T, Gamvrellis A, Crimeen-Irwin B, Pietersz GA, Li J, Mottram PL, et al. [203] Judenhofer MS, Wehrl HF, Newport DF, Catana C, Siegel SB, Becker M, et al.
Size-dependent immunogenicity: therapeutic and protective properties of Simultaneous PET-MRI: a new approach for functional and morphological
nano-vaccines against tumors. Journal of Immunology 2004;173:314854. imaging. Nature Medicine 2008;14:45965.
[188] De Temmerman M-L, Rejman J, Demeester J, Irvine DJ, Gander B, De Smedt SC. [204] Hafner AM, Corthsy B, Merkle HP. Particulate formulations for the deliv-
Particulate vaccines: on the quest for optimal delivery and immune response. ery of poly(I:C) as vaccine adjuvant. Advanced Drug Delivery Reviews
Drug Discovery Today 2011;16:56982. 2013;65:138699.
[189] Manolova V, Flace A, Bauer M, Schwarz K, Saudan P, Bachmann MF. Nanoparti- [205] Zhao CX, He LZ, Qiao SZ, Middelberg APJ. Nanoparticle synthesis in microre-
cles target distinct dendritic cell populations according to their size. European actors. Chemical Engineering Science 2011;66:146379.
Journal of Immunology 2008;38:140413.

All in-text references underlined in blue are linked to publications on ResearchGate, letting you access and read them immediately.

Potrebbero piacerti anche