Recent advances in peptide-based subunit

1
2
3
4
5
6
Recent advances in peptide-based subunit nanovaccines.
7
Abstract
8
Vaccination is the most efficient way to protect humans against pathogens. Peptide-
9
based vaccines offer several advantages over classical vaccines, which utilized whole
10
organisms or proteins. However, peptides alone are not immunogenic and need a delivery
11
system which can boost their recognition by the immune system. In recent years,
12
nanotechnology-based approaches have become one of the most promising strategies in
13
peptide vaccine delivery. This review summarizes knowledge on peptide vaccines and
14
nanotechnology-based approaches for their delivery. The recently reported nano-sized
15
delivery platforms for peptide antigens are reviewed, including nanoparticles composed of
16
polymers, peptides, lipids, inorganic materials and nanotubes. The future prospects for
17
peptide-based nanovaccines are discussed.
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
Keywords
36
Adjuvant, peptide vaccine, vaccine delivery, nanoparticles, polymer, lipids, self-assembly,
37
macromolecules, dendrimers, nanotechnology
38
39
Introduction
40
The introduction of a vaccine for human treatment was one of the most
41
revolutionizing discoveries in health care. While Edward Jenner and Louis Pasteur are
42
considered as the fathers of vaccination, the first vaccination attempt reaches back hundreds
43
years, when the first small pox inoculations were applied in China. Considering that until the
44
18th century smallpox caused about 10% of global mortalities in Europe, the success of
45
vaccine against this disease can be only compared with the introduction of penicillin.
46
Vaccinology has changed greatly since its early development but the classical vaccine
47
strategy based on attenuated or inactivated pathogens is still used. Problems associated with
48
conventional vaccines include the risk of infection, especially in the case of immune
49
compromised humans, difficulties and impurities associated with the production of pathogens
50
in vitro, and instability of the biological material. Therefore, there is increasing interest in
51
development of vaccines which use only minimal components from pathogens. Such vaccines
52
can be based on recombinant proteins or even minimal fragments carrying immunological
53
information from this protein, namely peptide epitopes.
54
Vaccine efficacy is largely dependent on its biochemical composition, which
55
predominantly includes antigen and immunostimulator (adjuvant). However, recently it has
56
been shown that morphological properties and particle size of the antigen/adjuvant system
57
play a major role in a vaccine’s ability to induce the desired immune responses. Therefore,
58
development of nanovaccines has been growing extensively in recent years [1-4].
59
Nanomaterials, which are usually defined as structures that have at least one size of 1-100 nm
60
dimension (according to American Chemistry Council-Nanotechnology Panel), have started
61
to be widely used for vaccine development. Such materials can be composed of polymers,
62
lipids, peptides, or inorganic constituents. This review summarizes the latest advances (with
63
special focus on the last five years) in delivery of peptide-based vaccines using nanomaterials
64
as carriers, as well as self-assembly delivery systems which are produced by self-organization
65
of appropriately modified peptide antigens. Most of the historical data as well as the study on
66
the use of nanoadjuvants such as Iscomatrix and MF59 have been reviewed elsewhere [1, 3,
67
5-9]. In this review, following the common understanding existing in the published literature,
68
we are defining nanovaccine as immunogenic nanomaterial including any particles with sizes
69
that do not exceed 1 micrometer.
70
71
Figure 1. Simplified diagram of the immune response to nanoparticles (or pathogens).
72
Antigen presenting cells (APCs) are major components of innate immunity. APCs recognize
73
uptake by the endocytosis or phagocytosis process and display antigen. The antigen then is
74
presented to the adaptive immune system and with the help of T-helper cells, appropriate
75
humoral or cellular responses are induced.
76
77
78
79
80
Immune response
81
Vaccines are designed to induce an adaptive immune response; cellular and/or
82
humoral responses. In general, antigen presenting cells (APCs), including dendritic cells
83
(DCs), are parts of an innate immune system and are positioned at the first line of
84
pathogen/vaccine recognition. Antigen can be recognized by DCs localized in peripheral
85
tissue and then transported to the lymph nodes or can travel independently to lymph nodes
86
where they are taken up by lymph node-resident DCs. DCs stimulate T-cells to respond to the
87
antigen by sensing immunogens usually through pattern recognition receptors (PRRs) which
88
recognize pathogen components. Examples of PRRs are Toll like receptors (TLRs) 1 to 13
89
[10] and mannose receptors [11]. The TLR family of receptors recognize a variety of
90
bacterial and viral molecules including free DNA, lipoprotein, lipopolysaccharide, flagellin,
91
etc [12]. Following recognition by PRRs on DCs, pathogen/antigen is taken up. The
92
mechanism of uptake is size-dependent (e.g. nanoparticles (<150 nm) are usually taken up by
93
clathrin-mediated endocytosis, while microparticles are taken up by phagocytosis) which
94
partially explains size-dependent immunogenicity of particles [9]. The antigen is processed
95
inside the APCs, and loaded onto major histocompatibility complex (MHC) class-1 or MHC
96
class-2 (Figure 1). Exogenous particles, toxins, or pathogens are usually endocytosed or
97
phagocytosed and processed into small antigens which are loaded inside vesicles on MHC
98
class-2 molecules. MHC class-2 presentation leads to activation of T-helper cells which
99
further stimulate antibody production or cellular immunity. The MHC-1 pathway, required
100
for production of cellular immunity, is activated through the processing of endogenous
101
antigen presented in the cytosol. However, the production of immune responses through
102
vaccination requires induction of the MHC-1 pathway through exogenous antigen. This
103
process, known as cross-presentation, includes uptake, processing and presentation via MHC
104
class-1 molecules of external antigen. It is not well understood but generally it is believed
105
that exogenous antigen is transported via phagocytosis to the cytosol where it can be
106
processed in the usual manner for endogenous antigens [13]. However, direct delivery of
107
antigen to the cytosol (e.g. with the help of fusogenic liposome) or endosomal escape of
108
antigen (e.g. in a virus-like manner) cannot be ruled out for some antigen delivery platforms.
109
Finally, alongside antigen presentation, signaling protein (cytokines) production is stimulated
110
and adaptive immunity is induced with the help of T-helper cells. Recognition of antigen on
111
MHC class-1 by T-helper cells subtype 1 is primarily responsible for activating and
112
regulating the development of cytotoxic T-lymphocytes (CTLs, CD8+ T-cells). T-helper cells
113
subtype 2 (Th2) favor humoral response (B-cell activation and antibody production).
114
Humoral immune responses are usually targeted to extracellular or intracellular pathogens
115
during or before infection. For example, a vaccine against human papilloma virus (HPV) was
116
developed using virus major capsid protein and thus targeting the virus in the pre-infection
117
stage [14]. Cellular immune responses are responsible for destroying already infected or
118
abnormal human cells. Therefore, in vaccine development this type of immunity is needed to
119
be induced against intracellular pathogen or tumors.
120
121
Peptide-based subunit vaccine
122
A peptide-based subunit vaccine is defined as a vaccine which contains only the
123
peptide component, derived mainly from bacterial, viral or parasite protein, necessary to
124
stimulate appropriate immune responses [15]. Its minimalistic composition is associated with
125
several benefits over the use of whole pathogenic microorganisms or protein. However,
126
removal of vast numbers of components typical for a pathogen (known also as “danger
127
signal”) brings significant reduction in vaccine efficacy and additional additives are required
128
to counteract this problem [16].
129
The major advantages of peptide-based vaccines are as follows:
130
1.
131
132
the use of microorganisms;
2.
133
134
they are non-infectious: cannot revert to virulent state, their production does not include
some pathogens are problematic to culture (e.g. sporozoites for malaria vaccines), and a
subunit-based vaccine (including peptide) might be the only solution in such cases;
3.
135
they do not possess redundant components, which significantly reduce the risk of allergic
or autoimmune responses;
136
4.
they can be designed (customized) to recognize certain pathogen-associated targets;
137
5.
they might be especially useful for development of anticancer vaccines in cases where
138
whole protein cannot be used due to its similarity to endogenous human protein or
139
carcinogenic properties;
140
141
6.
they can include several peptide epitopes targeting different stages in the life cycle or
subtypes of a pathogen;
142
7.
143
144
they can be easily produced, using solid phase peptide synthesis (SPPS), in a pure state,
in a highly reproducible manner, economically and in large scale; and
8.
145
are generally water-soluble, stable under storage conditions even at room temperature,
and can be freeze dried.
146
The major disadvantages of peptide-based vaccines are as follows:
147
1.
they require the use of an immunostimulant (adjuvant) to trigger the desired immune
148
response. Currently available experimental adjuvants suffer from side-toxicity, while
149
commercially available (safe for human) adjuvants are mostly limited to aluminum
150
derivatives that have limited potency in stimulating humoral immune responses and are
151
not effective at inducing cellular immunity; and
152
153
2.
they often lack a T-helper epitope that needs to be incorporated for optimal vaccine
efficacy.
154
155
Thus, in the peptide-based vaccine significant reduction of side effects and production
156
difficulties has been made at the cost of general vaccine efficacy (Figure 2). Finally, it is
157
necessary to take into account that protein-based vaccination can be similar or even more
158
valuable depending on the circumstances. Development of peptide vaccines is usually
159
considered in situations where the recombinant protein-based approach is unproductive. More
160
information on development of peptides as vaccine components can be found in recent
161
reviews [12, 15-17].
162
163
164
165
Figure 2. Vaccines progression - from whole pathogen to nanoparticles. Antigens and their
166
properties; (A) whole pathogen, (B) protein, (C) peptide, and (D) nanoparticles incorporating
167
peptide epitopes (peptides can be both presented on particle surface and/or encapsulated) .
168
169
170
Nanotechnology
171
The nanotechnology-based approach is considered to be one of the most advantageous
172
for development of peptide-based vaccines. Nano-sized vaccines are produced based on
173
nanomaterials with properties as described in the introduction. Such nanoparticles can be
174
built from inert (non-immunogenic) material, in/on which antigen is incorporated or from
175
appropriately modified antigen, which can self-assemble to form nanoparticles [18, 19].
176
Additional immunostimulant or PRR-targeting moieties can be incorporated in their structure.
177
The major advantages of nanovaccines include:
178
1. enhanced uptake by APCs:
179
-
180
181
182
size driven uptake (usually smaller particle are more easily uptaken and therefore are
more immunogenic)
-
cationic particles are more effectively uptaken into macrophages and DCs (due to the
attraction to negatively charged APC cell membranes);
183
2. larger particles can form a depot effect, that is, they retain the antigen at the injection site
184
and in this manner increase the time of vaccine exposure to the immune cells (however, it
185
is necessary to indicate that the depot effect is usually associated with micro rather than
186
nanoparticles);
187
188
189
190
191
192
193
194
195
196
3. particulate vaccines can potentially cross-present antigen (via MHC class-1). Antigen
cross-presentation is especially important to induce CD8+ T-cell immune responses;
4. particles might be covered by multiple copies of the same peptide antigen, mimicking
natural pathogen antigen recurrence;
5. antigens formulated into particles are also at least partially protected against enzymatic
degradation, which is an important issue for highly susceptible peptide antigens;
6. small nanoparticles can easy travel to lymph nodes (without participation of peripheral
DCs), and the nodes are the fighting core of the human immune system;
7. immunological properties of nanoparticles can be altered by changing their size, surface
charge, hydrophobicity, shape, etc.
197
198
Polymer-based nanoparticles
199
A polymer-based drug delivery system is one of the most dynamically growing fields
200
of research. Taking into account that the first polymeric drugs have been approved for human
201
treatment[20], this class of compounds have started to become very attractive from a
202
commercial point of view. Polymeric nanoparticles are usually stable in vivo but also may
203
have biodegradable properties; can protect incorporated antigen from metabolism and
204
elimination; their size, charge and hydrophobicity can be easily altered; and they usually have
205
low or no toxicity [21]. They can be used to form a depot effect to improve vaccine efficacy
206
via elongated exposure/release of antigen at the site of vaccine injection. Such factors as the
207
speed of polymer biodegradation and its shelf-life, rate of antigen release, loading capacity
208
and antigen stability during this loading can be controlled through the choice of polymer and
209
process of antigen incorporation.
210
The pioneering study in the use of polymer nanoparticles for peptide vaccine delivery
211
was performed by Plebanski and co-workers [22]. They showed that polystyrene
212
nanoparticles loaded with ovalbumin (OVA) derived peptide epitopes induced immune
213
responses in a size-dependent manner without the need of additional stimulation with an
214
adjuvant. Among tested particles with a variety of sizes (20, 40, 100, 200, 500, 1000 and
215
2000 nm), 40 nm particles induced the strongest cellular and humoral immunity. Covalent
216
linkage of the peptide was necessary for particle efficacy and therefore nanoparticles served
217
as the delivery system with self-adjuvanting properties rather than as a classical adjuvant, that
218
is, a physical mixture of polystyrene beads and the epitope was not effective. The induction
219
of stronger immune responses by 40-50 nm nanoparticles was later correlated with
220
preferential uptake of these nanoparticles by DCs [23]. It has been also shown using
221
polyhydroxylated nanoparticles of different sizes, that small nanoparticles (25 nm) are
222
capable of trafficking to lymph nodes by themselves and therefore induce stronger immune
223
responses than their larger counterparts [24, 25].
224
One of the most commonly used biodegradable polymer for drug delivery is poly(D,L-
225
lactic-co-glycolide) (PLGA) [26]. This polymer is often used as a first choice for polymeric
226
vaccine delivery systems mainly due to its excellent safety profile and established use in
227
commercial products for controlled delivery of peptide-based drugs [27]. Zhang et al. loaded
228
PLGA nanoparticles (80 ± 27 nm) prepared using the double emulsion method with tumor
229
associated peptide antigens (hgp10025-33 or TRP2180-188) [28]. The nanoparticles were
230
efficiently uptaken by murine DCs and induced stronger cellular immune responses in the
231
mouse model than the peptides mixed with Freund’s adjuvant. Both complete Freund’s
232
adjuvant (CFA) and incomplete Freund’s adjuvant (IFA) are commonly used as the “gold”
233
standard for stimulation of immune responses against peptide-based antigens; however, they
234
are not allowed for human use (particularly because CFA has shown high toxicity).
235
Nanoparticles formulated with TRP2180-188 were able to significantly reduce tumor growth in
236
mice following prophylactic subcutaneous immunization (mice were immunized trice prior to
237
a melanoma cells injection). Similarly, PLGA nanoparticles with a diameter of 215 and 330
238
nm loaded with tumor associated peptide antigen were able to stimulate cellular immunity
239
[29, 30]. To improve the efficacy of PLGA nanoparticles, several additives to the basic
240
nanoparticle formulation were tested. One of the approaches was designed to target human
241
follicle-associated epithelium derived M-cells, which are responsible of internalizing luminal
242
antigen and delivering it to lymphoid tissue [31]. Peptides targeting M-cells were conjugated
243
to PLGA nanoparticles and subsequently showed improved transport of antigen-loaded
244
nanoparticles across the intestinal mucosal barrier [32]. In other studies, Messmer and co-
245
workers conjugated DCs inducing peptide (Hp91) to PLGA and demonstrated that this
246
construct formulated into particles (~ 200 nm) activated both human and mouse DCs more
247
efficiently than peptide alone [33]. Lipid (1,2-dioleoyl-sn-glycero-3-phosphocholine) coated
248
PLGA nanoparticles (with diameters of 100 nm but smaller particles were also observed by
249
TEM) were studied [34]. Interestingly, a mixture of nanoparticles incorporating several tumor
250
associated antigens showed reduced stimulation of T-cells (assessed by IFN-γ production) but
251
improved prophylactic antitumor effect in mice when compared to any other nanoparticle-
252
bearing single antigen. It was suggested that improved antitumor efficacy was related to
253
reduction of the risk of tumor escape as the host immune system attacked multiple targets
254
simultaneously. PLGA nanoparticles have been recently used to generate immune responses
255
against tetanus and diphtheria toxoid and universal memory T-cell helper peptide, active in
256
vitro in human and in vivo in non-human primates, was developed [35]. PLGA nanoparticles
257
were also tested as a peptide-based vaccine candidate against Chlamydia trachomatis [36].
258
Chitosan is a chitin derived natural cationic polymer with adjuvanting properties [19].
259
It is recognized by cell surface receptors including macrophage mannose receptors and TLR-
260
2 [37]. Jackson and co-workers studied chitosan-based nano- and microparticles for delivery
261
of luteinizing hormone-releasing hormone (LHRH) as a peptide antigen [38]. They
262
demonstrated that antigen was mostly localized on the surface of chitosan particles.
263
Confirming previous observations with polyhydroxylated nanoparticles, the nanoparticles (~
264
200 nm) travelled from the injection site to the draining lymph nodes faster than
265
microparticles (~ 2 µm). However, no significant difference in antibody production was
266
observed for both types of particles after subcutaneous immunization in mice. Another
267
commonly used polymer for drug delivery is poly glutamic acid (PGA) which is
268
biodegradable, highly water-soluble, non-toxic and non-immunogenic [39]. Tumor specific
269
peptide antigen (EphA2 peptide), conjugated to PGA nanoparticles grafted with phenyl
270
alanine (246 ± 88 nm), demonstrated activity against liver tumor similar to that of the peptide
271
mixed with toxic CFA (which induced liver damage), but did not show any toxic side-effects
272
[40].
273
Recently nano-self-assembling strategies are receiving growing recognition in
274
biomedical fields [18] and it has been suggested that self-assembling amphiphilic polymers
275
might be useful systems for development of subunit vaccines [41]. To prove this concept,
276
Toth and co-workers applied a non-toxic tert-butyl polyacrylate as an dendrimer core and
277
chemically conjugated it with multiple copies of Group A Streptococcus (GAS) B-cell
278
epitope [42]. The produced construct was self-assembled to form 20 nm nanoparticles, which
279
were able to induce the desired helical conformation of attached peptides and elicit high
280
levels of antigen-specific antibodies without the aid of an adjuvant. These nanoparticles were
281
effective when administered via subcutaneous or intranasal routes and were also capable of in
282
vitro opsonization of GAS [43]. Furthermore, it was proved that smaller nanoparticles (~ 20
283
nm) were more immunogenic than larger ones (~ 500 nm) even after single immunization
284
[44]. Interestingly, when cervical cancer associated peptide epitopes were conjugated to
285
branched tert-butyl polyacrylate, nanoparticles as well as microparticles (depending on the
286
peptide structures) were formed in water. When the same conjugates were formulated in PBS
287
buffer all of them aggregated into large microparticles. Despite their large size, these particles
288
were able to reduce tumor growth in a therapeutical setup (vaccine treated existing tumor)
289
and even eradicate a model of cervical tumor in mice after a single immunization, without the
290
help of any external adjuvant [45]. In another approach, tumor-associated MUC1 peptide as
291
the B-cell epitope and a T-helper cell epitope, with or without a lipophilic unit (lauryl
292
methacrylate) were assembled on poly(N-(2-hydroxypropyl)methacrylamide) to form linear
293
polymeric amphiphiles with self-assembling properties [46]. The formed nanoparticles were
294
able to induce strong humoral immune responses only when mixed with CFA, consistently
295
with an older study, which used epitope polymerization technique based on the formation of
296
linear polyacrylate [47].
297
298
Lipid-based nanoparticles
299
Lipid carriers have been studied extensively for vaccine delivery and liposomes are
300
one of the most widely used lipid-based vaccine delivery vehicles [48, 49]. Surprisingly,
301
liposomes have been rarely used for peptide-based nanovaccine delivery. In a recent study,
302
multiepitope peptides from the rat HER2/neu oncogene were incorporated into liposome-
303
polycations with CpG oligonucleotides adjuvant (LPD) nanoparticles (~150 nm) [50]. Lead
304
liposomal formulation (with p5 peptide) was able to completely protect mice in a
305
prophylactic TUBO tumor model (overexpressing the rHER2/neu protein) challenge. In
306
another approach, highly conserved influenza-derived peptides were encapsulated into
307
liposomes (30 – 100 nm) with monophosphoryl lipid A (MPL) and trehalose 6,6’-dimycolate
308
as adjuvants [51]. While the peptides alone were practically non-effective, a liposomal
309
formulation was able to induce protective immune responses after intranasal administration
310
against a lethal influenza challenge in mice. The immune responses were T-cell dependent
311
with macrophages playing a major role (rather than DCs) in response induction.
312
Unfortunately, both the above liposomal strategies required the use of an adjuvant in the
313
formulation. A more popular lipid-based strategy used lipidation of peptide antigens to form
314
amphiphiles, which were self-assembled into nanoparticles. During study on the conserved
315
peptide epitope-based vaccine against GAS, it was demonstrated that the balance between
316
hydrophilic and hydrophobic properties of individual segments of such lipopeptides was
317
responsible for the size of formed particles and the more polar peptide epitopes attached to
318
the lipid core produced smaller nanoparticles [52]. In this approach the lipid peptide core
319
(LCP) strategy was used, in which unnatural lipidic amino acids (amino acids with long
320
aliphatic side chains) were conjugated via the branching moiety (based on polylysine,
321
carbohydrate, etc) to the desired peptide epitopes [53, 54]. In the LCP, lipid moieties served
322
as a hydrophobic core to allow self-assembly and act as a self-adjuvanting moiety with TLR-
323
2 agonist properties [54]. When multiple copies of GAS-derived B-cell epitopes (J14) were
324
incorporated into LCP constructs, large nanoparticles were formed (200-1000 nm) that
325
induced rather moderate B-cell response in comparison to the CFA-based control [55]. In
326
contrast, an LCP construct possessing modified J14 epitope (dJ14i), when self-assembled into
327
small nanoparticles (15-20 nm), was able to induce the same level of anti-dJ14i IgG titers as
328
the peptide formed with CFA when administered subcutaneously in mice [56]. However,
329
heterogeneous size distribution of nanoparticles with no clear size-dependent immune
330
responses were also reported for a variety of LCP-based vaccine candidates [57]. Robinson
331
and coworkers used lipopeptides to form self-assembled homogenous nanoparticles (20-25
332
nm) which were able to induce strong humoral immunity with or without the use of CFA [58,
333
59]. They also demonstrated that DCs used multiple endocytic routes even for uptake of
334
small nanoparticles. While the above particles were taken up mainly by macropinocytosis,
335
clathrin independent uptake was also observed [60].
336
337
Self-assembled peptide
338
The ability of certain peptides to self-assemble into particlse or fibrils is a well-known
339
phenomenon and peptide self-assembly has been used for biomedical purposes [61]. Peptide
340
self-assembled nanomaterials are biologically compatible, multifunctional, multivalent, well-
341
chemically defined, usually low or non-toxic and the position of attachment of an antigen can
342
be well controlled. Collier and co-workers have been intensively studying a vaccine delivery
343
system based on β-sheet forming Q11 peptide. Several different peptide epitopes were
344
conjugated to this peptide and self-assembled into fibrils (5-15 nm thick) [62-65]. They
345
observed strong humoral responses in mice when OVA peptide epitopes were covalently
346
bonded to Q11, however, a relatively large quantity of immunogen was required to induce
347
production of high antibody titers (0.3 mg per injection) [65]. They demonstrated that two
348
conjugates, incorporating Q11, linked with two single malaria-related peptide antigens can be
349
co-assembled together to produce an immune response without help of adjuvant through the
350
MyD88 pathway but without participation of TLR-2 and TLR-5 [64]. The fibres induced
351
immune responses with the help of CD4+ cells, were non-toxic and did not induce
352
inflammation [63]. When OVA-derived CTL epitope was conjugated to Q11, the formed
353
fibrils elicited robust CD8+ T-cell responses [62]. Toth and co-workers demonstrated that
354
such peptide antigen-bound fibrils can be formed upon request from non-fibrilizing
355
precursors using an isopeptide strategy. Stable in solid form, O-acyl isopeptide (ester isomer
356
of original peptide) showed high aqueous solubility and released native peptide through
357
physiological pH-triggered O-N acyl migration reaction with simultaneous fibril formation.
358
They claimed that this strategy can overcome potential problems related to over-aggregation,
359
precipitation, and changes in other properties during storage of fibril-based vaccines [66].
360
Burkhard and co-workers previously demonstrated that peptides which possess coil-
361
coil conformation were able to aggregate and upon conjugation with malaria peptide epitope
362
form nanoparticles (~25 nm). These nanoparticles induced protective immunity in mice in a
363
malaria challenge experiment [67]. Recently, they incorporated into their delivery system
364
tumor targeting moiety (bombesin) and formed nanoparticles (33-36 nm) [68]. While these
365
particles did not demonstrate tumor targeting properties, their spleen uptake was significantly
366
increased, proportionally to the increasing level of the bombesin in the particles. As spleen is
367
a primary organ of the immune system, it was suggested that such particles can be used for
368
design of vaccine candidates with improved efficacy. When this delivery system incorporated
369
CD8+ epitope from Toxoplasma gondii, it was able to self-assemble into ∼38 nm
370
nanoparticles and induce strong cellular immunity (assessed by IFN-γ production) [69]. The
371
nanoparticle was also able to reduce T. gondii parasite burden in vivo.
372
373
Inorganic nanoparticles and nanotubes
374
Nanoparticles built from inorganic material such as a gold or ferric oxide have
375
recently become attractive drug delivery vehicles [70]. They have unique physicochemical
376
properties such as porous structures, facile surface functionalization with a variety of ligands,
377
and their size and shape can be controlled. Interestingly, commercially available alum
378
adjuvant (which can form inorganic nanoparticles) was found to be safe and an effective
379
immunostimulant for whole pathogen or protein-based vaccine delivery; however, its
380
adjuvanting properties are generally too mild for stimulation of immune responses against
381
peptide antigen [71]. To overcome alum pure immunostimulatory activity, Neutra and
382
coworkers conjugated peptide epitopes derived from HIV-1 gp120 glycoprotein to the
383
aluminum oxide nanoparticles (~350 nm). These particles were able to stimulate a moderate
384
antibody response after intraperitoneal injection; however, they failed to stimulate mucosal
385
immunity [72, 73]. Further study was discontinued. Huang and co-workers used foot-and-
386
mouth disease virus associated peptide antigen conjugated to several gold
387
nanoparticles with sizes ranging from 2 to 50 nm (2, 5, 8, 12, 17, 37, 50 nm) [74]. The
388
highest antibody titers were observed for mice immunized with 8 nm nanoparticles,
389
while the 37 and 50 nm were ineffective. Generally, 2-17 nm particles induce strong
390
humoral response. The highest spleen uptake was observed for nanoparticles with size
391
12 nm while uptake was also high for particles of size 8-50 nm. Larger particles, which
392
are more easily endocytosed, were absorbed at the injection site and therefore their
393
concentration in the circulation (blood) was low. As size-dependant spleen uptake of
394
nanoparticles was similar to their efficacy profile, it was suggested that the ability of particles
395
to travel and accumulate in the spleen was crucial to induce immunity. Baneyx and co-
396
workers applied calcium phosphate to form peptide antigen-coated nanoparticles (50-70 nm)
397
which showed the ability to induce humoral immunity in mice [75]. In another study, calcium
398
carbonate nanoparticles were coated with polylysine and polyglutamic acid based on opposite
399
charge attraction [76]. During coating process, OVA and influenza peptide epitope were also
400
incorporated to form nanoparticles with diameter of ~ 250 nm and ~150 nm, respectively.
401
These nanoparticles were able to induce both humoral and cellular immunity after a single
402
injection in mice without the help of an adjuvant. Importantly, no immune responses to the
403
matrix components were detected.
404
The recent discoveries of carbon nanotubes as a drug delivery agent [77, 78] triggered
405
interest in developing this nanomaterial for vaccine delivery purposes [79, 80]. Early attempts
406
have shown that peptides conjugated to nanotubes were able to induce high titres of antibody
407
when CFA was used as an adjuvant [81, 82]. More recently, Villa et al. demonstrated that
408
peptide derived from Wilm's tumor protein conjugated to nanotubes of high length variability
409
could be rapidly internalized into APCs, and induced humoral immunity; however, external
410
adjuvant was still necessary for nanotube efficacy [83]. These data suggested that carbon
411
nanotubes are a rather poor immunostimulator for peptide-based vaccines.
412
413
414
Conclusion
Nanomaterial-based approaches for peptide vaccine development are clearly of high
415
importance in current vaccine delivery research. Particle size of these peptide vaccines plays
416
an important role for their immunostimulatory properties. Interestingly, size-dependent
417
activity is not as consistent as can be expected with different groups reporting different
418
optimal size for vaccine formulation. This phenomenon can be explained by the differences
419
in measurement techniques which are often determining diverse sizes of the same particles
420
(e.g. dynamic light scattering (DLS) measure hydrodynamic size, while transmission electron
421
microscopy (TEM) is showing the size of nanoparticles after drying and only the part which
422
efficiently absorbs light is visible). Particle size distribution also can vary significantly and
423
the immune system does not produce responses against single particle size but always against
424
a whole range of sizes present in the vaccine formulation. For simplicity however, dominant
425
size or “peak” size is usually reported. In addition, particles may differ not only in size but
426
also in (a) antigen loading; (b) level of antigen absorption on the surface against
427
encapsulated, and different loading methods may incorporate antigen into/onto nanoparticles
428
in different ways; (c) the nature of the composition material; (d) the ability of the antigen to
429
be released from nanoparticles; and (e) the level of its protection against biodegradation.
430
Moreover, dose and dosing frequencies differ between studies and the route of administration
431
can have major impact on vaccine efficacy. However, the message from most of the current
432
studies is clear; size plays an important role in vaccine efficacy. Smaller particles are more
433
immunogenic due to their easier uptake by DCs and their efficient transport in the lymphatic
434
system; however, large particles including microparticles can form a stable depot and in this
435
manner induce strong immune responses as well.
436
The antigen is often chemically conjugated into/onto nanoparticles, or the particles are
437
formed from self-assembled antigen-carrier conjugates; such stable composition ensures
438
delivery of the adjuvanting moieties and antigen to the same APCs. This limits systemic
439
distribution of adjuvant and its concentration required to boost immunity, therefore limiting
440
toxicity of the vaccine. Moreover, vaccines are not administered in a repetitive manner to the
441
host; therefore, the risk of excessive accumulation in the body of even relatively stable
442
particles is low. Nevertheless, nanoparticle-based formulations should undergo strict quality
443
control and such factors like reproducibility of formulation, storage related aggregation, and
444
surface charge changes need to be carefully monitored during production, storage and
445
transportation. This cost is warranted as in return, safe vaccines can be developed and the use
446
of classical adjuvant with their toxic side-effects can be omitted.
447
448
449
450
Future Perspectives
451
progress in nanotechnological approaches for vaccine delivery should overcome many of
452
existing obstacles. Especially, vaccine efficiency can be greatly improved and toxicity
453
reduced using an adjuvant-free nanovaccine strategy. In addition, the only immune adjuvant
454
commonly used for humans (alum) is not able to stimulate cellular immunity. Stimulation of
455
cellular immune responses has been found crucial for development of vaccines against
456
cancer, malaria, HIV and other intracellular pathogens. Thus, the ability of nanoparticles to
457
induce cellular immunity against incorporated peptide antigens would be of special interest in
458
the field of vaccine development. There are several examples of peptide-based vaccines in
459
clinical trials (e.g. vaccines against GAS or therapeutic vaccines against cervical cancer).
460
Thus, the prospect for commercial success of peptide-based vaccines is substantial and the
461
use of nanotechnology-based approaches can only increase this chance. In addition, many
462
current peptide vaccine delivery platforms have not been analyzed for their ability to form
463
particles, their size dependent immunity, and the influence of morphological properties on
464
their efficacy, but in the near future such analyses are expected to become standard in
465
peptide-based vaccine development. It is not anticipated that just one single size will be
466
found to be optimal for all vaccine deliveries; rather each delivery system and antigen will
467
have its unique optimal size and other properties (such as charge, shape etc.) and therefore,
There is no example of a peptide-based vaccine in the market so far; however, recent
468
each system will need to be optimized separately. Moreover, the use of a mixture of different
469
sizes might be advantageous in some cases (e.g, for the same antigen the stable depot with be
470
formed with large particles while at the same time small nanoparticles will be used to target
471
the antigen to lymph nodes). In future development, size-dependent toxicity needs to be
472
studied in more detail. Some recent reports have shown that very small cationic particles can
473
have significant toxicity. Thus, vaccine candidates, especially those with broad size
474
distribution might not be as safe as currently claimed. Even in such cases, the immune system
475
is expected to clear those nanoparticles before they can harm the human body. As vaccination
476
remains associated with some toxicity, approaches based on single immunization are
477
particularly advantageous and, as has been shown in this review, such immunization schedule
478
becomes possible with the help of nanoparticles. The use of fully biodegradable carriers is
479
also recommended. In future development, the cost of vaccine production needs to be taken
480
into account as well. For example, approaches for neglected tropical diseases, which are
481
slightly less affective but significantly cheaper, should be endorsed. Finally, in the near future
482
it is expected that nanoparticle-based formulations will not be limited to antigens and
483
immunostimulating moieties but additional functional elements will be incorporated (such as
484
targeting moieties, stabilizing coatings, or mucosal adhesive functionalities).
485
486
Executive Summary
487
Peptide-based subunit vaccine
488

489
490
effects.

491
492
The use of only minimal immunogenic component allows reduction of undesirable side-
Removal of danger signal reduces peptide-based vaccine immunogenicity; therefore,
external adjuvants or special delivery systems have to be used for vaccine efficacy.

Peptide-based vaccine can be relatively easy customized, produced, stored and
493
transported.
494
The nanotechnology
495

496
497
vaccine immunogenicity.

498
499
500
It has been widely accepted that the size of antigen particles plays an important role in
The use of nanoparticles can stimulate better antigen uptake by APCs, protect antigen
from degradation and elimination, and induce antigen cross-presentation to CTLs.

Nanoparticles can be engineered to contain multiple peptide epitopes, self-adjuvanting
moieties and targeting moieties.
501

502
Nanoparticles may mimic natural pathogen through size and display of multiple copies of
surface antigens.
503
Polymer-based nanoparticles
504

Peptide-based antigen can be encapsulated or attached on the surface of polymeric
505
nanoparticles
506
nanoparticles.
507

508
509
while
polymer-peptide
conjugates
can
be
self-assembled
into
Most of the data suggests that small polymer-based nanoparticles (20-50 nm) induce
optimal immune responses.

510
Poly(D,L-lactic-co-glycolide), chitosan, and acrylates are the most commonly used
polymeric carries for peptide vaccines delivery.
511
Lipid-based nanoparticles
512

Lipids have a natural tendency to self-assemble and might be recognized by TLRs.
513

Lipidation of peptides forms amphiphiles which are often able to self-assemble into
514
nanoparticles with self-adjuvanting properties and capacity to induce strong immune
515
responses.
516

517
Size-dependant immunogenicity of lipid-based peptide vaccine has not yet been
comprehensively studied.
518
Self-assembled peptides
519

520
521
Self-assembly properties of certain peptides can be used to form nanoparticle or
nanofibril structures.

522
Self-assembled peptides are fully biodegradable, biocompatible and can induce both
cellular and humoral immune responses without help of an adjuvant.
523
Inorganic nanoparticles and nanotubes
524

525
526
Peptide antigens can be conjugated to inorganic nanoparticles and induce size-dependent
immune responses.

While some studies have suggested optimal efficacy for small nanoparticles (2-17 nm),
527
larger nanoparticles are also effective in inducing immune responses without help of an
528
adjuvant.
529
530
531
532
533

Carbon nanotubes can serve as carriers for peptide-based vaccines but the use of an
adjuvant is still required for their efficacy.
Financial & competing interest disclosures
534
This work was supported by the National Health and Medical Research Council (NHMRC),
535
Australia. The authors have no relevant affiliations or financial involvement with any
536
organizations or entity with a financial interest in or financial conflict with the subject matter
537
or material discussed in the manuscript apart from those disclosed. The contents are solely
538
the responsibility of authors and do not necessarily represent the official views of the
539
NHMRC.
540
541
542
References
543
Papers of special note have been highlighted as:
544
 of interest
545
 of considerable interest
546
547
548
549
550
551
552
553
554
555
556
557
558
559
560
561
562
563
564
565
566
567
568
569
570
571
572
573
574
1.
Skwarczynski M, Toth I: Peptide-Based Subunit Nanovaccines. Curr. Drug Delivery
8(3), 282-289 (2011).
2.
Bachmann MF, Jennings GT: Vaccine delivery: a matter of size, geometry, kinetics
and molecular patterns. Nat. Rev. Immunol. 10, 787-796 (2010).
3.
Oyewumi MO, Kumar A, Cui ZR: Nano-microparticles as immune adjuvants:
correlating particle sizes and the resultant immune responses. Expert Rev. Vaccines
9(9), 1095-1107 (2010).
 Excellent review on relation between particles size and immunity.
4.
Fujita Y, Taguchi H: Current status of multiple antigen-presenting peptide vaccine
systems: Application of organic and inorganic nanoparticles. Chem. Cent. J. 5,
(2011).
5.
Zaman M, Good MF, Toth I: Nanovaccines and their mode of action. Methods 60(3),
226-231 (2013).
6.
Chadwick S, Kriegel C, Amiji M: Nanotechnology solutions for mucosal
immunization. Adv. Drug Delivery Rev. 62, 394–407 (2010).
7.
Nandedkar TD: Nanovaccines: recent developments in vaccination. J. Biosci. 34(6),
995-1003 (2009).
8.
Peek LJ, Middaugh CR, Berkland C: Nanotechnology in vaccine delivery. Adv. Drug
Delivery Rev. 60(8), 915-928 (2008).
9.
Xiang SD, Scholzen A, Minigo G et al.: Pathogen recognition and development of
particulate vaccines: Does size matter? Methods 40(1), 1-9 (2006).
10.
Hussein WM, Liu T-Y, Skwarczynski M, Toth I: Toll-like receptor agonists: a patent
review (2011-2013). Expert Opin. Ther. Pat. 24(4), 453-470 (2014).
11.
Keler T, Ramakrishna V, Fanger MW: Mannose receptor-targeted vaccines. Expert
Opin. Biol. Ther. 4(12), 1953-1962 (2004).
12.
Azmi F, Ahmad Fuaad AaH, Skwarczynski M, Toth I: Recent progress in adjuvant
discovery for peptide-based subunit vaccines. Human Vaccines &
Immunotherapeutics 10(3), 778–796 (2014).
575
576
577
578
579
580
581
582
583
584
585
586
587
588
589
590
591
592
593
594
595
596
597
598
599
600
601
602
603
604
605
606
607
608
609
610
611
612
613
614
615
616
617
618
619
620
621
622
623
13.
Joffre OP, Segura E, Savina A, Amigorena S: Cross-presentation by dendritic cells.
Nat. Rev. Immunol. 12(8), 557-569 (2012).
14.
Liu TY, Hussein WM, Toth I, Skwarczynski M: Advances in peptide-based human
papillomavirus therapeutic vaccines. Curr. Top. Med.Chem. 12, 1581-1592 (2012).
15.
Purcell AW, Mccluskey J, Rossjohn J: More than one reason to rethink the use of
peptides in vaccine design. Nat. Rev. Drug Discovery 6(5), 404-414 (2007).
 Excellent review on peptide-based vaccines
16.
Dudek NL, Perlmutter P, Aguilar MI, Croft NP, Purcell AW: Epitope Discovery and
Their Use in Peptide Based Vaccines. Curr. Pharm. Des. 16(28), 3149-3157 (2010).
17.
Yamada A, Sasada T, Noguchi M, Itoh K: Next-generation peptide vaccines for
advanced cancer. Cancer Sci. 104(1), 15-21 (2013).
18.
Doll TaPF, Raman S, Dey R, Burkhard P: Nanoscale assemblies and their biomedical
applications. J. Royal Soc. Interface 10(80), (2013).
19.
Sahdev P, Ochyl LJ, Moon JJ: Biomaterials for Nanoparticle Vaccine Delivery
Systems. Pharm. Res., (2014).
20.
Mccoy M: Long-term partners. Chem. Eng. News 91(9), 18-19 (2013).
21.
Akagi T, Baba M, Akashi M: Biodegradable Nanoparticles as Vaccine Adjuvants and
Delivery Systems: Regulation of Immune Responses by Nanoparticle-Based Vaccine.
In: Polymers in Nanomedicine, Kunugi S,Yamaoka T (Ed.^(Eds). 31-64 (2012).
22.
Fifis T, Gamvrellis A, Crimeen-Irwin B et al.: Size-dependent immunogenicity:
Therapeutic and protective properties of nano-vaccines against tumors. J. Immunol.
173(5), 3148-3154 (2004).
 Excellent early work on peptide-based nanovaccines.
23.
Mottram PL, Leong D, Crimeen-Irwin B et al.: Type 1 and 2 immunity following
vaccination is influenced by nanoparticle size: Formulation of a model vaccine for
respiratory syncytial virus. Mol. Pharm. 4(1), 73-84 (2007).
24.
Reddy ST, Van Der Vlies AJ, Simeoni E et al.: Exploiting lymphatic transport and
complement activation in nanoparticle vaccines. Nat. Biotechnol. 25(10), 1159-1164
(2007).
25.
Reddy ST, Rehor A, Schmoekel HG, Hubbell JA, Swartz MA: In vivo targeting of
dendritic cells in lymph nodes with poly(propylene sulfide) nanoparticles. J.
Controlled Release 112(1), 26-34 (2006).
26.
Mundargi RC, Babu VR, Rangaswamy V, Patel P, Aminabhavi TM: Nano/micro
technologies for delivering macromolecular therapeutics using poly(D,L-lactide-coglycolide) and its derivatives. J. Controlled Release 125(3), 193-209 (2008).
27.
Jiang WL, Gupta RK, Deshpande MC, Schwendeman SP: Biodegradable poly(lacticco-glycolic acid) microparticles for injectable delivery of vaccine antigens. Adv. Drug
Delivery Rev. 57(3), 391-410 (2005).
28.
Zhang Z, Tongchusak S, Mizukami Y et al.: Induction of anti-tumor cytotoxic T cell
responses through PLGA-nanoparticle mediated antigen delivery. Biomaterials
32(14), 3666-3678 (2011).
29.
Ma W, Smith T, Bogin V et al.: Enhanced presentation of MHC class Ia, Ib and class
II-restricted peptides encapsulated in biodegradable nanoparticles: a promising
strategy for tumor immunotherapy. Journal of Translational Medicine 9, (2011).
30.
Silva AL, Rosalia RA, Sazak A et al.: Optimization of encapsulation of a synthetic
long peptide in PLGA nanoparticles: Low-burst release is crucial for efficient CD8(+)
T cell activation. Eur. J. Pharm. Biopharm. 83(3), 338-345 (2013).
31.
Marasini N, Skwarczynski M, Toth I: Oral delivery of nanoparticle-based vaccines. .
Expert Rev. Vaccines in press, (2014).
624
625
626
627
628
629
630
631
632
633
634
635
636
637
638
639
640
641
642
643
644
645
646
647
648
649
650
651
652
653
654
655
656
657
658
659
660
661
662
663
664
665
666
667
668
669
670
671
672
32.
Fievez V, Plapied L, Plaideau C et al.: In vitro identification of targeting ligands of
human M cells by phage display. Int. J. Pharm. 394(1-2), 35-42 (2010).
33.
Clawson C, Huang C-T, Futalan D et al.: Delivery of a peptide via poly(D,L-lacticco-glycolic) acid nanoparticles enhances its dendritic cell-stimulatory capacity.
Nanomed.-Nanotechnol. Biol. Med. 6(5), 651-661 (2010).
34.
Tan S, Sasada T, Bershteyn A, Yang K, Ioji T, Zhang Z: Combinational delivery of
lipid-enveloped polymeric nanoparticles carrying different peptides for anti-tumor
immunotherapy. Nanomedicine (London, England) 9(5), 635-647 (2014).
 Lipid-coated polymer-based nanoparticles caring several different peptide epitopes showed
significantly stronger antitumor efficacy in contrast to single peptide loaded counterparts.
35.
Fraser CC, H Altreuter D, Ilyinskii P et al.: Generation of a universal CD4 memory T
cell recall peptide effective in humans, mice and non-human primates. Vaccine
32(24), 2896-2903 (2014).
36.
Taha MA, Singh SR, Dennis VA: Biodegradable PLGA85/15 nanoparticles as a
delivery vehicle for Chlamydia trachomatis recombinant MOMP-187 peptide.
Nanotechnology 23(32), (2012).
37.
Li X, Min M, Du N et al.: Chitin, chitosan, and glycated chitosan regulate immune
responses: the novel adjuvants for cancer vaccine. Clin. Dev. Immunol. 2013, 387023
(2013).
38.
Chua BY, Al Kobaisi M, Zeng W, Mainwaring D, Jackson DC: Chitosan
Microparticles and Nanoparticles as Biocompatible Delivery Vehicles for Peptide and
Protein-Based Immunocontraceptive Vaccines. Mol. Pharm. 9(1), 81-90 (2012).
39.
Shih IL, Van YT, Shen MH: Biomedical applications of chemically and
microbiologically synthesized poly(glutamic acid) and poly(lysine). Mini-Rev. Med.
Chem. 4(2), 179-188 (2004).
40.
Yamaguchi S, Tatsumi T, Takehara T et al.: EphA2-derived peptide vaccine with
amphiphilic poly(gamma-glutamic acid) nanoparticles elicits an anti-tumor effect
against mouse liver tumor. Cancer Immunol. Immunother. 59(5), 759-767 (2010).
41.
Akagi T, Baba M, Akashi M: Preparation of nanoparticles by the self-organization of
polymers consisting of hydrophobic and hydrophilic segments: Potential applications.
Polymer 48(23), 6729-6747 (2007).
42.
Skwarczynski M, Zaman M, Urbani CN et al.: Polyacrylate Dendrimer Nanoparticles:
A Self-Adjuvanting Vaccine Delivery System. Angewandte Chemie-International
Edition 49(33), 5742-5745 (2010).
 First example of self-assembling peptide-polymer conjugates for vaccine delivery.
43.
Zaman M, Skwarczynski M, Malcolm JM et al.: Self-adjuvanting polyacrylic
nanoparticulate delivery system for group A streptococcus (GAS) vaccine. Nanomed.Nanotechnol. Biol. Med. 7(2), 168-173 (2011).
44.
Fuaad A, Jia ZF, Zaman M et al.: Polymer-peptide hybrids as a highly immunogenic
single-dose nanovaccine. Nanomedicine 9(1), 35-43 (2014).
45.
Liu T-Y, Hussein WM, Jia Z et al.: Self-Adjuvanting Polymer–Peptide Conjugates As
Therapeutic Vaccine Candidates against Cervical Cancer. Biomacromolecules 14(8),
2798-2806 (2013).
46.
Nuhn L, Hartmann S, Palitzsch B et al.: Water-soluble polymers coupled with
glycopeptide antigens and T-cell epitopes as potential antitumor vaccines. Angew.
Chem. Int. Ed. Engl. 52(40), 10652-10656 (2013).
47.
Brandt ER, Sriprakash KS, Hobb RI et al.: New multi-determinant strategy for a
group A streptococcal vaccine designed for the Australian Aboriginal population. Nat.
Med. 6(4), 455-459 (2000).
673
674
675
676
677
678
679
680
681
682
683
684
685
686
687
688
689
690
691
692
693
694
695
696
697
698
699
700
701
702
703
704
705
706
707
708
709
710
711
712
713
714
715
716
717
718
719
720
48.
Ghaffar KA, Giddam AK, Zaman M, Skwarczynski M, Toth I: Liposomes as
nanovaccine delivery systems. Curr. Top. Med. Chem. 14(9), 1194-1208 (2014).
49.
Giddam AK, Zaman M, Skwarczynski M, Toth I: Liposome-based delivery system
for vaccine candidates: constructing an effective formulation. Nanomedicine 7(12),
1877-1893 (2012).
 Interesting review on liposomes-based vaccine formulation.
50.
51.
52.
53.
54.
55.
56.
57.
58.
59.
60.
61.
62.
63.
Jalali SA, Sankian M, Tavakkol-Afshari J, Jaafari MR: Induction of tumor-specific
immunity by multi-epitope rat HER2/neu-derived peptides encapsulated in LPD
Nanoparticles. Nanomedicine 8(5), 692-701 (2012).
Tai W, Roberts L, Seryshev A et al.: Multistrain influenza protection induced by a
nanoparticulate mucosal immunotherapeutic. Mucosal Immunol. 4(2), 197-207
(2011).
Skwarczynski M, Parhiz BH, Soltani F et al.: Lipid Peptide Core Nanoparticles as
Multivalent Vaccine Candidates against Streptococcus pyogenes. Aust. J. Chem. 65,
35-39 (2012).
Zhong W, Skwarczynski M, Toth I: Lipid Core Peptide System for Gene, Drug, and
Vaccine Delivery. Aust. J. Chem. 62(9), 956-967 (2009).
Skwarczynski M, Zaman M, Toth I: Lipo-peptides/saccharides in peptide vaccine
delivery. . In: Handbook of the Biologically Active Peptides, the 2nd Edition, Kastin A
(Ed.^(Eds). Elsevier Inc, Burlington 571-579 (2013).
Skwarczynski M, Fuaad AaHA, Rustanti L et al.: Group A streptococcal vaccine
candidates based on the conserved conformational epitope from M protein. Drug
Delivery Lett. 1(Copyright (C) 2012 American Chemical Society (ACS). All Rights
Reserved.), 2-8 (2011).
Skwarczynski M, Kamaruzaman KA, Srinivasan S et al.: M-Protein-derived
Conformational Peptide Epitope Vaccine Candidate against Group A Streptococcus.
Curr. Drug Delivery 10(1), 39-45 (2013).
Zaman M, Abdel-Aal A-BM, Fujita Y et al.: Structure-Activity Relationship for the
Development of a Self-Adjuvanting Mucosally Active Lipopeptide Vaccine against
Streptococcus pyogenes. J. Med. Chem. 55(19), 8515-8523 (2012).
Boato F, Thomas RM, Ghasparian A, Freund-Renard A, Moehle K, Robinson JA:
Synthetic virus-like particles from self-assembling coiled-coil lipopeptides and their
use in antigen display to the immune system. Angewandte Chemie-International
Edition 46(47), 9015-9018 (2007).
Riedel T, Ghasparian A, Moehle K, Rusert P, Trkola A, Robinson JA: Synthetic
Virus-Like Particles and Conformationally Constrained Peptidomimetics in Vaccine
Design. ChemBioChem 12(18), 2829-2836 (2011).
Sharma R, Ghasparian A, Robinson JA, Mccullough KC: Synthetic Virus-Like
Particles Target Dendritic Cell Lipid Rafts for Rapid Endocytosis Primarily but Not
Exclusively by Macropinocytosis. PLoS One 7(8), (2012).
Roy A, Franco OL, Mandal SM: Biomedical Exploitation of Self Assembled Peptide
Based Nanostructures. Current Protein & Peptide Science 14(7), 580-587 (2013).
Chesson CB, Huelsmann EJ, Lacek AT et al.: Antigenic peptide nanofibers elicit
adjuvant-free CD8(+) T cell responses. Vaccine 32(10), 1174-1180 (2014).
Chen JJ, Pompano RR, Santiago FW et al.: The use of self-adjuvanting nanofiber
vaccines to elicit high-affinity B cell responses to peptide antigens without
inflammation. Biomaterials 34(34), 8776-8785 (2013).
721
722
723
724
725
726
727
728
729
730
731
732
733
734
735
736
737
738
739
740
741
742
743
744
745
746
747
748
749
750
751
752
753
754
755
756
757
758
759
760
761
762
763
764
765
766
767
768
769
770
64.
Rudra JS, Mishra S, Chong AS et al.: Self-assembled peptide nanofibers raising
durable antibody responses against a malaria epitope. Biomaterials 33(27), 6476-6484
(2012).
65.
Rudra JS, Tian YF, Jung JP, Collier JH: A self-assembling peptide acting as an
immune adjuvant. Proc. Natl. Acad. Sci. U. S. A. 107(2), 622-627 (2010).
66.
Skwarczynski M, Kowapradit J, Ziora ZM, Toth I: pH-triggered peptide selfassembly into fibrils: a potential peptide-based subunit vaccine delivery platform.
Biochemical Compounds 1(1), (2013).
67.
Kaba SA, Brando C, Guo Q et al.: A Nonadjuvanted Polypeptide Nanoparticle
Vaccine Confers Long-Lasting Protection against Rodent Malaria. J. Immunol.
183(11), 7268-7277 (2009).
68.
Yang Y, Neef T, Mittelholzer C et al.: The biodistribution of self-assembling protein
nanoparticles shows they are promising vaccine platforms. J. Nanobiotechnol. 11,
(2013).
69.
El Bissati K, Zhou Y, Dasgupta D et al.: Effectiveness of a novel immunogenic
nanoparticle platform for Toxoplasma peptide vaccine in HLA transgenic mice.
Vaccine 32(26), 3243-3248 (2014).
70.
Ojea-Jimenez I, Comenge J, Garcia-Fernandez L, Megson ZA, Casals E, Puntes VF:
Engineered Inorganic Nanoparticles for Drug Delivery Applications. Curr. Drug
Metab. 14(5), 518-530 (2013).
71.
Lindblad EB: Aluminium adjuvants - in retrospect and prospect. Vaccine 22(27-28),
3658-3668 (2004).
72.
Frey A, Mantis N, Kozlowski PA et al.: Immunization of mice with peptomers
covalently coupled to aluminum oxide nanoparticles. Vaccine 17(23-24), 3007-3019
(1999).
73.
Frey A, Neutra MR, Robey FA: Peptomer aluminum oxide nanoparticle conjugates as
systemic and mucosal vaccine candidates: Synthesis and characterization of a
conjugate derived from the C4 domain of HIV-1(MN) gp120. Bioconjugate Chem.
8(3), 424-433 (1997).
74.
Chen Y-S, Hung Y-C, Lin W-H, Huang GS: Assessment of gold nanoparticles as a
size-dependent vaccine carrier for enhancing the antibody response against synthetic
foot-and-mouth disease virus peptide. Nanotechnology 21(19), (2010).
 Comprehensive study on size-dependent immunogenicity of gold-nanoparticles.
75.
Chiu D, Zhou W, Kitayaporn S et al.: Biomineralization and Size Control of Stable
Calcium Phosphate Core-Protein Shell Nanoparticles: Potential for Vaccine
Applications. Bioconjugate Chem. 23(3), 610-617 (2012).
76.
Powell TJ, Palath N, Derome ME, Tang J, Jacobs A, Boyd JG: Synthetic nanoparticle
vaccines produced by layer-by-layer assembly of artificial biofilms induce potent
protective T-cell and antibody responses in vivo. Vaccine 29(3), 558-569 (2011).
77.
Mehra NK, Mishra V, Jain NK: A review of ligand tethered surface engineered
carbon nanotubes. Biomaterials 35(4), 1267-1283 (2014).
78.
Foldvari M, Bagonluri M: Carbon nanotubes as functional excipients for
nanomedicines: II. Drug delivery and biocompatibility issues. Nanomed.Nanotechnol. Biol. Med. 4(3), 183-200 (2008).
79.
Fadel TR, Fahmy TM: Immunotherapy applications of carbon nanotubes: from design
to safe applications. Trends Biotechnol. 32(4), 198-209 (2014).
80.
Gottardi R, Douradinha B: Carbon nanotubes as a novel tool for vaccination against
infectious diseases and cancer. J. Nanobiotechnol. 11, (2013).
81.
Yandara N, Pastorin G, Prato M, Bianco A, Patarroyo ME, Lozano JM:
Immunological profile of a Plasmodium vivax AMA-1 N-terminus peptide-carbon
771
772
773
774
775
776
777
778
82.
83.
nanotube conjugate in an infected Plasmodium berghei mouse model. Vaccine 26(46),
5864-5873 (2008).
Pantarotto D, Partidos CD, Graff R et al.: Synthesis, structural characterization, and
immunological properties of carbon nanotubes functionalized with peptides. J. Am.
Chem. Soc. 125(20), 6160-6164 (2003).
Villa CH, Dao T, Ahearn I et al.: Single-Walled Carbon Nanotubes Deliver Peptide
Antigen into Dendritic Cells and Enhance IgG Responses to Tumor-Associated
Antigens. ACS Nano 5(7), 5300-5311 (2011).