G Model
ARTICLE IN PRESS
JVAC 16762 1–10
Vaccine xxx (2015) xxx–xxx
Contents lists available at ScienceDirect
Vaccine journal homepage: www.elsevier.com/locate/vaccine
Polymeric micro/nanoparticles: Particle design and potential vaccine delivery applications
1
2
Q1
3
Hua Yue a , Guanghui Ma a,b,∗ a
4
b
5
National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, China
6
7 19
a r t i c l e
i n f o
a b s t r a c t
8 9 10
Article history: Available online xxx
11
Particle based adjuvant showed promising signs on delivering antigen to immune cells and acting as stimulators to elicit preventive or therapeutic response. Nevertheless, the wide size distribution of available polymeric particles has so far obscured the immunostimulative effects of particle adjuvant, and compromised the progress in pharmacological researches. To conquer this hurdle, our research group has carried out a series of researches regarding the particulate vaccine, by taking advantage of the successful fabrication of polymeric particles with uniform size. In this review, we highlight the insight and practical progress focused on the effects of physiochemical property (e.g. particle size, charge, hydrophobicity, surface chemical group, and particle shape) and antigen loading mode on the resultant biological/immunological outcome. The underlying mechanisms of how the particles-based vaccine functioned in the immune system are also discussed. Based on the knowledge, particles with high antigen payload and optimized attributes could be designed for expected adjuvant purpose, leading to the development of high efficient vaccine candidates. © 2015 Published by Elsevier Ltd.
18
Keywords: Micro/nanoparticles Adjuvant Physiochemical property Preventive/therapeutic vaccine Antigen loading Immunological mechanism
20
1. Introduction
21
1.1. Adjuvants: avenue to the next-generation vaccine
12 13 14 15 16 17
Q2 22 23 24 25 26 27 28 29 30 31 32 33 34 35
Vaccination is the most effective and economic way to prevent infection and severe outcomes caused by bacterial, viruses, or other pathogen. Owing to hundred kinds of vaccines, global death has decreased, along with cost for treatment for infectious diseases. The development of vaccine products was accompanied with rapid advancement of biotechnology, immunology, and biomaterials. The first generation of vaccine was dated from 1796, when Edward Jenner inoculated healthy people with attenuated live cowpox virus, opening the era of vaccination. By the 1940s, the vaccines were developed when passage in cell culture permitted the selection of attenuated mutants. Following that era, heat or chemical inactivation was applied to pathogens (e.g. typhoid, plague, and so on) to reduce the toxicity while maintaining the immunogenicity [1]. Although live or inactivated vaccines are capable of eliciting robust
∗ Corresponding author at: National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, No. 1 Bei-Er-Tiao, Zhong Guan Cun, Beijing 100190, China. Tel.: +86 1082627072; fax: +86 1082627072. E-mail address:
[email protected] (G. Ma).
immune response with the assist of endogenous adjuvant (nonspecific pathogenic component), the safety concerns has limited their clinic use, especially when extremely dangerous pathogens, such as human immunodeficiency virus (HIV) and hepatitis virus, were involved [2]. Therefore, the modern generation of genetically engineered vaccine or subunit vaccine, which have defined components, good stability and high safety, emerged from the recombinant hepatitis B virus vaccine in 1986. However, most of the new generation vaccines suffer from shortcoming of poor immunogenicity, and the exploration of high-performance adjuvant has become a rapidly expanding research area [3]. In addition to the progress of preventive vaccines for healthy people, vaccine-based treatments of chronic infected diseases and malignant tumors have attracted tremendous interests in the recent decades [4,5]. Compared with conventional chemotherapy or radiotherapy, vaccines strategy possesses relatively higher specificity and lower side effect. For example, the first Food and Drug Administration (FDA)-approved tumor vaccine (Provenge) could suppress metastatic castration-resistant prostate cancer by activating the power of the patient’s own immune system [6]. A crucial requirement for therapeutic vaccine is to elicit sufficient cellular immune responses against the target cells, thus not only the strength but also the polarization of the immune response should be considered in adjuvant design, making it even more challenging [7].
http://dx.doi.org/10.1016/j.vaccine.2015.07.100 0264-410X/© 2015 Published by Elsevier Ltd.
Please cite this article in press as: Yue H, Ma G. Polymeric micro/nanoparticles: Particle design and potential vaccine delivery applications. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.07.100
36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
G Model JVAC 16762 1–10
ARTICLE IN PRESS H. Yue, G. Ma / Vaccine xxx (2015) xxx–xxx
2
81
Development of vaccines requires safe and efficient adjuvant or antigen delivery systems. Although being pursued, ideal adjuvants remain out of reach for now. Alum salt is the most commonly used adjuvant approved by FDA (in 1926), which can be found in diphtheria–tetanus–pertussis, human papillomavirus or hepatitis vaccines [8]. Alum salt induces the antibody related immune response by a depot effect and activation of antigen present cells (APCs) (via NLRP3 inflammasome). Nevertheless, the preventive effect was not satisfactory for some subunit vaccine (e.g. H5N1 split vaccine). Moreover, its incapacity in eliciting cellular response hindered its therapeutic application in treatment of infected or cancerous cells. In the following 80 years, adjuvants like water-in-oil emulsion (e.g. Freund’s adjuvant), microorganism component (lipopolysaccharide, LPS), and cytokines (Interleukine2) were developed to induce higher immune response. Albeit effective in animal models, these adjuvants were reported to induce adverse effects like fever, lethargy, and even shock. Their clinic applications were thus stagnated until MPL-A (Monophosphoryl Lipid A, a derivate of LPS) was adopted as an adjuvant in FDA approved cervical cancer vaccine in 2009. In this regard, there is still an unfulfilled need for a safe and potent adjuvant.
82
1.2. Potential micro/nanoparticle-based adjuvant
61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80
83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124
Compared with traditional adjuvant, the micro/nanoparticles (MP/NP) possess a particular advantage as they have similar size and structure as that of a pathogen, leading to a preferred interaction with antigen presenting cells (APCs). MP/NP, such as polymeric particles, liposomes and micelle, have shown promising signs owing to their attributes in bio-application [9–13]. Firstly, antigens that loaded in the particles can be protected from enzymatic degradation and rapid denaturation. Secondly, particles can improve the antigen uptake by APCs, promoting subsequent antigen process and cellular mediated response. Thirdly, the fine-tuned characteristics (e.g. size, surface charge or structure) of particles allow them to fulfill a prolonged antigen release for long-lasting humoral response, or targeted delivery and controllable release for specific cellular immunity [14–16]. Last but not least, the particulate vaccines can be administrated via alternative routes like oral, rectal, or nasal administration, rather than subcutaneously injection. The potential of polymeric particulate vaccine delivery systems has been widely recognized [17–19]. However, owing to the prevalent use of non-uniform sized particles, discrepant and even contradictory outcomes were often present in biological or immunological evaluations. This situation has kept compromising the progress in establishing reliable theoretical guidance for particle design [20]. Moreover, wide size distribution may lead to an uncontrollable tissue distribution of the adjuvant particles, magnifying the concern on safety. To resolve these problems, our research group has developed a special MP/NP fabrication method on the basis of microporous membrane emulsification and has achieved successes in preparing bio-degradable (e.g. polylactide PLA) and polysaccharide (e.g. chitosan) MP/NP with controllable and uniform size (Fig. 1) [21–24]. Particles at a range of 100 nm to 100 m have been successfully prepared by choosing membrane with specific pore size. In addition, the scale-up equipment (40 L/h when pore size is 5.2 m) has been established in our group, which can be applied for scaling-up test. Based on this kernel technique, MP/NP of different structure or physiochemical properties for specific characteristics and functions can be obtained (see our previous review [25]). The polymeric MP/NP with defined sizes and properties thus facilitated to exploring the relationship between the particle properties and immunomodulatory effects, providing enlightening paradigms in vaccine development. Herein, we provide a review on the correlation of the particle physiochemical properties and antigen loading mode with the
Fig. 1. Schematic diagram of membrane emulsification process (A) and the scanning electron microscope (SEM) images of as-prepared PLA microparticles (B) and chitosan microparticles (C).
resultant biological/immunological outcomes, mostly based on the study of using the aforementioned uniform MP/NP. The underlying mechanisms of how the particles-based vaccine functioned in the immune system were also discussed. On the basis of particle design concept, potential applications of polymeric MP/NP were implicated not only for prophylactic vaccine (against e.g. Influenza, Anthrax), but also for therapeutic vaccine (against e.g. chronically infected hepatitis and malignant tumor). 2. Particle design of uniform micro/nanoparticles for vaccine delivery 2.1. Particle property design for vaccine delivery The requirements of immune responses vary for specific antigen type and vaccination purpose. Antibody mediated humoral response is critical in the defense against extracellular pathogens for the preventive vaccine, while antigen specific cytotoxic T cells is mostly wanted for therapeutic vaccine. As APCs are pivots that translate the vaccine stimulation to immune system, their responses to MP/NP are very important for regulating the subsequent immune response. The major APCs responses include APCs uptake, intracellular trafficking pathway, and cytokine profile. For most vaccines, avidly taken up by APCs is preferred to provide sufficient antigen signal. Since lysosomes are correlated to the main degradation pathway of exogenous antigen for CD4 T cells mediated humoral response, lysosomal degradation is not a favored process in eliciting cellular mediated response. In contrast, lysosome escape allows the cross-presentation of exogenous antigen to activate CD8+ T cells (CTL), which is particularly desired for the therapeutic vaccine or cancer vaccine [26]. In addition, the type and level of cytokine secretion by APCs can also modulate the subsequent immune activation. Therefore, a deeper understanding of how the physical properties of particles regulate APCs mediated biological or immunological responses is highly required. These correlation results will benefit the design of a particulate vaccine with optimized delivery mode or stimulation efficacy as well as minimized potential hazards [27]. 2.1.1. Particle size Particle size is the primary attribute and plays a paramount role in the following fate of antigen when the antigen loaded
Please cite this article in press as: Yue H, Ma G. Polymeric micro/nanoparticles: Particle design and potential vaccine delivery applications. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.07.100
125 126 127 128 129 130 131 132
133 134
135
136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159
160 161 162
G Model JVAC 16762 1–10
ARTICLE IN PRESS H. Yue, G. Ma / Vaccine xxx (2015) xxx–xxx
163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217
218 219 220 221 222 223 224 225 226 227
particles encounter with APCs. In fact, some conflicting reports existed for a long term regarding the influence of particle size on immune responses. For example, a study suggested that 5 m (PLA-glycolide) PLGA particles more efficiently enhanced immune responses, while another study showed that 1 m particles were preferentially taken up by APCs and induced stronger immune responses than that of nanoparticles [28,29]. In contrast, polyacrylamide hydrogel particles of 35 nm and 3.5 m in size showed no difference in the magnitude of T-cell activation [30]. Such inconsistent results may originate from the intrinsic difference of the materials as well as the difficulty in preparing particles with controllable size and narrow size distribution. Uniform sized chitosan particles at different size (430 nm, 1.9 m and 4.8 m) (Fig. 2A–C) were prepared by utilizing the membrane emulsification technique, and particle size effect on biological response in macrophages (a professional APCs) was compared [31]. These chitosan particles showed bright autofluorescence, and the fluorescent intensity of cells was proved in direct proportion to the internalized particle volume. The particle number/surface area/volume of the internalized MP/NP was thus calculated according to the cellular fluorescence data, and corresponding antigen loading modes were suggested. 430 nm particles accumulated in the cells at a faster rate, and with a higher surface area (∼800 m2 ) and particle number (330) (Fig. 2D), indicating their advantage for antigen adsorption and conjugation. In contrast, the internalized 1.9 m microparticles displayed higher volume value (∼165 m3 ), suggesting their superiority in encapsulating therapeutic molecules (Fig. 2E and F). Moreover, the nanoparticles preferred to promote the secretion of Th1-specific molecule signals (e.g. IFN, IL-12) rather than immune suppressors, showing the capability of NP to improve vaccine efficacy. On the basis of this design concept, PLA (FDA-approved material which undergone extensive clinic evaluations) was employed as the material to prepare MP/NP adjuvant for Hepatitis B vaccine [32]. In comparison with MP, NP promoted higher antigen internalization and diverse degradation in lysosomes as well as proteasomes (Fig. 3). This result indicated a possibility to cross present antigen to CTL, which was indispensable for the direct cytotoxic lysis against infected cells or tumor cells. In vivo experiments established that NP-antigen group indeed enhanced the hallmarks of cellular mediated response (e.g. CTL cytotoxicity and IFN-␥ cytokine). Particles size was also found to contribute to the vaccine function routes, as smaller particles (mostly <50 nm) were able to penetrate the compact barrier of lymphatic system [33]. Compared with 100 nm particles, 25 nm NP transported from the injection site directly to draining lymph nodes (LNs) and activated the vast amount of immune cells settled there. Nevertheless, particles with a very small size (especially less than 10 nm) were reported to cause potential cytotoxicity, which partially precluded their further application [34]. On account of the ultrahigh specific surface area of NP (1.4 nm), cell necrosis was triggered by oxidative stress and mitochondrial damage during the fierce interaction between NP and intracellular organelles. As particle size is a parameter that could be geared toward eliciting a desired immune type while minimizing toxicity, it needs to be seriously considered in vaccine design 2.1.2. Particle charge Surface charge is another feature that affects the in vivo distribution and cellular uptake of particle. As cell membrane is negatively charged, the internalizing process and thus the antigen delivery of particles may be influenced by the particles with different surface charge. Surface charge was found to play a very significant role in determining the type and amount of the particle coronas (absorbed protein from human plasma) [35]. For example, 100-nm carboxyl-modified (negatively charged) particles absorbed a larger number of immunoglobulins while less apolipoproteins than that
3
for the positively charged particles or plain particles. However, different studies still show discrepancies in the correlation of particle charge and cell interaction. Cationic liposome-regulated immune responses were found to rely on the surface charge density, and low-charge particles failed to promote the antigen specific immune responses even at high concentrations [36]. In contrast, negatively charged [37] liposomes could also act as efficient adjuvants to induce cell-mediated immune response. To elucidate the role of particle charge on the APCs responses, three kinds of NP (∼215 nm) were compared, including negatively charged N-NP (carboxymethyl chitosan NP, ∼ 45.8), neutrally charged M-NP (chitosan coated carboxymethyl chitosan NP, ∼ 0.5), and positively charged P-NP (quaternized chitosan NP, ∼ 39.2) (Fig. 4) [38]. As these particles were fabricated by using the membrane emulsification technique, uniform size as well as identical physicochemical properties could be assured, excluding influence of other factors except for surface charge. Both the cellular uptake rate and amount were found positively correlated with the surface charge in eight cell lines. Surface charge also decided the intracellular trafficking of different particles. P-NP escaped from lysosome and exhibited peri-nuclear localization, whereas the NNP and M-NP were highly colocalized with lysosome. Driven by these findings, we coated the PLA particles with cationic polymers (e.g. chitosan (CS), chitosan chloride (CSC), and polyethylenimine (PEI)) to build positively charged surfaces and tested their adjuvanticity for therapeutic hepatitis B vaccine (Fig. 5) [39]. Owing to the merits of positive charge on improved antigen adsorption and lysosome-independent antigen processing, the cationic particlebased vaccine generated an enhanced antigen uptake, stimulation, and augmented Th1 polarized cytokines (e.g. IL-1, IL-12, for cellular response). In vivo testes showed a rapid and enhanced humoral immune response as well as a more efficient cell mediated immune responses, in comparison with aluminum-adsorbed vaccine and undecorated PLA particles. 2.1.3. Hydrophobicity Surface hydrophobicity also plays an integral role in the interaction between APCs and particles, since the cell membranes are formed by a lipid bilayer, exhibiting a hydrophobic interior. Particles prepared with hydrophobic and high-molecular weight polymers tended to be more efficiently and rapidly phagocytosed [40]. However, the understanding went short especially regarding the effect of hydrophobicity on biological response and antigen/drug delivery efficacy, as compared with that of size and charge. To understand the inherent relationship between the particle hydrophobicity and the APC activation, PLA were took as prototype materials, and the biological outcome of three particles (∼290 nm), including PLA, PLGA, and PEG-b-PLA (polyethylene glycol-PLA) were compared [41]. With the change of the molecular composition, the hydrophobicity increased gradually from PLA, PLGA, to PEG-b-PLA NPs, which dictated the decreased macrophage internalization and subsequent delivery performance. Compared with PEG-b-PLA particles, PLA and PLGA NPs were more preferentially internalized by macrophages. By using PLA, PLGA, and PEG-PLA particles, which were similar in size (∼1 m) and morphology but different in surface hydrophobicity, their abilities in inducing immune responses were further evaluated [42]. Increased surface hydrophobicity of PLA-based particles greatly promoted the cellular internalization of recombinant Bacillus anthracis protective antigen (rPA) by dendritic cells (DCs a professional APCs). DCs treated with rPA-PLA (the most hydrophobic MP) exhibited a higher percentage of rPA priming cells (52.37%) than those treated by rPA-PLGA (50.33%) or rPA-PELA (44.17%). Meanwhile, the number of rPA priming cells increased more than 10-fold in both the axillary and the popliteal LNs in the PLA particle group as compared with the PLGA or PELA particle groups.
Please cite this article in press as: Yue H, Ma G. Polymeric micro/nanoparticles: Particle design and potential vaccine delivery applications. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.07.100
228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261
262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292
G Model JVAC 16762 1–10 4
ARTICLE IN PRESS H. Yue, G. Ma / Vaccine xxx (2015) xxx–xxx
Fig. 2. Internalization profiles of macrophages after 24 h incubation with 430 nm particles (A), 1.9 m particles (B) and 4.8 m particles (C). The internalization data were further expressed by means of particle number (D), surface area (E), and volume (F).
Fig. 3. The cellular response of APCs after incubation with PLA nanoparticle adjuvanted vaccine.
Fig. 4. Schematic image of charge effect on cellular uptake and intracellular trafficking.
Fig. 5. A strategy of using cationic polymer-coated PLA particles for therapeutic hepatitis B vaccine.
Please cite this article in press as: Yue H, Ma G. Polymeric micro/nanoparticles: Particle design and potential vaccine delivery applications. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.07.100
G Model JVAC 16762 1–10
ARTICLE IN PRESS H. Yue, G. Ma / Vaccine xxx (2015) xxx–xxx
5
Fig. 6. Measurement adhesion forces between different particles and live cells. (A) Schematic representation of the experimental setup for obtaining force curves on the cell surface with particle-coated tips. (B) SEM images of PLA, PLGA and PELA particles prepared for coating the cantilever tips (scale bar 1 mm). (C–E) Analysis of the adhesion-force distribution histogram for PLA (C), PLGA (D), and PELA (E) particles.
293 294 295 296 297 298 299 300
301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324
325 326 327 328 329 330 331
Through coating the particles onto the automatic force microscope probes, the assay of relative adhesion forces exhibited that PLA MP exhibited the strongest interaction force with the cell membrane (2382.8 pN), followed by PLGA MP (1845.37 pN), and PELA MP (1631.36 pN) (Fig. 6). This mechanism investigation revealed that the increased surface hydrophobicity enhanced the physical interaction intensity between particles and cell membranes, which thus promoted the internalization of particles into cells. 2.1.4. Surface chemical group Most of the chemical groups on particles source from the original component of particle material. To fulfill the knowledge gap of intrinsic surface chemical group on biological and immunological outcome, we fabricated a novel kind of adjuvant (CS-NH2 MP, ∼1.0 m) with abundant amino groups by membrane emulsification and layer-by-layer (LbL) assembly technology [43]. Compared with the amino-cross-linked MP (control MP), CS-NH2 MP with a high surface density of amino groups lead to improved vaccine efficacy. The level of IL-4 and IFN-␥, the secretion of rPA-specific IgG titers, and the antigen specific T cell proliferative in splenocytes were found significantly enhanced. In addition to intrinsic component, surface modification is also a routine strategy to modulate the performances of particulate adjuvants [44]. In one of the most typical examples, CD206 antibody was conjugated to the particles to guide their tracking down of DCs. Such an active targeting strategy was practical for exploring the particles as tumor vaccine adjuvant. In a recent study, a chitosan based NP adjuvant that modeled after granules of mast cells was endowed with attributes of LNs targeting as well as timed release of cytokines [45]. When used as an adjuvant during vaccination of mice with haemagglutinin from the influenza virus, the particles enhanced adaptive immune responses and increased survival of mice on lethal challenge. 2.1.5. Particle shape Shape is an essential property of natural bio-particles (e.g. versatile virus or bacterial), which is believed to be associated with their infection efficacy and the subsequent replication. However, investigation of shape effect has lagged far behind that of the size and charge, as the fabrications of particles with non-spherical shape are constricted upon limited material choice and restricted techniques.
This bottleneck was not broken until the emergency of some initial studies until recently, and shape was becoming a design parameter for micro- and nanoscale delivery carriers [46,47]. For example, shape effect of filaments particles (via polymer micelle assembly) was addressed, and worm shape particles were found with prolonged blood circulation and enhanced drug delivery efficacy than spherical particles [48]. In our study, we investigated the cellular behavior of nanospheres and nanorods in the interplay with different kinds of cells (including endothelia cells and macrophages) [49]. The rod-like particles possessed an appreciable advantage of being quickly internalized by cells, reminiscent of the marvelous cell infecting capacity of bacillus. Although particle internalization is a complex process, which is dependent on cell types and particle raw materials, macropinocytosis and phagocytosis were found be the predominant uptake mechanism for non-spherical particles [50,51]. Desimone group prepared cylindrical particles and pointed out that cells were readily to internalize particles with a diameter aspect ratio of 3 by using several different mechanisms of endocytosis. After optimizing the manufacture process of PRINT (Particle replication in non-wetting templates) technique, Liquidia Technologies drove the non-spherical PLGA-based particles (LIQ001, 80 × 80 × 320 nm) forward in the area of influenza vaccines [52]. These particles were safe and showed enhanced responses to influenza hemagglutinin in murine models. Recently, they have completed a Phase 1/2a clinical trial of LIQ001 + Fluzone involving both young and elderly healthy adults. In addition to the non-spherical particles, the two dimensional material graphene oxide (GO) has renewed interest in biomaterialbased application. We uncovered that the particle shape regulated both the intracellular distribution of particle and the cytokine profile response (Fig. 7) [53]. Intriguingly, the strong steric effect of flat micro-sized forced the micro-sized GO folding from 2 m to 300 nm, and brought higher cytokine secretion of macrophage than that of nano-sized GO. This might be attributed to the continuous struggle of GO for a more stable state of GO, which betrayed its extrinsic properties inside cells and finally resulted in recognition and stronger response from APCs. With this pilot study, we constructed a high-performance tumor vaccine and preliminarily proved the concept of new dimensional material based delivery system.
Please cite this article in press as: Yue H, Ma G. Polymeric micro/nanoparticles: Particle design and potential vaccine delivery applications. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.07.100
332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372
G Model JVAC 16762 1–10
ARTICLE IN PRESS H. Yue, G. Ma / Vaccine xxx (2015) xxx–xxx
6
Fig. 7. Cellular response of macrophage after exposure with 350 nm and 2 m GO. (A) Transmission electron microscopy images showing eligible cell interactions with different sized GO. (B) Cytokine profile showing that the cells stimulated with microsized GO secrete more immune activation-related cytokines.
373
374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398
2.2. Antigen loading mode for vaccine delivery 2.2.1. Adsorption/encapsulation/mixture Efficient antigen loading is indispensable for subsequent delivery and immune stimulation. The major loading patterns are adsorption and encapsulation, which influence the bioactivity and release rate of antigen in a complex manner. On one hand, when antigens are microencapsulated with particle, inactivation of antigen may occur during this process and compromise the corresponding immunogenicity and even cause the adverse effect [54,55]. In contrast, antigens that formulated with particles by gentle adsorption process are expected to get a better chance to maintain their activities (the activity loss of adsorbed antigen on PLA NP is below 5%). On the other hand, the adsorbed antigen will be released relatively quickly, leaving a limited time window for APCs recruitment, whereas the encapsulated antigens show controlledrelease characteristics and lead to superior long-lasting antibody levels (more than 32 weeks) [56]. To achieve a better understanding of how the antigen loading way impact on the antigen exposure and the resultant antigen-specific immune responses, we compared three antigenNP formulations (Fig. 8) [57]. The combined formulation (composed of antigen encapsulated in nanoparticles and antigen absorbed on nanoparticles through simple mixture) induced more powerful antigen-specific immune responses than each single-component formulation (antigen encapsulated within nanoparticles, or soluble antigen mixed with blank nanoparticles). The potential of
combined formulation might be attributed to the antigen-depot effect at the injection site, effective supply of both adequate initial antigen exposure and long-term antigen persistence, and efficient induction of DC activation and follicular helper T cell differentiation in LNs. Therefore, understanding the effect of different antigen loading on the resultant immune responses has significant implications for rational vaccine design.
2.2.2. Other method: NP-based multi-adjuvant modality Besides the traditional antigen loading methods, a whole cell tumor antigen modality was constructed based on the particle adjuvants. This modality of whole cell tumor vaccine (WCTV) is especially suitable for the tumors whose specific antigens have not been identified. On account of the carrier function of CPP modified PLGA NP, two encapsulated stimulators (IL-2 and GM-CSF) in this particle were efficiently introduced, via the entry of PLA NP into tumor cell. In addition to the reinforced internalization of stimulators with the assist of NP, a lysosome-escape behavior of NP also contributed a lot to the significantly improved bioavailability of imported GM-CSF and IL-2. These adjuvant-primed tumor cells were further used as multi-adjuvant WCTV after inactivation. In vitro data demonstrated the programed promotions of multi-adjuvants on DC recruitment, antigen presentation, and T-cell activation. In vivo evaluations demonstrated the satisfactory effects on tumor growth suppression, metastasis inhibition, and recurrence prevention. In this aspect, the nanoparticles-based
Please cite this article in press as: Yue H, Ma G. Polymeric micro/nanoparticles: Particle design and potential vaccine delivery applications. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.07.100
399 400 401 402 403 404 405
406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423
G Model JVAC 16762 1–10
ARTICLE IN PRESS H. Yue, G. Ma / Vaccine xxx (2015) xxx–xxx
7
Fig. 8. Illustration of different antigen loading modes.
Fig. 9. Immunological mechanism of alum and MP/NP adjuvanted vaccine.
424 425
426
427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442
443 444 445 446 447
multi-adjuvant served as a high-performance modality for therapeutic vaccine. 2.3. Immunological mechanism of the particle based adjuvant 2.3.1. Antigen reservoir In contrast to the rapid clearance of soluble antigen at injection site after administration of soluble antigen, the alum salt significantly prolonged antigen deposition. This “antigen reservoir” effect provided a longer time window for the recruitment of inflammation related cells (e.g. DCs, macrophage, and neutrophil) and promoted their subsequent interaction between these APCs and antigen. However, the antigen reservoir capability of particles varied with their components and the physiochemical properties. Whereas chitosan based particles deposited at the injection site for a longer period [39], PLA particles that were cleared (internalized by APCs) at a quick speed were favorable for further antigen process [58]. Moreover, in design of LN targeted antigen delivery, there’s a need for particles to directly migrate to the immune APC/T cells that settled in the draining lymph node, rather than stay at the injection sites. 2.3.2. APCs uptake and activation APCs are the leading actors in processing vaccine antigens, which receive the vaccine stimulation and motivate the other part of the immune system. These cells engulf the antigen and present the most characteristic peptide (major histocompatibility
complex MHC molecule, recognition signal) on cell surface. Based on specific MHC peptide signals and costimulators (stimulation signal), immune cells like CD4 and CD8 T cells will be activated to release their weapons (antibody or granule/perforin), exerting their functions in neutralizing the virus or attacking the infected cells, respectively. On the contrary, lacking of any MHC molecules (especially MHC I for CD8 T) and costimulators (CD80/86) or cytokines (communication signal), will lead to weak cellular response or immunity tolerance, thus conniving chronic diseases (chronic hepatitis B) or cancer progression. For instance, the incapacity of alum adjuvant in cellular response of alum adjuvant was found highly associated with downregulation of MHC I level expression in APCs [32]. Guided by this immunological process, most of the strategies (nano-sized, positively charged, rod-like) focused on increasing the chances of antigen to be massively and efficiently processed by APCs. The aforementioned vaccine adjuvants (chitosan NP, PEI modified PLA MP, non-spherical MP/NP) enjoyed the advantage of improving cellular uptake of antigen or activating the recognition or costimulator signals for T cells. Moreover, with the stimulation of exogenous particles, APCs secreted a range of signaling molecules or cytokines, which were vital to the following immunological regulation and activation. 2.3.3. Lysosome escape The internalized exogenous antigens were usually entrapped and processed by lysosome for CD4 T mediated antibody response,
Please cite this article in press as: Yue H, Ma G. Polymeric micro/nanoparticles: Particle design and potential vaccine delivery applications. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.07.100
448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470
471 472 473
G Model JVAC 16762 1–10 8
ARTICLE IN PRESS H. Yue, G. Ma / Vaccine xxx (2015) xxx–xxx
2.3.4. Complement activation Complement activation is capable of acquiring systematic immune response and is considered an alternative approach toward immunotherapy [67]. In the case of CS-NH2 MP, C3a (an index molecule for complement activation) was significantly enhanced upon the particle exposure, resulting in stronger antigen specific humoral response. Notwithstanding a similar cellular uptake level of the MP with different extends of amino group, the abundant amino groups on CS-NH2 MP could promote the covalent binding of C3b molecular and efficiently activate completion pathway. Considering possible acute inflammatory responses provoked by C3a, we carefully checked the pathological changes in the subcutis, and verified the fair cell infiltration and safety of CS-NH2 MP for using as a vaccine-delivery platform.
Fig. 10. Multiple mechanisms of immunity enhancement by the Gel MP based vaccine delivery system.
whereas the endogenous (self) antigens were processed via protease for CD8 T. A well-documented benefit of alum adjuvant is to elicit robust preventive humoral response. This can be partly 476 ascribed to the undivided antigen supply to lysosome-mediated 477 antigen processing, and the consequent MHC II signal upregulation 478 Q3 for efficient CD4 T cell activation (Fig. 9). However, the high fre479 quent antigenic variations among circulating virus (e.g. H1N1 and 480 H5N1) drove us to develop vaccines to elicit cross-protective CD8 T 481 cell response against both viruses [59]. Aiming to treat intracellular 482 infection and malignant cells, exogenous antigen also needs to be 483 cross-present through MHC I pathway to activate CD8 T cytotoxic 484 responses. Strategies to cross-present antigen include lysosome 485 escape of antigen, targeting Toll-like receptors with additive of dan486 ger signals (e.g. CpG) [60–62] and so on. In this review, we focused 487 on the lysosome escape way. 488 Antigen escape from lysosome was preferred when strong cel489 lular immune response was expected, and well-designed particle 490 adjuvants could trigger this process very efficiently. Considering 491 the advantage of a facile preparation condition, particles that bear 492 this merit but need no modification were intriguing candidates. 493 Under the “proton-sponge” effect, the positive charge of NPs (as 494 referred in P-NP) may trigger the influx of chloride ions to main495 tain charge neutrality, leading to osmotic swelling, physical rupture 496 of lysosomes, and the final antigen escape to cytoplasm [63,64]. 497 To further enhance the efficacy of lysosome escape, some stud498 ies attempted to modulate the intracellular trafficking of particles 499 by further functionalization or fine-tuning the particle component. 500 For example, we successfully developed a novel adjuvant candidate 501 (Gel MP) based on the thermal-gelation property and pH sensitive 502 properties of quaternized chitosan (Fig. 10) [65,66]. The majority 503 (close to 80%) of antigens (H5N1) in Gel MP/HA group were disso504 ciated from lysosomes, demonstrating an efficient antigen escape 505 from lysosome and release to the cytoplasm. A possible reason 506 for lysosome escape was the swelling and dissolution of pH sensi507 tive Gel MP under acidic microenvironment, which might result in 508 osmotic imbalance as proved by the improvement of erythrocyte 509 lysis. Another reason was the above-mentioned “proton-sponge” 510 effect in acidic endosome, which was related to high buffering 511 capacity (BC) of polymers. Compared with the positive PEI control, 512 a significantly higher BC was revealed for Gel MP in the pH range 513 for endosomal escape (pH 4.5–7), which was likely due to added 514 phosphate groups during NP preparation. 515 474 475
516 517 518 519 520 521 522 523 524 525 526 527 528 529
3. Conclusions and perspective
530
3.1. Conclusions
531
This article reviewed the recent insight and practical progress of MP/NP based adjuvant/antigen delivery system in our research group, emphasizing on the uniform-sized polymeric particles. In-depth understandings of particle–bio interactions make an important contribution to support the rapid expanding researches on particle adjuvants. However, it is not an easy task to draw an uncontested conclusion due to the complexity originated from the particle fabrication. By using a unique microporous membrane emulsification technique, we prepared particles with uniform and controllable size with good reproducibility, paving the way for further investigation on biological/immunological response. With these particles, the effect of a single property (e.g. size, charge, shape) can be clarified, and the influence of other factors is minimized to ensure reliable results. Taking advantage of successful exploration of particle-bio interaction, smaller size (raging from 20 nm to 1 m), positive charge, rod shape, hydrophobic surface, specific chemical component were implicated in the active immune response. Moreover, the underlying versatile mechanisms of how the particles modulated in the immune system were also unveiled. Given the extensive knowledge, the tailored design of particle with high antigen payload and optimized attributes for expected purpose became feasible. 3.2. Perspectives In this article, we focused on constructing particles adjuvant under the knowledge of the influences of particle property on the immunological outcome. Intelligent micro/nanoparticles combining both antigen delivery and multi-stimulation are promising candidates for the new generation vaccines. Nevertheless, the capacity of adjuvant should still be optimized to cater for the vaccine demand of immune efficacy or longevity. With the thorough comprehension of the immunological effect of each property, more than one attribute (e.g. charge and shape) can be introduced into a single particle, via component control, ligand graft or surface modification, thus to achieve specific function (carrier or targeting) and activity (stimulation). In addition, the thorough theoretical progress of novel or traditional adjuvant addressed on molecular, cellular and animal level is highly appreciated [68], which will provide the design concept for maximizing the efficiency of the integrated multi-attributes while minimizing the potential hazard. Recalling the latest developments in preventive vaccine, particle-based vaccine promises compelling advantages even beyond adjuvant. Nevertheless, most of the studies are still in its research stage, and the clinical trial fairly lagged behind the overwhelming research work. Only scattered and few examples were
Please cite this article in press as: Yue H, Ma G. Polymeric micro/nanoparticles: Particle design and potential vaccine delivery applications. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.07.100
532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553
554
555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575
G Model JVAC 16762 1–10
ARTICLE IN PRESS H. Yue, G. Ma / Vaccine xxx (2015) xxx–xxx
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
translated from bench to the clinical Phase. The major handicap is associated with the quite limited material options due to the difficultly in developing material with good safety profile. To circumvent the problem, the well-proved materials (PLA/PLGA) with excellent biocompatibility are preferred for preventive vaccine, which is come-at-able for clinical trial. In the meantime, it is urgent to improve the quality of candidate materials (e.g. chitosan) base on the control of the preparation process and the comprehensive verification of the safety issue. Regarding the material that has not been fully approved but extensively proved, selecting a relative safe administration route, such as the transdermal (micro-needle), oral or mucosal delivery (e.g. chitosan based adjuvant) [69,70], is also an acceptable way to reduce the safety concern. As for both preventive and therapeutic vaccines, an overall challenge is the scale-up production of particles with uniform size and specific property. To achieve this goal, the corresponding manufacture apparatus should be developed to guarantee the repeatability of products, as well as provide enough samples for clinical trials. In our lab, pilot-scale equipment with automatic process control has been established, and a large amount of studies on fine-tuning the particles preparation conditions and process optimization are being performed. This progress may make an important contribution to the particle-based adjuvants, and is waiting to reach its full potential for further applications for humans. Author contribution Both authors have (1) screened relevant literature and discussed the review outline, (2) drafted the article or revised it critically for important intellectual content, and (3) approved the final version submitted. Conflict of interest statement The authors have no conflict of interest to declare. Acknowledgments
612
This work was supported by National Natural Science Foundation of China (51302265), National High Technology Research and Development Program of China (2014AA093604), and Major Project of the Ministry of Science and Technology of China (2014ZX09102045).
613
References
Q4 608 609 610 611
614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636
[1] Plotkin S. History of vaccination. Proc Natl Acad Sci U S A 2014;111(34):12283–7. [2] Blower SM, Koelle K, Kirschner DE, Mills J. Live attenuated HIV vaccines: predicting the tradeoff between efficacy and safety. Proc Natl Acad Sci U S A 2001;98(6):3618–23. [3] Coffman RL, Sher A, Seder RA. Vaccine adjuvants putting innate immunity to work. Immunity 2010;33(4):492–503. [4] Schlom J. Therapeutic cancer vaccines: current status and moving forward. J Natl Cancer Inst 2012;104(8):599–613. [5] Silva JM, Videira M, Gaspar R, Preat V, Florindo HF. Immune system targeting by biodegradable nanoparticles for cancer vaccines. J Control Release 2013;168(2):179–99. [6] Cheever MA, Higano CSPROVENGE. (Sipuleucel-T) in prostate cancer: the first FDA-approved therapeutic cancer vaccine. Clin Cancer Res 2011;17(11):3520–6. [7] Banday AH, Jeelani S, Hruby VJ. Cancer vaccine adjuvants – recent clinical progress and future perspectives. Immunopharmacol Immunotoxicol 2015;37(1):1–11. [8] Marrack P, McKee AS, Munks MW. Towards an understanding of the adjuvant action of aluminium. Nat Rev Immunol 2009;9(4):287–93. [9] Xiang SD, Scalzo-Inguanti K, Minigo G, Park A, Hardy CL, Plebanski M. Promising particle-based vaccines in cancer therapy. Expert Rev Vaccines 2008;7(7):1103–19.
9
[10] Cheng Z, Al Zaki A, Hui JZ, Muzykantov VR, Tsourkas A. Multifunctional nanoparticles: cost versus benefit of adding targeting and imaging capabilities. Science 2012;338(6109):903–10. [11] van Riet E, Ainai A, Suzuki T, Kersten G, Hasegawa H. Combatting infectious diseases; nanotechnology as a platform for rational vaccine design. Adv Drug Deliv Rev 2014;74C:28–34. [12] Singh M, Chakrapani A, O’Hagon D. Nanoparticles and microparticles as vaccine-delivery systems. Exp Rev Vaccines 2007;6(5):797–808. [13] Zhao L, Seth A, Wibowo N, Zhao CX, Mitter N, Yu C, et al. Nanoparticle vaccines. Vaccine 2014;32(3):327–37. [14] Zhou W, Moguche AO, Chiu D, Murali-Krishna K, Baneyx F. Just-in-time vaccines: biomineralized calcium phosphate core-immunogen shell nanoparticles induce long-lasting CD8(+) T cell responses in mice. Nanomedicine 2014;10(3):571–8. [15] Carcaboso AM, Hernandez RM, Igartua M, Rosas JE, Patarroyo ME, Pedraz JL. Potent, long lasting systemic antibody levels and mixed Th1/Th2 immune response after nasal immunization with malaria antigen loaded PLGA microparticles. Vaccine 2004;22(11-12):1423–32. [16] Shah RR, O’Hagan DT, Amiji MM, Brito LA. The impact of size on particulate vaccine adjuvants. Nanomedicine (Lond) 2014;9(17):2671–81. [17] Rice-Ficht AC, Arenas-Gamboa AM, Kahl-McDonagh MM, Ficht TA. Polymeric particles in vaccine delivery. Curr Opin Microbiol 2010;13(1):106–12. [18] Ferreira SA, Gama FM. Biodegradable polymeric nanoparticles as vaccine delivery systems. Vaccine 2008. [19] Akagi T, Baba M, Akashi M. Biodegradable nanoparticles as vaccine adjuvants and delivery systems: regulation of immune responses by nanoparticle-based vaccine. Polym Nanomed 2012;247:31–64. [20] Oyewumi MO, Kumar A, Cui ZR. Nano-microparticles as immune adjuvants: correlating particle sizes and the resultant immune responses. Exp Rev Vaccines 2010;9(9):1095–107. [21] Wang LY, Ma GH, Su ZG. Preparation of uniform sized chitosan microspheres by membrane emulsification technique and application as a carrier of protein drug. J Control Release 2005;106(1-2):62–75. [22] Liu R, Ma GH, Meng FT, Su ZG. Preparation of uniform-sized PLA microcapsules by combining Shirasu Porous Glass membrane emulsification technique and multiple emulsion-solvent evaporation method. J Control Release 2005;103(1):31–43. [23] Wei Q, Wei W, Tian R, Wang LY, Su ZG, Ma GH. Preparation of uniform-sized PELA microspheres with high encapsulation efficiency of antigen by premix membrane emulsification. J Colloid Interface Sci 2008;323(2):267–73. [24] Wei W, Yuan L, Hu G, Wang LY, Wu H, Hu X, et al. Monodisperse chitosan microspheres with interesting structures for protein drug delivery. Adv Mater 2008;20(12):2292–6. [25] Ma GH. Microencapsulation of protein drugs for drug delivery: strategy, preparation, and applications. J Control Release 2014;193:324–40. [26] Flinsenberg TW, Compeer EB, Boelens JJ, Boes M. Antigen cross-presentation: extending recent laboratory findings to therapeutic intervention. Clin Exp Immunol 2011;165(1):8–18. [27] Mitragotri S, Lahann J. Physical approaches to biomaterial design. Nat Mater 2009;8(1):15–23. [28] Gupta V, Ahsan F. Influence of PEI as a core modifying agent on PLGA microspheres of PGE(1), a pulmonary selective vasodilator. Int J Pharm 2011;413(1–2):51–62. [29] Murphy A, Taggart G. A comparison of predicted and experimental critical micelle concentration values of cationic and anionic ternary surfactant mixtures using molecular-thermo dynamic theory and pseudophase separation theory. Colloids Surf A 2002;205(3):237–48. [30] Cohen JA, Beaudette TT, Tseng WW, Bachelder EM, Mende I, Engleman EG, et al. T-cell activation by antigen-loaded pH-sensitive hydrogel particles in vivo: the effect of particle size. Bioconjug Chem 2009;20(1):111–9. [31] Yue H, Wei W, Yue ZG, Lv PP, Wang LY, Ma GH, et al. Particle size affects the cellular response in macrophages. Eur J Pharma Sci 2010;41(5):650–7. [32] Yue H, Wei W, Fan B, Yue ZG, Wang LY, Ma GH, et al. The orchestration of cellular and humoral responses is facilitated by divergent intracellular antigen trafficking in nanoparticle-based therapeutic vaccine. Pharmacol Res 2012;65(2):189–97. [33] Reddy ST, van der Vlies AJ, Simeoni E, Angeli V, Randolph GJ, O’Neil CP, et al. Exploiting lymphatic transport and complement activation in nanoparticle vaccines. Nat Biotechnol 2007;25(10):1159–64. [34] Pan Y, Leifert A, Ruau D, Neuss S, Bornemann J, Schmid G, et al. Gold nanoparticles of diameter 1.4 nm trigger necrosis by oxidative stress and mitochondrial damage. Small 2009;5(18):2067–76. [35] Lundqvist M, Stigler J, Elia G, Lynch I, Cedervall T, Dawson KA. Nanoparticle size and surface properties determine the protein corona with possible implications for biological impacts. Proc Natl Acad Sci U S A 2008;105(38):14265–70. [36] Ma YF, Zhuang Y, Xie XF, Wang C, Wang F, Zhou DM, et al. The role of surface charge density in cationic liposome-promoted dendritic cell maturation and vaccine-induced immune responses. Nanoscale 2011;3(5):2307–14. [37] Yotsumoto S, Aramaki Y, Kakiuchi T, Tsuchiya S. Induction of antigendependent interleukin-12 production by negatively charged liposomes encapsulating antigens. Vaccine 2004;22(25–26):3503–9. [38] Yue ZG, Wei W, Lv PP, Yue H, Wang LY, Su ZG, et al. Surface charge affects cellular uptake and intracellular trafficking of chitosan-based nanoparticles. Biomacromolecules 2011;(7):2440–6. [39] Chen X, Liu Y, Wang L, Liu Y, Zhang W, Fan B, et al. Enhanced humoral and cell-mediated immune responses generated by cationic
Please cite this article in press as: Yue H, Ma G. Polymeric micro/nanoparticles: Particle design and potential vaccine delivery applications. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.07.100
637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722
G Model JVAC 16762 1–10 10 723 724 725
[40]
726 727 728
[41]
729 730 731 732
[42]
733 734 735
[43]
736 737 738
[44]
739 740 741 742
[45]
743 744 745
[46]
746 747 748
[47]
749 750 751
[48]
752 753 754
[49]
755 756 757
[50]
758 759 760
[51]
761 762 763
[52]
764 765 766
[53]
767 768 769 770 771
[54]
ARTICLE IN PRESS H. Yue, G. Ma / Vaccine xxx (2015) xxx–xxx
polymer-coated PLA microspheres with adsorbed HBsAg. Mol Pharma 2014;11(6):1772–84. Nel AE, Madler L, Velegol D, Xia T, Hoek EMV, Somasundaran P, et al. Understanding biophysicochemical interactions at the nano–bio interface. Nat Mater 2009;8(7):543–57. Yue ZG, You ZX, Yang QZ, Lv PP, Yue H, Wang B, et al. Molecular structure matters: PEG-b-PLA nanoparticles with hydrophilicity and deformability demonstrate their advantages for high-performance delivery of anti-cancer drugs. J Mater Chem B 2013;1(26):3239–47. Liu Y, Yin Y, Wang LY, Zhang WF, Chen XM, Yang XX, et al. Surface hydrophobicity of microparticles modulates adjuvanticity. J Mater Chem B 2013;1(32):3888–96. Liu Y, Yin Y, Wang LY, Zhang WF, Chen XM, Yang XX, et al. Engineering biomaterial-associated complement activation to improve vaccine efficacy. Biomacromolecules 2013;14(9):3321–8. 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 2011;29(3):558–69. John ALS, Chan CY, Staats HF, Leong KW, Abraham SN. Synthetic mast-cell granules as adjuvants to promote and polarize immunity in lymph nodes. Nat Mater 2012;11(3):250–7. Mathaes R, Winter G, Besheer A, Engert J. Non-spherical micro- and nanoparticles: fabrication, characterization and drug delivery applications. Expert Opin Drug Deliv 2015;12(3):481–92. Champion JA, Katare YK, Mitragotri S. Particle shape: a new design parameter for micro- and nanoscale drug delivery carriers. J Control Release 2007;121(12):3–9. Geng Y, Dalhaimer P, Cai S, Tsai R, Tewari M, Minko T, et al. Shape effects of filaments versus spherical particles in flow and drug delivery. Nat Nanotechnol 2007;2(4):249–55. Yue ZG, Wei W, You ZX, Yang QZ, Yue H, Su ZG, et al. Iron oxide nanotubes for magnetically guided delivery and pH-activated release of insoluble anticancer drugs. Adv Funct Mater 2011;21(18):3446–53. Barua S, Yoo JW, Kolhar P, Wakankar A, Gokarn YR, Mitragotri S. Particle shape enhances specificity of antibody-displaying nanoparticles. Proc Natl Acad Sci U S A 2013;110(9):3270–5. Agarwal R, Singh V, Jurney P, Shi L, Sreenivasan SV, Roy K. Mammalian cells preferentially internalize hydrogel nanodiscs over nanorods and use shapespecific uptake mechanisms. Proc Natl Acad Sci U S A 2013;110(43):17247–52. Galloway AL, Murphy A, DeSimone JM, Di J, Herrmann JP, Hunter ME, et al. Development of a nanoparticle-based influenza vaccine using the PRINT (R) technology. Nanomed Nanotechnol Biol Med 2013;9(4):523–31. Yue H, Wei W, Yue Z, Wang B, Luo N, Gao Y, et al. The role of the lateral dimension of graphene oxide in the regulation of cellular responses. Biomaterials 2012;33(16):4013–21. Varshochian R, Jeddi-Tehrani M, Mahmoudi AR, Khoshayand MR, Atyabi F, Sabzevari A, et al. The protective effect of albumin on bevacizumab activity and stability in PLGA nanoparticles intended for retinal and choroidal neovascularization treatments. Eur J Pharma Sci 2013;50(3-4):341–52.
[55] Yadav SC, Kumari A, Yadav R. Development of peptide and protein nanotherapeutics by nanoencapsulation and nanobioconjugation. Peptides 2011;32(1):173–87. [56] Singh M, Li XM, Wang HY, McGee JP, Zamb T, Koff W, et al. Immunogenicity and protection in small-animal models with controlled-release tetanus toxoid microparticles as a single-dose vaccine. Infect Immun 1997;65(5):1716–21. [57] Zhang W, Wang L, Liu Y, Chen X, Liu Q, Jia J, et al. Immune responses to vaccines involving a combined antigen–nanoparticle mixture and nanoparticleencapsulated antigen formulation. Biomaterials 2014;35(23):6086–97. [58] Zhang WF, Wang LY, Liu Y, Chen XM, Li JH, Yang TY, et al. Comparison of PLA microparticles and alum as adjuvants for H5N1 influenza split vaccine: adjuvanticity evaluation and preliminary action mode analysis. Pharmaceut Res 2014;31(4):1015–31. [59] Ju Y, Fan H, Liu J, Hu J, Li X, Li C, et al. Heat shock protein gp96 adjuvant induces T cell responses and cross-protection to a split influenza vaccine. Vaccine 2014;32(23):2703–11. [60] Wilson JT, Keller S, Manganiello MJ, Cheng C, Lee CC, Opara C, et al. pH-Responsive nanoparticle vaccines for dual-delivery of antigens and immunostimulatory oligonucleotides. ACS Nano 2013;7(5):3912–25. [61] Palucka K, Banchereau J. Dendritic-cell-based therapeutic cancer vaccines. Immunity 2013;39(1):38–48. [62] Shirota H, Klinman DM. Recent progress concerning CpG DNA and its use as a vaccine adjuvant. Exp Rev Vaccines 2014;13(2):299–312. [63] Richard I, Thibault M, De Crescenzo G, Buschmann MD, Lavertu M. Ionization behavior of chitosan and chitosan-DNA polyplexes indicate that chitosan has a similar capability to induce a proton-sponge effect as PEI. Biomacromolecules 2013;14(6):1732–40. [64] De Koker S, De Geest BG, Singh SK, De Rycke R, Naessens T, Van Kooyk Y, et al. Polyelectrolyte microcapsules as antigen delivery vehicles to dendritic cells: uptake, processing, and cross-presentation of encapsulated antigens. Angew Chem Int Ed 2009;48(45):8485–9. [65] Wu J, Wei W, Wang LY, Su ZG, Ma GH. Preparation of uniform-sized pH-sensitive quaternized chitosan microsphere by combining membrane emulsification technique and thermal-gelation method. Colloids Surf B 2008;63(2):164–75. [66] Wang YQ, Wu J, Fan QZ, Zhou M, Yue ZG, Ma GH, et al. Novel vaccine delivery system induces robust humoral and cellular immune responses based on multiple mechanisms. Adv Healthc Mater 2014;3(5):670–81. [67] Thomas SN, van der Vlies AJ, O’Neil CP, Reddy ST, Yu SS, Giorgio TD, et al. Engineering complement activation on polypropylene sulfide vaccine nanoparticles. Biomaterials 2011;32(8):2194–203. [68] Flach TL, Ng G, Hari A, Desrosiers MD, Zhang P, Ward SM, et al. Alum interaction with dendritic cell membrane lipids is essential for its adjuvanticity. Nat Med 2011;17(4), 479-U121. [69] Robert L, Atmar MD, David I, Bernstein MD, Clayton D, Harro MD, et al. Norovirus vaccine against experimental human Norwalk Virus illness. New Eng J Med 2011;365(23):2178–87. [70] Wu YB, Wei W, Zhou M, Wang YQ, Wu J, Ma GH, et al. Thermal-sensitive hydrogel as adjuvant-free vaccine delivery system for H5N1 intranasal immunization. Biomaterials 2012;33(7):2351–60.
Please cite this article in press as: Yue H, Ma G. Polymeric micro/nanoparticles: Particle design and potential vaccine delivery applications. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.07.100
772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820