10
UNCORRECTED PROOF 1 Aluminum hydroxide nanoparticles show a stronger vaccine adjuvant 2 activity than traditional aluminum hydroxide microparticles Xinran Q1 Li, Abdulaziz M. Aldayel, Zhengrong Cui 4 The University of Texas at Austin, College of Pharmacy, Pharmaceutics Division, Austin, TX 78712, USA 5 6 abstract article info 7 Article history: 8 Received 5 August 2013 9 Accepted 25 October 2013 10 Available online xxxx 11 12 13 14 Keywords: 15 Vaccine 16 Nanoparticles 17 Microparticles 18 Antibody responses 19 Local inammation 20 Aluminum hydroxide is used as a vaccine adjuvant in various human vaccines. Unfortunately, despite its favor- 21 able safety prole, aluminum hydroxide can only weakly or moderately potentiate antigen-specic antibody re- 22 sponses. When dispersed in an aqueous solution, aluminum hydroxide forms particulates of 120 μm. There is 23 increasing evidence that nanoparticles around or less than 200 nm as vaccine or antigen carriers have a more po- 24 tent adjuvant activity than large microparticles. In the present study, we synthesized aluminum hydroxide nano- 25 particles of 112 nm. Using ovalbumin and Bacillus anthracis protective antigen protein as model antigens, we 26 showed that protein antigens adsorbed on the aluminum hydroxide nanoparticles induced a stronger antigen- 27 specic antibody response than the same protein antigens adsorbed on the traditional aluminum hydroxide mi- 28 croparticles of around 9.3 μm. The potent adjuvant activity of the aluminum hydroxide nanoparticles was likely 29 related to their ability to more effectively facilitate the uptake of the antigens adsorbed on them by antigen- 30 presenting cells. Finally, the local inammation induced by aluminum hydroxide nanoparticles in the injection 31 sites was milder than that induced by microparticles. Simply reducing the particle size of the traditional alumi- 32 num hydroxide adjuvant into nanometers represents a novel and effective approach to improve its adjuvanticity. 33 © 2013 Published by Elsevier B.V. 34 35 36 37 38 1. Introduction 39 Many vaccines and antigens require an adjuvant to induce a strong 40 immune response [1]. Aluminum-containing adjuvants are approved 41 by the United States Food and Drug Administration for human use. 42 There are two main aluminum-containing adjuvants, aluminum hy- 43 droxide and aluminum phosphate. Aluminum hydroxide adjuvant is 44 composed of small primary bers with an average calculated dimension 45 of 4.5 × 2.2 × 10 nm, whereas the primary particles of aluminum phos- 46 phate adjuvant are around 50 nm [2]. In an aqueous solution, however, 47 the size of the primary particles of both aluminum hydroxide and alu- 48 minum phosphate becomes 120 μm as a result of aggregation [3]. 49 The mechanisms of immunopotentiation by aluminum-containing ad- 50 juvants have yet been fully elucidated. Originally, Glenny et al. (1931) 51 proposed that aluminum-containing adjuvants could form an antigen 52 depot in the injection site, from where the antigens are slowly released, 53 and thereby the adsorption efciency of antigens on aluminum- 54 containing adjuvants is thought to be critical [1]. However, data from 55 Hansen et al. showed that the tight binding of antigens onto 56 aluminum-containing adjuvants may signicantly reduce the amount 57 of antigens that can elute from the aluminum salts, resulting in a weak 58 antibody response [4]. Berthold et al. examined whether the full adsorp- 59 tion of antigens onto adjuvants is necessary by comparing the immune 60 responses induced by two vaccine formulations: Bacillus anthracis 61 recombinant protective antigen (PA) protein adsorbed onto aluminum 62 hydroxide with a high binding efciency, and PA admixed with alumi- 63 num phosphate with a negligible binding [5]. It was found that both for- 64 mulations induced comparable anti-PA antibody responses, suggesting 65 that the adjuvant activity of aluminum salts may not be entirely 66 depended on the adsorption of the antigens onto the adjuvants [5]. 67 Other mechanisms of immunopotentiation by aluminum-containing 68 adjuvants have been proposed as well [2,6,7]. HogenEsch [6] summa- 69 rized that aluminum-containing adjuvants may enhance immune re- 70 sponses by (i) direct or indirect stimulation of dendritic cells (DCs) 71 [8]; (ii) activation of complements [9]; and (iii) induction of the release 72 of chemokines [6,9]. More recently, aluminum-containing adjuvants 73 have been shown to promote caspase-1 activation and IL-1β secretion 74 through the NALP3 inammasomes [10]. 75 Due to their favorable safety prole, aluminum-containing adju- 76 vants have been widely used in human vaccines for decades. Unfortu- 77 nately, aluminum-containing adjuvants can only weakly or 78 moderately potentiate antigen-specic antibody responses and are gen- 79 erally considered incapable of helping antigens to induce cellular im- 80 mune responses [11]. As aforementioned, when dispersed in an 81 aqueous solution, both aluminum hydroxide and aluminum phosphate 82 form 120 μm particulates [3]. Recently, there had been extensive ef- 83 forts in identifying the relationship between the size of particulate vac- 84 cine carriers and their adjuvant activities [1214]. Although it remains 85 controversial as to what particle size is associated with the most potent 86 adjuvant activity, it is clear that the size of particulate vaccine carriers 87 signicantly affects their adjuvant activities, and there are data showing Journal of Controlled Release xxx (2013) xxxxxx Corresponding author. Tel.: +1 512 495 4758; fax: +1 512 471 7474. E-mail address: [email protected] (Z. Cui). COREL-06939; No of Pages 10 0168-3659/$ see front matter © 2013 Published by Elsevier B.V. http://dx.doi.org/10.1016/j.jconrel.2013.10.032 Contents lists available at ScienceDirect Journal of Controlled Release journal homepage: www.elsevier.com/locate/jconrel Please cite this article as: X. Li, et al., Aluminum hydroxide nanoparticles show a stronger vaccine adjuvant activity than traditional aluminum hydroxide microparticles, J. Control. Release (2013), http://dx.doi.org/10.1016/j.jconrel.2013.10.032 NANOMEDICINE

Aluminum hydroxide nanoparticles show a stronger vaccine adjuvant activity than traditional aluminum hydroxide microparticles

Embed Size (px)

Citation preview

1

2

3Q1

4

5

678910111213141516171819

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

Journal of Controlled Release xxx (2013) xxx–xxx

COREL-06939; No of Pages 10

Contents lists available at ScienceDirect

Journal of Controlled Release

j ourna l homepage: www.e lsev ie r .com/ locate / jconre l

NANOMEDICIN

E

Aluminum hydroxide nanoparticles show a stronger vaccine adjuvantactivity than traditional aluminum hydroxide microparticles

FXinran Li, Abdulaziz M. Aldayel, Zhengrong Cui ⁎The University of Texas at Austin, College of Pharmacy, Pharmaceutics Division, Austin, TX 78712, USA

O

⁎ Corresponding author. Tel.: +1 512 495 4758; fax: +E-mail address: [email protected] (Z. C

0168-3659/$ – see front matter © 2013 Published by Elsehttp://dx.doi.org/10.1016/j.jconrel.2013.10.032

Please cite this article as: X. Li, et al., Aluminhydroxide microparticles, J. Control. Release

Oa b s t r a c t

a r t i c l e i n f o

20

21

22

23

24

25

26

27

28

29

Article history:Received 5 August 2013Accepted 25 October 2013Available online xxxx

Keywords:VaccineNanoparticlesMicroparticlesAntibody responsesLocal inflammation

30

31

32

33

TED P

RAluminum hydroxide is used as a vaccine adjuvant in various human vaccines. Unfortunately, despite its favor-able safety profile, aluminum hydroxide can only weakly or moderately potentiate antigen-specific antibody re-sponses. When dispersed in an aqueous solution, aluminum hydroxide forms particulates of 1–20 μm. There isincreasing evidence that nanoparticles around or less than 200 nmas vaccine or antigen carriers have amore po-tent adjuvant activity than largemicroparticles. In the present study, we synthesized aluminumhydroxide nano-particles of 112 nm. Using ovalbumin and Bacillus anthracis protective antigen protein as model antigens, weshowed that protein antigens adsorbed on the aluminum hydroxide nanoparticles induced a stronger antigen-specific antibody response than the same protein antigens adsorbed on the traditional aluminum hydroxide mi-croparticles of around 9.3 μm. The potent adjuvant activity of the aluminum hydroxide nanoparticles was likelyrelated to their ability to more effectively facilitate the uptake of the antigens adsorbed on them by antigen-presenting cells. Finally, the local inflammation induced by aluminum hydroxide nanoparticles in the injectionsites was milder than that induced by microparticles. Simply reducing the particle size of the traditional alumi-numhydroxide adjuvant into nanometers represents a novel and effective approach to improve its adjuvanticity.

© 2013 Published by Elsevier B.V.

3435

C

61

62

63

64

65

66

67

68

69

70

71

72

73

74

75

76

77

78

79

80

81

82

83

84

UNCO

RRE1. Introduction

Many vaccines and antigens require an adjuvant to induce a strongimmune response [1]. Aluminum-containing adjuvants are approvedby the United States Food and Drug Administration for human use.There are two main aluminum-containing adjuvants, aluminum hy-droxide and aluminum phosphate. Aluminum hydroxide adjuvant iscomposed of small primary fiberswith an average calculated dimensionof 4.5 × 2.2 × 10 nm,whereas the primary particles of aluminumphos-phate adjuvant are around 50 nm [2]. In an aqueous solution, however,the size of the primary particles of both aluminum hydroxide and alu-minum phosphate becomes 1–20 μm as a result of aggregation [3].The mechanisms of immunopotentiation by aluminum-containing ad-juvants have yet been fully elucidated. Originally, Glenny et al. (1931)proposed that aluminum-containing adjuvants could form an antigendepot in the injection site, fromwhere the antigens are slowly released,and thereby the adsorption efficiency of antigens on aluminum-containing adjuvants is thought to be critical [1]. However, data fromHansen et al. showed that the tight binding of antigens ontoaluminum-containing adjuvants may significantly reduce the amountof antigens that can elute from the aluminum salts, resulting in a weakantibody response [4]. Berthold et al. examinedwhether the full adsorp-tion of antigens onto adjuvants is necessary by comparing the immuneresponses induced by two vaccine formulations: Bacillus anthracis

85

86

871 512 471 7474.ui).

vier B.V.

um hydroxide nanoparticles(2013), http://dx.doi.org/10.1

recombinant protective antigen (PA) protein adsorbed onto aluminumhydroxide with a high binding efficiency, and PA admixed with alumi-num phosphatewith a negligible binding [5]. It was found that both for-mulations induced comparable anti-PA antibody responses, suggestingthat the adjuvant activity of aluminum salts may not be entirelydepended on the adsorption of the antigens onto the adjuvants [5].Other mechanisms of immunopotentiation by aluminum-containingadjuvants have been proposed as well [2,6,7]. HogenEsch [6] summa-rized that aluminum-containing adjuvants may enhance immune re-sponses by (i) direct or indirect stimulation of dendritic cells (DCs)[8]; (ii) activation of complements [9]; and (iii) induction of the releaseof chemokines [6,9]. More recently, aluminum-containing adjuvantshave been shown to promote caspase-1 activation and IL-1β secretionthrough the NALP3 inflammasomes [10].

Due to their favorable safety profile, aluminum-containing adju-vants have been widely used in human vaccines for decades. Unfortu-nately, aluminum-containing adjuvants can only weakly ormoderately potentiate antigen-specific antibody responses and are gen-erally considered incapable of helping antigens to induce cellular im-mune responses [11]. As aforementioned, when dispersed in anaqueous solution, both aluminum hydroxide and aluminum phosphateform 1–20 μm particulates [3]. Recently, there had been extensive ef-forts in identifying the relationship between the size of particulate vac-cine carriers and their adjuvant activities [12–14]. Although it remainscontroversial as to what particle size is associated with themost potentadjuvant activity, it is clear that the size of particulate vaccine carrierssignificantly affects their adjuvant activities, and there are data showing

show a stronger vaccine adjuvant activity than traditional aluminum016/j.jconrel.2013.10.032

T

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

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

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

2 X. Li et al. / Journal of Controlled Release xxx (2013) xxx–xxx

NANOMEDICIN

E

UNCO

RREC

that particulate vaccine carriers of around 200 nm (or less) may be op-timal. For examples, Fifis et al. reported that ovalbumin (OVA)-conju-gated polystyrene particles of 230 nm induced stronger OVA-specificantibody and cellular immune responses than other larger OVA-conjugated polystyrene particles after intradermally injected into mice[13,15]. In a previous study, we also showed that small solid lipid nano-particles of 200 nm have a more potent adjuvant activity than largersolid lipid nanoparticles of 700 nm, when OVA as an antigen issurface-conjugated on them [16]. The ability of the smaller nanoparti-cles to more effectively facilitate the uptake of antigens carried bythem by antigen-presenting cells (APCs) and to up-regulate the expres-sion of major histocompatibility complex and co-stimulatory moleculesis likely related to their potent adjuvant activity [16]. Based on thesefindings,we proposed to improve the adjuvant activity of the traditionalaluminum-containing adjuvants by reducing their particle size. We hy-pothesized that small aluminum hydroxide nanoparticles of ≤200 nmhave amore potent vaccine adjuvant activity than the traditional alumi-numhydroxide adjuvantwith a particle size of 1–20 μm. To test this hy-pothesis, we synthesized aluminum hydroxide nanoparticles with amean diameter of 112 nm and compared their adjuvant activity withthat of the traditional aluminum hydroxide suspension with a mean di-ameter of 9.3 μm. OVA and B. anthracis PA protein were used as modelantigens.

2. Materials and methods

2.1. Materials

Dried aluminum hydroxide gel was from Spectrum (Gardena, CA).Aluminum chloride hexahydrate, sodium hydroxide, OVA, horseserum, Laemmli sample buffer, fluorescein-5(6)-isothiocyanate (FITC),sodium bicarbonate, sodium carbonate, phosphate-buffered saline(PBS), and incomplete Freund's adjuvant (IFA) were from Sigma-Aldrich (St. Louis, MO). Goat anti-mouse immunoglobulins (IgG) werefrom Southern Biotechnology Associates, Inc. (Birmingham, AL).Carbon-coated 400-mesh gridswere fromElectronMicroscopy Sciences(Hatfield, PA). Vectashield mounting medium with 4′,6-diamidino-2-phenylindole (DAPI) was from Vector Laboratories, Inc. (Burlingame,CA). B. anthracis PA protein was from List Biological Laboratories, Inc.(Campbell, CA). Bio-Safe™ Coomassie blue staining solution and Bio-Rad DC™ protein assay reagents were from Bio-Rad Laboratories (Her-cules, CA). GM-CSF was from R&D Systems, Inc. (Minneapolis, MN).Tissue-Tek® O.C.T. compound medium was from Sakura Finetek USA,Inc. (Torrance, CA). Cell culture medium and fetal bovine serum (FBS)were from Invitrogen (Carlsbad, CA).

2.2. Mice and cell lines

Female BALB/c and C57BL/6 mice, 6–8 weeks of age, were fromCharles River Laboratories, Inc. (Wilmington, MA). The OVA-expressing B16-OVA cell line was generously provided by Dr. Edith M.Lord and Dr. John Frelinger (University of Rochester Medical Center,Rochester, NY) [17] and cultured in RPMI1640 medium supplementedwith 5% FBS and 400 μg/ml of geneticin (Sigma). Mouse J774A.1macro-phage cells (# TIB-67™) were from the American Type and Culture Col-lection (Manassas, VA) and grown in DMEM supplemented with 10%FBS, 100 U/ml of penicillin and 100 μg/ml of streptomycin, all fromInvitrogen. DC2.4 cells (a mouse dendritic cell line) were originally cre-ated by Dr. Kenneth Rock (University of Massachusetts Medical School,Worcester, MA) [18] and grown in RPMI1640 medium supplementedwith 10% FBS, 100 U/ml of penicillin and 100 μg/ml of streptomycin.

2.3. Preparation of aluminum hydroxide nanoparticles and microparticles

Aluminum hydroxide nanoparticles of less than 200 nm were syn-thesized by reacting aluminum chloride with sodium hydroxide in a

Please cite this article as: X. Li, et al., Aluminum hydroxide nanoparticleshydroxide microparticles, J. Control. Release (2013), http://dx.doi.org/10.1

ED P

RO

OF

solution. An equal volume of a 3.6 mg/ml AlCl3·6H2O solution and a0.04 M NaOH solution were added into a glass vial, and a small volumeof 0.01 M NaOHwas added to adjust the pH to 7.0. After 20 min of stir-ring at room temperature, particle suspensionwas sonicated for 15 minto break down the particle size. A PD10 desalting column (AmershamBiosciences, Piscataway, NJ) was then used to remove sodium chloridein the suspension, and the eluted fractions were analyzed for nanopar-ticles by measuring the particle size using a Malvern Zetasizer NanoZS (Westborough, MA), and for aluminum content using a Varian 710-ES Inductively Coupled Plasma Optical Emission Spectrometer in theCivil Architectural and Environmental Engineering Department at TheUniversity of Texas at Austin. The fourth fraction with the highest con-centration of aluminum was used for further studies. Endotoxin wasnot detectable in the nanoparticle preparation with a ToxinSensor™chromogenic limulus amebocyte lysate endotoxin assay kit fromGenScript (Piscataway, NJ) [16]. Aluminum hydroxide microparticleswere prepared by dispersing dried aluminum hydroxide gel into sterilewater, followed by vigorous vortexing and 5 min of water-bath sonica-tion, if needed. The size of the microparticles was determined using aSympatec Helos laser diffraction instrument (Sympatec GmbH,Germany) equipped with a R3 lens.

2.4. Stability of aluminum hydroxide particles

The stability of aluminum hydroxide particles in suspension at 4 °Cor room temperature was initially examined before adsorption withproteins. The particles in suspension were kept at 4 °C for 30 days,and their sizes were measured on days 0 and 30. In another study, theparticles in suspension were kept at room temperature for 15 days,and their sizes were measured on days 0, 1, 7 and 15.

2.5. X-ray diffraction

The X-ray diffractograms of aluminum hydroxide particles were ob-tainedwith a Scintag X1 theta–theta powder diffractometer using Cu K-alpha radiation and a solid state Si(Li) detector in the TexasMaterials In-stitute X-ray Facility in the Chemical Engineering Department at TheUniversity of Texas at Austin.

2.6. Adsorption of protein antigens on aluminum hydroxide particles

The adsorption of proteins (OVAor PA) on aluminumhydroxide par-ticles was carried out by mixing the particles in suspension with theprotein in solution. Briefly, a certain volume of the protein solutionwas added into a tube (10 μg OVA or 4 μg PA), followed by the additionof particles in suspension at a weight ratio of 1:5 to 1:1 (OVA vs. parti-cles) or 1:5 (PA vs. particles). After 20 min of gentle stirring, the pro-tein–particle mixtures were stored at 4 °C or freeze-dried, if needed,before further use.

The OVA-adsorbed aluminum hydroxide nanoparticles were lyoph-ilized using a FreeZone plus 4.5 l cascade console freeze dry system(Labconco, Kansas City, MO). A proper cryoprotectant such as trehalose(2%, w/v) was needed to successfully freeze-dry the nanoparticles(Fig. S1A). In a short-term 28-day study and when stored as a lyophi-lized powder at 4 °C, the size of the lyophilized, OVA-adsorbed alumi-num hydroxide nanoparticles did not significantly change (Fig. S1B),indicating that the antigen-adsorbed aluminum hydroxide nanoparti-cles may be stored long-term as a lyophilized powder.

2.7. Transmission electron microscopy (TEM)

The OVA-adsorbed aluminum hydroxide nanoparticles were exam-ined using an FEI Tecnai Transmission Electron Microscope in the Insti-tute for Cellular andMolecular Biology (ICMB)Microscopy and ImagingFacility at The University of Texas at Austin [19]. Carbon-coated 400-mesh grids were activated for 1–2 min. One drop of the OVA-

show a stronger vaccine adjuvant activity than traditional aluminum016/j.jconrel.2013.10.032

T

205

206

207

208

209

210

211

212

213

214

215

216

217

218

219

220

221

222

223

224

225

226

227

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

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

3X. Li et al. / Journal of Controlled Release xxx (2013) xxx–xxx

NANOMEDICIN

E

UNCO

RREC

adsorbed nanoparticle suspension was deposited on the grids and incu-bated for 2 min at room temperature. The grids were washed withwater and dried for 1 min. Extra water was removed using filterpaper. The grids were then stained with uranyl acetate for 2 min,washed with water, and allowed to dry for 15 min before observation.

2.8. Scanning electron microscope (SEM)

The OVA-adsorbed aluminum hydroxide nanoparticles and micro-particles were examined using a Zeiss Supra 40 VP Scanning ElectronMicroscope in the ICMB Microscopy and Imaging Facility. One drop ofaluminum hydroxide particle suspension was deposited on the speci-men stub using a double stick carbon tape and allowed to dry overnight.The specimen stubswith sampleswere then placed in the sputter coaterchamber and coated with a very thin film of iridium before SEM exam-ination [20].

2.9. SDS-PAGE

SDS-PAGE assay was used to determine the extent towhich the pro-tein antigen was bound onto the aluminum hydroxide particles. Briefly,OVA (10 μg) was mixed with various amounts of aluminum hydroxideparticles in suspension (0, 1, 2, 5, 10, 20, 50, and 100 μg). The OVA–par-ticle mixtures were then lyophilized. The resultant powders werereconstituted in de-ionized water and mixed with Laemmli samplebuffer (62.5 mM Tris–HCl, pH 6.8, 25% glycerol, 2% SDS, and 0.01%Bromophenol Blue). Electrophoresis was performed with 7.5% Mini-PROTEAN® TGX™ precast polyacrylamide gels (Bio-Rad). Precisionplus protein standards were run along with the samples at 130 V for1 h. The gelswere stained in a Bio-Safe Coomassie blue staining solutionand scanned using a Kodak Image Station (Rochester, NY).

2.10. Preparation of bone marrow dendritic cells

Bone marrow dendritic cells (BMDCs) were generated from bonemarrow precursors from C57BL/6 mice [21]. Briefly, femur bones wereremoved from C57BL/6 mice and purified from surrounding tissues.The bones were left in 70% ethanol for 2 min for disinfection andwashedwith sterile PBS. After both ends of femur bones were removed,bone marrow was flushed out with PBS using a hypodermic needle at-tached to a syringe. After 3 washes with PBS, all leukocytes obtainedwere transferred into bacteriological petri dishes and cultured with10 ml of RPMI1640 medium supplemented with 10% FBS, 100 U/ml ofpenicillin, 100 μg/ml of streptomycin, 2-mercaptomethanol (50 μM)and granulocyte-macrophage-colony stimulating factor (GM-CSF)(100 ng/ml). Cells were allowed to grow at 37 °C under 5% CO2 for3 days, and another 10 ml of culture medium was added into the orig-inal dish. Onday6, half of the supernatantwas collected and centrifugedat 800 rpm for 4 min. Cell pellet was re-suspended in culture mediumand added back into the original dish. Cells on days 7 or 8 were usedfor further studies. In order to examine the purity of BMDCs, the cellswere stained with antibodies against CD11c (BD Pharmingen, SanDiego, CA) [21], and analyzed using a Guava EasyCyte 8HTmicrocapillary flow cytometer (Millipore Corporation, Hayward, CA).A high purity of 86.5% BMDCs was obtained after 8 days in culturemedium.

2.11. Uptake of the OVA-adsorbed particles by BMDCs, DC2.4 cells andJ77A4.1 cells in culture

In vitro uptake studies were carried out using OVA that was pre-labeled with FITC [16]. BMDCs, DC2.4 or J77A4.1 cells (50,000 cells/well) were seeded into 24-well plates and allowed to grow overnightat 37 °C, 5% CO2. FITC-labeled OVA-particles were added into the cellculture and incubated at 37 °C under 5% CO2 or at 4 °C. After further in-cubation (3 h for DC2.4 and J77A4.1 cells, 6 h for BMDCs), cells were

Please cite this article as: X. Li, et al., Aluminum hydroxide nanoparticleshydroxide microparticles, J. Control. Release (2013), http://dx.doi.org/10.1

ED P

RO

OF

washed with PBS (10 mM, pH 7.4) three times, lyzed with Triton X-100 (0.17%, v/v) and then applied to a BioTek Synergy HT microplatereader to measure the fluorescence intensity. Endocytosis is inhibitedat 4 °C. Therefore, a subtraction of the fluorescence intensity of thecells incubated at 4 °C from the fluorescence intensity of the cells incu-bated at 37 °C, 5% CO2, allowed us to estimate the amount of FITC-OVAthat was internalized.

2.12. Fluorescence microscopy

DC2.4 cells (1.5 × 104) were plated on poly-D-lysine-coated glasscoverslips overnight. FITC-labeled OVA-adsorbed particles were addedand incubated with the cells for 30 to 60 min at 37 °C, 5% CO2. Cellswere then washed with PBS, fixed in 3% paraformaldehyde for 20 minat room temperature, followed by three times of washwith PBS. Cover-slips were mounted on the slides using Vectashield mounting mediumwith DAPI. Fluorescent images were acquired using an Olympus BX60Biological Microscope (Center Valley, PA).

2.13. Immunization studies

All animal studies were carried out following the National Institutesof Health guidelines for care and use of laboratory animals. The animalprotocol was approved by the Institutional Animal Care and Use Com-mittee at The University of Texas at Austin. When OVA was used asthe antigen, female BALB/c mice (18–20 g) were subcutaneouslyinjected with OVA-adsorbed aluminum hydroxide particles once aweek for three consecutive weeks. The dose of the OVA was 10 μg permouse per injection; 20 (or 50) μg per mouse per injection for the par-ticles. Sterile PBS or OVA (10 μg) dissolved in PBS was used as controls.Twenty seven days after the first dose, mice were bled for antibodyassay. When PA was used as the antigen, female BALB/c mice(18–20 g) were immunized subcutaneously with PA-adsorbed alumi-num hydroxide particles on days 0 and 14. As negative controls, micewere injected with sterile PBS or PA alone. The dose of PA was 4 μgper mouse per injection, and the dose of the particles was 20 μg permouse per injection. Mice were bled one week and one month afterthe second immunization for antibody assay. Specific antibody levelsin serum samples were determined using enzyme-linked immunosor-bent assay (ELISA) as previously described [19].

2.14. Tumor prevention assays

Female C57BL/6 mice (18–20 g) were immunized with OVA-adsorbed particles, PBS, or OVA alone on days 0, 7, and 14 by subcutane-ous injection. The dose of OVA was 10 μg per mouse per injection, andthe particles were 20 μg. As a positive control, mice were also immu-nized with OVA adjuvanted with IFA (50 μl per mouse per injection).On day 21, B16-OVA cells (50,000/mouse) were subcutaneouslyinjected in the right flank of the mice (different from the sites of immu-nization). Tumor growth was monitored daily. Tumor size was mea-sured using a caliper, and the volume was calculated using thefollowing equation [22]: tumor volume (mm3) = 1/2 [length × (-width)2].

2.15. Histological examination

BALB/c mice were injected with PA adsorbed aluminum hydroxideparticles on day 0 and 19. As negative controls, mice were injectedwith sterile PBS or PA alone. On day 40, micewere euthanized for histo-logical examination. The hair on the injection site was initially removedusing Nair® lotion (Church and Dwight Co, Princeton, NJ). The skin atthe injection sites, including skin and muscle tissues, were removedand spread out on a piece of index paper. The tissue and paper togetherwere cut into a 1 cm × 1 cmsquare and transferred to tissue cryomolds(25 mm × 20 mm × 5 mm, Sakura Finetek USA, Inc.). Any residual

show a stronger vaccine adjuvant activity than traditional aluminum016/j.jconrel.2013.10.032

T

321

322

323

324

325

326

327

328

329

330

331

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

373

374

375

376

377

378

379

380

381

4 X. Li et al. / Journal of Controlled Release xxx (2013) xxx–xxx

NANOMEDICIN

E

spaces in the cryomolds were filled with Tissue-Tek® O.C.T. compoundmedium and fixed in the vapor of liquid nitrogen for 10 min. After theO.C.T. compound medium was frozen into a solid white color, thewhole cryomoles were removed and wrapped with aluminum foil.The prepared samples were stored at −80 °C for cryostat sectioningand staining with Hematoxylin and eosin (H&E, Sigma, St. Louis, MO)in the Histology and Tissue Analysis Facility in the Dell Pediatric Re-search Institute, The University of Texas at Austin [23].

2.16. Statistics

Statistical analyses were completed using analysis of variancefollowed by Fisher's protected least significant difference procedure. Ap-value of ≤0.05 (two-tail) was considered statistically significant.

3. Result and discussion

3.1. Synthesis and characterization of aluminum hydroxide particles

In order to evaluate the effect of the size of aluminumhydroxide par-ticles on their adjuvant activity, aluminum hydroxide nanoparticles andmicroparticles with mean diameters of 112 ± 6.2 nm and9.3 ± 2.2 μm, respectively, were prepared (Fig. 1A). At neutral pH, thezeta potentials of both particles were positive (Fig. 1A), but the zeta po-tential of the aluminumhydroxidemicroparticles was less positive thanthat of the aluminum hydroxide nanoparticles (Fig. 1A). The positivecharge of aluminum hydroxide particles was likely due to the metallichydroxyls on their surface, which could accept protons and show a pos-itive zeta potential [24]. Since the reduction of particle size increases thetotal surface area of the particles [25], the aluminum hydroxide nano-particles are expected to have a relatively larger surface area than themicroparticles, and thusmoremetallic hydroxyl groups on their surface,resulting in a more positive zeta potential. The aluminum hydroxidenanoparticles were stable when stored at 4 °C (or room temperature

UNCO

RREC

0

10

20

30

40

50

60

1

10

100

1000

10000

100000

NPs MPs

Zet

a p

ote

nti

al (

mV

)

Par

ticl

e si

ze (

nm

)

Particle sizeZeta potential

A

C

Fig. 1. Physical properties of aluminum hydroxide nanoparticles (NPs) andmicroparticles (MPshydroxide nanoparticles and microparticles when stored in suspension at 4 °C for 30 days. (C–Data shown in A and B are mean ± S.D. (n = 3).

Please cite this article as: X. Li, et al., Aluminum hydroxide nanoparticleshydroxide microparticles, J. Control. Release (2013), http://dx.doi.org/10.1

ED P

RO

OF

(Fig. S2)), whereas the microparticles were slightly less stable(Fig. 1B), likely because the zeta potential of the nanoparticles wasN30 mV, whereas the zeta potential of the microparticles wasb30 mV, at which the electrostatic repulsion was not strong enoughto prevent aggregation [26]. The X-ray powder patterns of aluminumhydroxide particles are shown in Fig. 1C–D. The nanoparticles werecompletely amorphous (Fig. 1C). The microparticles were mostly crys-talline Al(OH)3 (Fig. 1D), although the large peak in the left indicatedthat some amorphous AlO(OH) materials existed as well (Fig. 1D).

3.2. Characterization of OVA-adsorbed aluminum hydroxide particles

Shown in Fig. 2A are the size and zeta potentials of the aluminumhy-droxide nanoparticles and microparticles after the adsorption of OVAprotein at a 1:2 ratio (OVA vs. particle, w/w). The mean diameters ofthe OVA-adsorbed nanoparticles and microparticles were129 ± 20 nm and 9.4 ± 1.7 μm, respectively; and their zeta potentialswere 16 ± 1.8 and−23 ± 1.9, respectively. The sizes of both particlesincreased after the adsorption of OVA. Since OVA is net negativelycharged at neutral pH (isoelectric point (pI), 4.7), after the adsorptionof OVA, the zeta potentials of the resultant nanoparticles became lesspositive, and the zeta potential of microparticles even changed frompositive to negative (Fig. 2A).

Shown in Fig. 2B are the fractions of free OVA when a fixed amountof OVAwasmixedwith increasing amounts of the aluminumhydroxidenanoparticles or microparticles. As expected, the fraction of unboundOVA decreased when the amount of aluminum hydroxide particlesadded was increased. When the ratio of OVA to nanoparticles was de-creased to 1:2 and 1:5, theOVA protein bands can no longer be detectedon the SDS-PAGE, indicating that all the OVA proteins were bound onthe particles when OVA and nanoparticles were mixed at 1:2 ratio orlower. The adsorption of theOVA to the aluminumhydroxidemicropar-ticles was not as extensive as to the nanoparticles. Only when theweight ratio of microparticles to OVA was increased to 5:1, the OVA

B

1

10

100

1000

10000

100000

NPs MPs

Par

ticl

e si

ze (

nm

)

Day 0Day 30

p = 0.85

p = 0.002

D

). (A). Particle sizes (open bar) and zeta potentials (●). (B). The stability of the aluminumD). X-ray diffractograms of aluminum hydroxide nanoparticles (C) and microparticles (D).

show a stronger vaccine adjuvant activity than traditional aluminum016/j.jconrel.2013.10.032

CTED P

RO

OF

382

383

384

385

386

387

388

389

390

391

392

393

394

395

396

397

398

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

424

425

426

427

-50

-40

-30

-20

-10

0

10

20

30

1

100

10000

OVA-NPs OVA-MPs

Zet

a p

ote

nti

al (

mV

)

Par

ticl

e si

ze (

nm

)

Particle size

Zeta potential

1 2 3 4 5 6

A B

D OVA-NPsC OVA-MPs

Fig. 2. Physical characterization of aluminum hydroxide nanoparticles and microparticles after adsorption of OVA. (A). Particle sizes (open bar) and zeta potentials (●) of OVA-adsorbedaluminum hydroxide nanoparticles (OVA-NPs) and OVA-adsorbed aluminum hydroxide microparticles (OVA-MPs). Data shown are mean ± S.D. (n = 3). (B). The binding efficiency ofOVA to the aluminumhydroxide nanoparticles andmicroparticles. Shown is the fraction of free (unbound) OVA at various OVAs to particle ratios (w/w) as determined from the intensitiesof the protein bands on the SDS-PAGE (inset, OVA to nanoparticle ratio: lane 1, 10:0; lane 2, 10:1; lane 3, 5:1; lane 4, 2:1; lane 5, 1:1; lane 6, 1:2). (C). A representative SEMpicture of OVA-MPs. (D). A representative TEM picture of OVA-NPs.

5X. Li et al. / Journal of Controlled Release xxx (2013) xxx–xxx

NANOMEDICIN

E

UNCO

RREprotein bands were no longer detectable using SDS-PAGE (Fig. 2B). The

mechanisms of the adsorption of OVA to aluminum hydroxide particlesare likely twofold: (1) the electrostatic interaction between OVA andaluminum hydroxide particles because they have opposite net chargesat neutral pH; and (2) ligand exchange as OVA protein contains up totwo phosphate groups, which could strongly bind to aluminum insteadof a hydroxyl group [27]. The higher protein adsorption capacity of thealuminum hydroxide nanoparticles is consistent with the larger totalsurface area of the nanoparticles, which contain more binding sites forprotein adsorption. The relatively smaller total surface area of the alu-minum hydroxide microparticles limited the amount of proteinsadsorbed on them. Besides the effect of the surface area, the zeta poten-tial of the aluminum hydroxide particles may also have contributed tothe adsorption capacity. The zeta potential of the aluminum hydroxidenanoparticles was more positive than that of the microparticles(Fig. 1A). Therefore, the aluminum hydroxide nanoparticles may haveattracted more OVA proteins to their surface.

Shown in Fig. 2C is a representative SEM picture of the OVA-adsorbed aluminum hydroxide microparticles. A representative TEMpicture of the OVA-adsorbed aluminum hydroxide nanoparticles isshown in Fig. 2D. A representative SEM picture of the OVA-adsorbedaluminumhydroxide nanoparticles is also available in Fig. S3. The nano-particles are rod-shaped with a smooth surface, whereas the micropar-ticles have a rough surface and are in irregular shapes.

428

429

430

431

432

Please cite this article as: X. Li, et al., Aluminum hydroxide nanoparticleshydroxide microparticles, J. Control. Release (2013), http://dx.doi.org/10.1

3.3. OVA-adsorbed small aluminum hydroxide nanoparticles induced astronger OVA-specific antibody immune response than OVA-adsorbed largealuminum hydroxide microparticles

Aluminum hydroxide particles with diameters in the range of1–20 μm have been widely used in human vaccines [11]. Previousdata showed that nanoparticles with a mean diameter of around orless than 200 nm have a more potent adjuvant activity than larger par-ticles [16,19]. To test whether small aluminum hydroxide nanoparticlesof less than 200 nm can help an antigen to induce a stronger immuneresponse than larger aluminum hydroxide microparticles, the anti-OVA immune responses induced by OVA-adsorbed aluminum hydrox-ide nanoparticles and microparticles at the OVA to particles ratio of1:2 were compared. Data in Fig. 3A showed that the anti-OVA IgGlevel in mice that were immunized with the OVA-adsorbed aluminumhydroxide nanoparticles was significantly higher than that in micethat were immunized with OVA-adsorbed microparticles or OVA alone(at 100-fold dilution, p b 0.001, OVA-NPs vs. OVA; p = 0.018, OVA-NPs vs. OVA-MPs; p = 0.05, OVA alone vs. OVA-MPs).

There were data showing that nanoparticles as a carrier may allowproteins to induce antitumor responses. For example, Falo et al. evaluat-ed the ability of particulate antigens against tumor growth and foundthat immunization ofmicewith OVA-conjugated iron beadsmore effec-tively prevented the growth of tumor cells that overexpress OVA (B16-OVA) in the immunized mice, as compared to immunization with OVAalone [28]. Falo et al. also concluded that the antitumor immunity wasessentially contributed by specific CD8+ T cells [28]. Accordingly, atumor prevention study was carried out to evaluate the ability of our

show a stronger vaccine adjuvant activity than traditional aluminum016/j.jconrel.2013.10.032

RECTED P

RO

OF

433

434

435

436

437

438

439

440

441

442

443

444

445

446

447

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

474

475

476

0

0.1

0.2

0.3

0.4

0.5

0.6

100 1000 10000

An

ti-O

VA

IgG

(O

D45

0)

Serum dilution factor

PBS

OVA

OVA-NPs

OVA-MPs

a

b

c

0

1

2

3

100X 1000X 10,000X

Dilution factor

An

ti-O

VA

IgG

(O

D45

0)

PBS

OVA

OVA-NPs

OVA-MPs

a

b

1

10

100

1000

Tu

mo

r V

olu

me

(mm

3 )

PBS OVA NPs MPs IFA

A B

C

Fig. 3. OVA-adsorbed aluminum hydroxide nanoparticles induced a stronger OVA-specific antibody response and more effectively protected the growth of OVA-expressing tumor cellsthan OVA-adsorbed aluminum hydroxide microparticles. (A). Serum total IgG response. Mice (n = 5) were dosed with OVA-NPs or OVA-MPs at an OVA to particle ratio of 1:2 (w/w)on days 0, 7 and 14. Total anti-OVA IgG level in serum samples was measured on day 27 (a p b 0.001, OVA vs. OVA-NPs; b p = 0.02, OVA-NPs vs. OVA-MPs; also, OVA vs. OVA-NPs,p = 0.005 at 1000-fold dilution, p = 0.05 at 10,000-fold dilution). (B). The volumes of B16-OVA tumors in mice immunized with OVA-NPs or OVA-MPs. Tumor volumes shown are31 days after tumor cell implantation. B16-OVA tumor cells were s.c. implanted into mice (n = 5) 10 days after the third immunization. OVA adjuvanted with IFA (OVA/IFA) was a pos-itive control (Notes: A volume of 1 mm3 is assigned tomice that do not have tumors because the Y-axis in this figure is in log scale). (C). Serum total anti-OVA IgG responses.Mice (n = 5)were dosed with OVA-NPs or OVA-MPs at an OVA to particle ratio of 1:5 (w/w) on days 0, 7 and 14. Anti-OVA IgG level in serum samples was measured on day 24 (a–c p = 0.02 at alldilutions, OVA-NPs vs. OVA-MPs).

6 X. Li et al. / Journal of Controlled Release xxx (2013) xxx–xxx

NANOMEDICIN

E

UNCO

ROVA-adsorbed aluminum hydroxide nanoparticles in preventing tumorgrowth. Twenty-one days after C57BL/6 mice were immunized withOVA-adsorbed aluminum hydroxide nanoparticles or microparticles,they were challenged with the OVA-expressing B16-OVA tumor cells,and the tumor growth was monitored. As shown in Fig. 3B, 31 daysafter tumor cell injection, tumors were detected only in one of the 5mice that were immunized with the OVA-adsorbed aluminum hydrox-ide nanoparticles. In contrast, all mice immunized with the OVA-adsorbed microparticles or with OVA alone developed tumors, indicat-ing that the immune responses induced by OVA-adsorbed aluminumhydroxide nanoparticles can inhibit tumor growth. However, the anti-tumor activity was likely antibody-mediated because we were notable to consistently detect OVA-specific CD8+ T cell response in the im-munized mice (data not shown).

As shown in Fig. 2B, when the ratio of particles to OVA was 1:2, theOVAproteinwas fully adsorbedon the nanoparticles, but the adsorptionof the OVA to the aluminumhydroxidemicroparticleswas not as exten-sive. Only when themicroparticles to OVAweight ratio reached 5:1, theOVA protein was fully adsorbed on both nanoparticles and microparti-cles. To make sure that the stronger immune response induced by theOVA-adsorbed aluminum hydroxide nanoparticles shown in Fig. 3Awas not due to the difference in the extent to which the OVA wasadsorbed on the aluminumhydroxide nanoparticles andmicroparticles,

Please cite this article as: X. Li, et al., Aluminum hydroxide nanoparticleshydroxide microparticles, J. Control. Release (2013), http://dx.doi.org/10.1

another mouse immunization study was carried out by immunizingmice with OVA-adsorbed aluminum hydroxide nanoparticles or micro-particles at an aluminum hydroxide to OVA weight ratio 5:1. As shownin Fig. 3C, the serum anti-OVA IgG response induced by the OVA-adsorbed aluminum hydroxide nanoparticles was still stronger thanthat induced by the OVA-adsorbed aluminum hydroxidemicroparticles(p = 0.02 at all three dilutions, OVA-NPs vs. OVA-MPs), demonstratingthat the stronger immune response induced by the OVA-adsorbed alu-minum hydroxide nanoparticles was truly due to their smaller size.

3.4. PA-adsorbed aluminum hydroxide nanoparticles induced a strongerPA-specific antibody response than PA-adsorbed aluminum hydroxidemicroparticles

To test whether the potent adjuvant activity of the aluminum hy-droxide nanoparticles was unique to OVA as an antigen, we completeda similar immunization study with the anthrax PA protein. Anthrax isa toxin-mediated disease, and anthrax toxin is consisted of three pro-teins, PA, lethal factor, and edema factor [29]. PA proteins form aheptamer on the surface of cells, fromwhich the edema factor and lethalfactor enter cells [29]. Therefore, the induction of anti-PA antibody re-sponses is critical and sufficient for a vaccine to protect against anthrax[30]. PA was absorbed on aluminum hydroxide particles at a particle to

show a stronger vaccine adjuvant activity than traditional aluminum016/j.jconrel.2013.10.032

477

478

479

480

481

482

483

484

485

486

487

488

489

490

491

492

493

494

495

496

497

498

499

500

501

502

503

504

7X. Li et al. / Journal of Controlled Release xxx (2013) xxx–xxx

NANOMEDICIN

E

PA ratio of 5:1 (w/w). Themean diameters of the resultant PA-adsorbedaluminum hydroxide nanoparticles and microparticles were204 ± 25 nm and 7.1 ± 3.4 μm, respectively (Fig. 4A). Mice werethen immunizedwith the PA-adsorbed aluminumhydroxidenanoparti-cles or microparticles on days 0 and 14. One week after the first dose,anti-PA IgG was not detectable in any mice (data not shown). Oneweek after the second dose, significant anti-PA IgG responses were de-tected in mice that were immunized with the PA-adsorbed aluminumhydroxide nanoparticles or microparticles, but the levels of the anti-PA IgG were not different (Fig. 4B). However, four weeks after the sec-ond immunization, the anti-PA IgG level in mice that were immunizedwith the PA-adsorbed aluminum hydroxide nanoparticles was signifi-cantly higher than that in mice that were immunized with the PA-adsorbed aluminum hydroxide microparticles (Fig. 4C). The anti-PA

UNCO

RRECT

0

0.5

1

1.5

2

10,000 100,000 1,000,000

An

ti-P

A Ig

G (

OD

450)

PBS

PA

PA-NPs

PA-MPs

Serum dilution factor

-30

-25

-20

-15

-10

-5

0

0

1000

2000

3000

4000

PA-NPs PA-MPs

Zet

a p

ote

nti

al (

mV

)

Par

ticl

e si

ze (

nm

)

Particle size

Zeta potential

0

0.5

1

1.5

2

W1 W4

An

ti-P

A Ig

G le

vel (

OD

450)

PBSPAPA-NPsPA-MPs

f

eE

b

c

*

*

C

A

Fig. 4. PA-adsorbed aluminum hydroxide nanoparticles induced a stronger PA-specific antibodybar) and zeta potentials (●) of PA-adsorbed aluminum hydroxide nanoparticles (PA-NPs) andsponses. Mice (n = 5) were dosed with PA-adsorbed aluminum hydroxide nanoparticles (PA-per injection). Anti-PA IgG and IgG1 levels were measured one week (B) and four weeks afterthat were diluted 10,000-fold (a p = 0.0002, PA vs. PA-NPs; p = 0.001, PA vs. PA-MPs). In Cp = 0.007, PA-NPs vs. PA-MPs at 100,000-fold dilution; and p = 0.0002, PA-NPs vs. PA-MPs at 1parison of the anti-PA IgG levels in mice one week (W1) and four weeks (W4) after the last(n = 5). e,f p b 0.05, week 1 vs.week 4 for both PA-NPs and PA-MPs.

Please cite this article as: X. Li, et al., Aluminum hydroxide nanoparticleshydroxide microparticles, J. Control. Release (2013), http://dx.doi.org/10.1

IgG1 levels 4 weeks after the second immunization are shown inFig. 4D. A significantly higher level of anti-PA IgG1 was detected inmice that were immunized with PA-adsorbed aluminum hydroxidenanoparticles than inmice that were immunizedwith PA-adsorbed alu-minum hydroxide microparticles (Fig. 4D). PA-specific IgE was not de-tected 4 weeks after immunization with PA-adsorbed aluminumhydroxide nanoparticles or microparticles (data not shown). The kinet-ics of the total serum anti-PA IgG levelswithin 4 weeks after the last im-munization is shown in Fig. 4E. It is clear that during the 4-week periodafter the second immunization, the total serum anti-PA IgG level signif-icantly increased in mice that were immunized with the PA-adsorbedaluminum hydroxide nanoparticles (p = 0.005, week 1 vs. week 4),but decreased in mice that were immunized with the PA-adsorbed alu-minum hydroxide microparticles (p = 0.005, week 1 vs. week 4).

ED P

RO

OF

10,000 100,000

An

ti-P

A Ig

G1

(OD

450)

Serum dilution factor

PBS

PA

PA-NPs

PA-MPs

0

0.5

1

1.5

2

PB

S

PA

PA

-NP

s

PA

-MP

s

An

ti-P

A Ig

G (

OD

450)

a

d

D

B

0

0.5

1

1.5

2

2.5

3

3.5

4

4.5

response than PA-adsorbed aluminum hydroxide microparticles. (A). Particle sizes (openPA-adsorbed aluminum hydroxide microparticles (PA-MPs). (B–D). Anti-PA antibody re-NPs) or microparticles (PA-MPs), or PA alone on days 0 and 14 (PA dose, 4 μg per mousethe second immunization (C–D). Data shown in B are anti-PA IgG level in serum samplesat 10,000-fold dilution, b p = 0.0002, PA vs. PA-NPs; c p = 0.002, PA-NPs vs. PA-MPs. *,000,000-fold dilution. In D at 100,000 dilution, d p b 0.05, PA-NPs vs. PA-MPs. (E). A com-immunization (serum samples were diluted 10,000-fold). Data shown are mean ± S.E.

show a stronger vaccine adjuvant activity than traditional aluminum016/j.jconrel.2013.10.032

505

506

507

508

509

510

511

512

513

514

515

516

517

518

519

520

521

522

523

524

525

526

527

528

529

8 X. Li et al. / Journal of Controlled Release xxx (2013) xxx–xxx

NANOMEDICIN

E

3.5. The uptake of OVA-adsorbed aluminum hydroxide particles by antigen-presenting cells in culture

One important step for an antigen to induce an immune response isits uptake byAPCs. Therefore, we evaluated the extent towhichDCs andmacrophages, two critical APCs, can take upOVA as an antigen adsorbedon aluminumhydroxide nanoparticles or microparticles. BMDCs, DC2.4,or J774A.1 cells in culture were incubated with fluorescein-labeled OVAadsorbed on aluminum hydroxide nanoparticles or microparticles forup to 6 h, and the % of OVA internalized by the cells was determined.As shown in Fig. 5A, in all three cell lines, more OVA was internalizedwhen adsorbed on the aluminum hydroxide nanoparticles than whenadsorbed on the aluminum hydroxide microparticles. The fluorescence

UNCO

RRECT

0

0.3

0.6

0.9

BMDCs

% O

VA

inte

rnal

ized

I. OVA

II. OVA-NPs

III. OVA-MPs

DAPI Fluores

*

A

B

Fig. 5. The uptake of OVA-adsorbed aluminum hydroxide nanoparticles and microparticles. (A)minumhydroxide nanoparticles ormicroparticles adsorbedwith FITC-labeledOVAwere incubathe % of FITC-labeled OVA that was internalized was determined (*p b 0.05, OVA-NPs vs. OVA-cencemicroscopic pictures of DC2.4 cells after incubationwith aluminumhydroxide nanoparticOVA-NPs; (III). OVA-MPs. Cell nuclei were stained with DAPI (blue). (For interpretation of thearticle.)

Please cite this article as: X. Li, et al., Aluminum hydroxide nanoparticleshydroxide microparticles, J. Control. Release (2013), http://dx.doi.org/10.1

microscopic pictures in Fig. 5B are also supportive of the data inFig. 5A and may explain why the aluminum hydroxide nanoparticleswere more effective than the microparticles in facilitating the uptakeof OVA by DC2.4 cells. Green fluorescence signal, an indication of the lo-cation of the OVA protein, was detected only inside cells that were incu-bated with OVA-adsorbed aluminum hydroxide nanoparticles, but notin cells that were incubated with OVA-adsorbed aluminum hydroxidemicroparticles (Fig. 5B). In fact, for cells that were incubated with theOVA-adsorbed aluminum hydroxide microparticles, almost all fluores-cence signals were extracellular (Fig. 5B), and it appeared that someOVA-adsorbed aluminum hydroxide microparticles were even largerthan the cells (Fig. 5B), which may explain why the OVA adsorbed onthe aluminum hydroxide microparticles did enter the cells (Fig. 5A).

ED P

RO

OF

DC2.4 J774A.1

OVA

NPs

MPs

cein Overlay

*

*

The uptake of OVA-NPs and OVA-MPs by BMDCs, DC2.4, and J774A.1 cells in culture. Alu-tedwith BMDCs,DC2.4 cells or J774A.1 cells for 3–6 h at 37 °Cwith 5%of CO2 or at 4 °C, andMPs in all three cells). Data shown are mean ± S.D. (n = 6). (B). Representative fluores-les ormicroparticles pre-adsorbedwith FITC-labeled OVA. (I). FITC-labeled OVA alone; (II).references to color in this figure legend, the reader is referred to the web version of this

show a stronger vaccine adjuvant activity than traditional aluminum016/j.jconrel.2013.10.032

T

530

531

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

576

577

578

579

580

581

582

583

584

585

586

587

588

589

590

591

592

593

594

595

596

597

598

599

9X. Li et al. / Journal of Controlled Release xxx (2013) xxx–xxx

NANOMEDICIN

E

C

Previous data showed that antigens eluted from adjuvants are takenup by DCs bymacropinocytosis, while those remaining adsorbed are in-ternalizedwith adjuvant particles by phagocytosis [31]. Because close to100% of the OVA was adsorbed on the aluminum hydroxide nanoparti-cles, it is likely that phagocytosis or endocytosis was the predominantmechanism for the internalization of the OVA that was adsorbed onthe aluminum hydroxide nanoparticles. In contrast, only less than 20%OVA was adsorbed onto the microparticles (at the OVA to particleratio of 1:2, w/w). The small percentage of OVA that was internalizedby DC2.4 cells incubated with the OVA-adsorbed aluminum hydroxidemicroparticles was probably from themacropinocytosis of the unboundOVA and the OVA that was eluted from the microparticles. Morefieldet al. showed that DCs are able to internalize particles with a diameterlarger than that of cells [32], but we could not find any internalizationof theOVA-adsorbed aluminumhydroxidemicroparticles usingfluores-cence microscopy. Previously, Kanchan and Panda also reported thatnanoparticles (200–600 nm) were more efficiently taken up by macro-phages in comparison tomicroparticles (2–8 μm) [33]. Thus, we suspectthat the ability of the aluminum hydroxide nanoparticles to more effec-tively facilitate the uptake of the OVA adsorbed on them by APCs is re-lated to their potent adjuvant ability (Figs. 3–4).

Finally, a comparison of the internalization of the OVA by themacro-phages (J774A.1 cells) and DCs (BMDCs and DC2.4 cells) indicated thatthe percentage of OVA adsorbed on the aluminumhydroxidemicropar-ticles that was internalized by the macrophages was relatively higherthan by the DCs (Fig. 5A). This finding is in agreement with a previousreport that macrophages can effectively take up particles larger than500 nm, whereas DCs are more effective in taking up smaller nanopar-ticles (b200 nm) [34].

3.6. Aluminum hydroxide nanoparticles induced milder local inflammationreactions than aluminum hydroxide microparticles

Aluminum adjuvants have been administered safely to humanssince 1932 [35]. Adverse reactions that have been reported with vac-cines adjuvanted with aluminum salts are generally local reactions in-cluding subcutaneous (s.c.) nodule, granulomatous inflammation, andsterile abscesses [36]. In order to preliminarily evaluate the safety

UNCO

RRE

PBS

PA-NPs

Fig. 6. Representative H&E histograms of the skin in the injection sites. BALB/c mice were s.c. introls, mice were injected with sterile PBS or PA alone. On day 40, mice were euthanized and thestaining (scale bar, 200 μm).

Please cite this article as: X. Li, et al., Aluminum hydroxide nanoparticleshydroxide microparticles, J. Control. Release (2013), http://dx.doi.org/10.1

ED P

RO

OF

profile of our aluminum hydroxide nanoparticles, the injection siteswere examined histologically. As shown in Fig. 6, microparticles andnanoparticles both induced local cutaneous inflammation in the injec-tion sites when examined 40 days after the last injection, but the in-flammation induced by the PA-adsorbed microparticles was muchmore severe, as indicated by the greater number of neutrophils accumu-lated in the injection sites and the more pronounced epidermal hyper-plasia. Clearly the aluminum hydroxide nanoparticles are lessproinflammatory than the microparticles.

4. Conclusions

In the present study, we synthesized aluminum hydroxide nanopar-ticles with a mean diameter of 112 nm and showed that the adjuvantactivity of the aluminum hydroxide nanoparticles was more potentthan that of the traditional aluminum hydroxide microparticles(~9 μm). The specific antibody responses induced by protein antigensadjuvanted with the aluminum hydroxide nanoparticles were strongerand more durable than that induced by the same amount of antigensadjuvanted with the traditional aluminum hydroxide microparticles.Themore potent adjuvant activity of the aluminumhydroxide nanopar-ticles may be partially attributed to their ability to more extensivelybind to antigens and increase the uptake of the antigens adsorbed onthem by APCs. Moreover, the aluminum hydroxide nanoparticles wereless proinflammatory than the microparticles in the injection sites.The new aluminum hydroxide nanoparticles have the potential to bedeveloped into a more effective and safer adjuvant to formulate newvaccines and reformulate existing vaccines.

Acknowledgments

This work was supported in part by the National Institutes of Healthgrants (AI070538, AI078304, and Al105789) to Z. Cui. A.M.A is a KingAbdullah International Medical Research Center (KAIMRC) scholar andis supported by the KAIMRC scholarship program. The authors wouldlike to thank Dr. Hugh Smyth in the College of Pharmacy at The Univer-sity of Texas at Austin for kindly allowing us to use the Sympatec Heloslaser diffraction instrument available in his lab.

PA

PA-MPs

jectedwith PA-adsorbed aluminumhydroxide particles on days 0 and 19. As negative con-skin samples in the injection sites were collected for cryostat sectioning followed by H&E

show a stronger vaccine adjuvant activity than traditional aluminum016/j.jconrel.2013.10.032

T

600

601

602

603

604605606607608609610611612613614615616617618619620621622623624625626627628629630631632633634635636637638639640641642643644645646647648649650651652653

654655656657658659660661662663664665666667668669670671672673674675676677678679680681682683684685686687688689690691692693694695696697698699700701702703704705706707708709

710

10 X. Li et al. / Journal of Controlled Release xxx (2013) xxx–xxx

NANOMEDICIN

E

RREC

Appendix A. Supplementary data

Supplementary data to this article can be found online at http://dx.doi.org/10.1016/j.jconrel.2013.10.032.

References

[1] D.T. O'Hagan, M.L. MacKichan, M. Singh, Recent developments in adjuvants for vac-cines against infectious diseases, Biomol. Eng. 18 (2001) 69–85.

[2] I.Z. Romero Mendez, Y. Shi, H. HogenEsch, S.L. Hem, Potentiation of the immune re-sponse to non-adsorbed antigens by aluminum-containing adjuvants, Vaccine 25(2007) 825–833.

[3] S.L. Hem, H. Hogenesch, Relationship between physical and chemical properties ofaluminum-containing adjuvants and immunopotentiation, Expert Rev. Vaccine 6(2007) 685–698.

[4] B. Hansen, A. Sokolovska, H. HogenEsch, S.L. Hem, Relationship between thestrength of antigen adsorption to an aluminum-containing adjuvant and the im-mune response, Vaccine 25 (2007) 6618–6624.

[5] I. Berthold, M.L. Pombo, L.Wagner, J.L. Arciniega, Immunogenicity inmice of anthraxrecombinant protective antigen in the presence of aluminum adjuvants, Vaccine 23(2005) 1993–1999.

[6] H. HogenEsch, Mechanisms of stimulation of the immune response by aluminumadjuvants, Vaccine 20 (Suppl. 3) (2002) S34–S39.

[7] L.S. Jones, L.J. Peek, J. Power, A. Markham, B. Yazzie, C.R. Middaugh, Effects of adsorp-tion to aluminum salt adjuvants on the structure and stability of model protein an-tigens, J. Biol. Chem. 280 (2005) 13406–13414.

[8] J.W. Mannhalter, H.O. Neychev, G.J. Zlabinger, R. Ahmad, M.M. Eibl, Modulation ofthe human immune response by the non-toxic and non-pyrogenic adjuvant alumin-ium hydroxide: effect on antigen uptake and antigen presentation, Clin. Exp.Immunol. 61 (1985) 143–151.

[9] M. Ulanova, A. Tarkowski, M. Hahn-Zoric, L.A. Hanson, The common vaccine adju-vant aluminum hydroxide up-regulates accessory properties of human monocytesvia an interleukin-4-dependent mechanism, Infect. Immun. 69 (2001) 1151–1159.

[10] S.C. Eisenbarth, O.R. Colegio, W. O'Connor, F.S. Sutterwala, R.A. Flavell, Crucial rolefor the Nalp3 inflammasome in the immunostimulatory properties of aluminiumadjuvants, Nature 453 (2008) 1122–1126.

[11] R.K. Gupta, Aluminum compounds as vaccine adjuvants, Adv. Drug Deliv. Rev. 32(1998) 155–172.

[12] I. Gutierro, R.M. Hernandez, M. Igartua, A.R. Gascon, J.L. Pedraz, Size dependent im-mune response after subcutaneous, oral and intranasal administration of BSA loadednanospheres, Vaccine 21 (2002) 67–77.

[13] M. Kalkanidis, G.A. Pietersz, S.D. Xiang, P.L. Mottram, B. Crimeen-Irwin, K.Ardipradja, M. Plebanski, Methods for nano-particle based vaccine formulationand evaluation of their immunogenicity, Methods 40 (2006) 20–29.

[14] J. Wendorf, J. Chesko, J. Kazzaz, M. Ugozzoli, M. Vajdy, D. O'Hagan, M. Singh, A com-parison of anionic nanoparticles and microparticles as vaccine delivery systems,Hum. Vaccine 4 (2008) 44–49.

[15] T. Fifis, A. Gamvrellis, B. Crimeen-Irwin, G.A. Pietersz, J. Li, P.L. Mottram, I.F.McKenzie, M. Plebanski, Size-dependent immunogenicity: therapeutic and protec-tive properties of nano-vaccines against tumors, J. Immunol. 173 (2004) 3148–3154.

[16] X.R. Li, B.R. Sloat, N. Yanasarn, Z.R. Cui, Relationship between the size of nanoparti-cles and their adjuvant activity: data from a study with an improved experimentaldesign, Eur. J. Pharm. Biopharm. 78 (2011) 107–116.

[17] A.A. Lugade, J.P. Moran, S.A. Gerber, R.C. Rose, J.G. Frelinger, E.M. Lord, Local radia-tion therapy of B16 melanoma tumors increases the generation of tumorantigen-specific effector cells that traffic to the tumor, J. Immunol. 174 (2005)7516–7523.

UNCO

Please cite this article as: X. Li, et al., Aluminum hydroxide nanoparticleshydroxide microparticles, J. Control. Release (2013), http://dx.doi.org/10.1

ED P

RO

OF

[18] Z.H. Shen, G. Reznikoff, G. Dranoff, K.L. Rock, Cloned dendritic cells can present ex-ogenous antigens on both MHC class I and class II molecules, J. Immunol. 158(1997) 2723–2730.

[19] B.R. Sloat, M.A. Sandoval, A.M. Hau, Y. He, Z. Cui, Strong antibody responses inducedby protein antigens conjugated onto the surface of lecithin-based nanoparticles, J.Control. Release 141 (2010) 93–100.

[20] A.Y. Hao, Y.Y. Geng, Q. Xu, Z.Y. Lu, L. Yu, Study of different effects on foaming processof biodegradable PLA/starch composites in supercritical/compressed carbon dioxide,J. Appl. Polym. Sci. 109 (2008) 2679–2686.

[21] M.B. Lutz, N. Kukutsch, A.L. Ogilvie, S. Rossner, F. Koch, N. Romani, G. Schuler, An ad-vanced culture method for generating large quantities of highly pure dendritic cellsfrom mouse bone marrow, J. Immunol. Methods 223 (1999) 77–92.

[22] M.M. Tomayko, C.P. Reynolds, Determination of subcutaneous tumor size in athymic(nude) mice, Cancer Chemother. Pharmacol. 24 (1989) 148–154.

[23] J. Lim, Y. Kim, W. Lee, M. Kim, E.J. Lee, C.S. Kang, K. Han, Fresh-frozen, optimal cut-ting temperature (OCT) compound-embedded bonemarrow aspirates: a reliable re-source for morphological, immunohistochemical and molecular examinations, Int. J.Lab. Hematol. 32 (2010) e34–e39.

[24] W. Stumm, L. Sigg, B. Sulzberger, Chemistry of the Solid–Water Interface: Processesat the Mineral–Water and Particle–Water Interface in Natural Systems, Wiley, NewYork, 1992.

[25] S. Lowell, S. Lowell, Characterization of Porous Solids and Powders: Surface Area,Pore Size, and Density, Kluwer Academic Publishers, Dordrecht; Boston, 2004.

[26] A.R. Bumb, C.A.S., M. Bernardo, P.J. Dobson, P. Choyke, M.W. Brechbiel, Multi-modalSuperparamagnetic Nanoprobe: Harnessing Magnetic, Nuclear, and Optical Powerfor Diagnostic Imaging Applications, NSTI-Nanotech, 32010.

[27] S. Iyer, H. HogenEsch, S.L. Hem, Effect of the degree of phosphate substitution in alu-minum hydroxide adjuvant on the adsorption of phosphorylated proteins, Pharm.Dev. Technol. 8 (2003) 81–86.

[28] L.D. Falo Jr., M. Kovacsovics-Bankowski, K. Thompson, K.L. Rock, Targeting antigeninto the phagocytic pathway in vivo induces protective tumour immunity, Nat.Med. 1 (1995) 649–653.

[29] K.A. Bradley, J. Mogridge, M. Mourez, R.J. Collier, J.A. Young, Identification of the cel-lular receptor for anthrax toxin, Nature 414 (2001) 225–229.

[30] B.R. Sloat, Z. Cui, Evaluation of the immune response induced by a nasal anthrax vac-cine based on the protective antigen protein in anaesthetized andnon-anaesthetized mice, J. Pharm. Pharmacol. 58 (2006) 439–447.

[31] H.L. Davis, R.Weeratna, T.J.Waldschmidt, L. Tygrett, J. Schorr, A.M. Krieg, CpG DNA isa potent enhancer of specific immunity in mice immunized with recombinant hep-atitis B surface antigen, J. Immunol. 160 (1998) 870–876.

[32] G.L. Morefield, A. Sokolovska, D. Jiang, H. HogenEsch, J.P. Robinson, S.L. Hem, Role ofaluminum-containing adjuvants in antigen internalization by dendritic cells in vitro,Vaccine 23 (2005) 1588–1595.

[33] V. Kanchan, A.K. Panda, Interactions of antigen-loaded polylactide particles withmacrophages and their correlation with the immune response, Biomaterials 28(2007) 5344–5357.

[34] S.D. Xiang, A. Scholzen, G. Minigo, C. David, V. Apostolopoulos, P.L. Mottram, M.Plebanski, Pathogen recognition and development of particulate vaccines: doessize matter? Methods 40 (2006) 1–9.

[35] N. Goto, H. Kato, J. Maeyama, K. Eto, S. Yoshihara, Studies on the toxicities of alumin-ium hydroxide and calcium phosphate as immunological adjuvants for vaccines,Vaccine 11 (1993) 914–918.

[36] N. Goto, H. Kato, J. Maeyama, M. Shibano, T. Saito, J. Yamaguchi, S. Yoshihara, Localtissue irritating effects and adjuvant activities of calcium phosphate and aluminiumhydroxide with different physical properties, Vaccine 15 (1997) 1364–1371.

show a stronger vaccine adjuvant activity than traditional aluminum016/j.jconrel.2013.10.032