formulation and evaluation of oral microparticulate ... formulation and evaluation o… · eudragit...

7
Please cite this article in press as: Tawde SA, et al. Formulation and evaluation of oral microparticulate ovarian cancer vaccines. Vaccine (2012), http://dx.doi.org/10.1016/j.vaccine.2012.05.073 ARTICLE IN PRESS G Model JVAC 13243 1–7 Vaccine xxx (2012) xxx–xxx Contents lists available at SciVerse ScienceDirect Vaccine jou rn al h om epa ge: www.elsevier.com/locate/vaccine Formulation and evaluation of oral microparticulate ovarian cancer vaccines 1 Suprita A. Tawde a , Lipika Chablani a , Archana Akalkotkar a , Cherilyn D’Souza a , Periasamy Selvaraj b , Q1 Martin J. D’Souza a,2 3 a Vaccine Nanotechnology Laboratory, Department of Pharmaceutical Sciences, Mercer University, College of Pharmacy and Health Sciences, 3001 Mercer University Drive, Atlanta, GA 30341, USA 4 5 b Department of Pathology and Laboratory Medicine, Emory University, 7309 Woodruff Memorial Research Building, 101 Woodruff Circle, Atlanta, GA 30322, USA 6 7 a r t i c l e i n f o 8 9 Article history: 10 Received 16 April 2012 11 Accepted 25 May 2012 12 Available online xxx 13 Keywords: 14 Microparticles 15 Ovarian cancer 16 Oral delivery 17 M-cells 18 ID8 cells 19 Spray drying 20 a b s t r a c t Ovarian cancer is the fifth most leading cause of cancer related deaths in women in the US. Cus- tomized immunotherapeutic strategies may serve as an alternative method to control the recurrence or progression of ovarian cancer and to avoid severe adverse effects of chemotherapy. In this study, a microparticulate vaccine using whole cell lysate of a murine ovarian cancer cell line, ID8 was prepared with the use of a spray dryer. These particles were designed for oral delivery using enteric polymers such as methacrylic copolymer, Eudragit ® FS30D and hydroxyl propyl methyl cellulose acetate succinate. These particles were targeted for uptake via microfold cell (M-cell) in Peyer’s patches of small intestine using M-cell targeting ligand, Aleuria aurantia lectin. The interleukins (ILs) such as IL-2 and IL-12 were added to the vaccine formulation to further enhance the immune response. The particles obtained were of 1.58 ± 0.62 m size with a charge of 12.48 ± 2.32 mV. The vaccine efficacy was evaluated by adminis- tering the particles via oral route to C57BL/6 female mice. At the end of vaccination, mice were challenged with live tumor cells. Vaccinated mice showed significant (around six-fold) retardation of tumor volume in comparison to non-vaccinated animals for 3 weeks after the tumor challenge (p < 0.001). The serum IgG antibody levels were found to be elevated in case of vaccinated animals in comparison to non-vaccinated group (p < 0.05). Analysis of IgG1 titers (indicative of Th2 response) and IgG2a titers (indicative of Th1 response) showed a mixed Th1 and Th2 immune response in case vaccine alone and Th2 response in case of vaccine with interleukins group. Moreover, CD8+ T-cell, CD4+ T-cell and B-cell populations in different lymphatic organs were elevated in case of vaccinated mice. Thus, whole cell lysate vaccine microparticles formulated by spray drying could trigger humoral as well as cellular immune response when adminis- tered orally. Such vaccine could potentially be an effective treatment for patients with residual tumor or high tumor-relapse probability. © 2012 Elsevier Ltd. All rights reserved. 1. Introduction 21 Ovarian cancer is the most lethal gynecological cancer in the 22 US. Only about 10% of ovarian cancers are usually found in early 23 stages. Patients with epithelial tumors, which account for approx- 24 imately 90% of ovarian cancer, generally have poor overall survival 25 [1]. First-line treatment for advanced cancer involves surgery 26 followed by chemotherapy. However, cancer relapses within rel- 27 atively short periods of time even after treatment [2]. Therefore, 28 alternative approaches, immunotherapy is being investigated to 29 prevent relapse of cancer. 30 Several vaccines are underway in clinical trials and most of 31 them have not progressed beyond phase I/II studies. Even though 32 antigen-specific response is obtained with different approaches of 33 Corresponding author. Tel.: +1 678 547 6353; fax: +1 678 547 6423. E-mail address: dsouza [email protected] (M.J. D’Souza). antigen specific immunization, there is no consistency in clinical 34 benefit [3,4]. In this study, we have investigated whether oral vac- 35 cination with microparticles containing the ovarian cancer antigens 36 can prevent/retard ovarian cancer growth. A murine ovarian can- 37 cer cell line, ID8 was used as a source of antigens as it correlates 38 closely to human ovarian cancer cell lines in signaling pathways 39 and results in development of tumor in mice models similar to 40 human ovarian cancer [5]. Despite of advancement in vaccine tech- 41 nology, the whole cell lysate vaccine still remains a very promising 42 approach as it can overcome the demerits associated with a sin- 43 gle antigen/epitope vaccine. Whole cell lysate provides a pool of 44 tumor-associated antigens (TAAs) which can induce both CD8+ and 45 CD4+ T cells [6]. Therefore, we proceeded with a whole cell lysate 46 of ID8 cells to prepare the vaccine for this study. 47 Oral vaccine delivery is an attractive mode of immunization 48 because of its ease of administration, low manufacturing costs and 49 patient compliance. However, for an oral vaccine to be effective, 50 it must withstand gastro-intestinal degradation. For this purpose, 51 0264-410X/$ see front matter © 2012 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.vaccine.2012.05.073

Upload: others

Post on 18-Oct-2020

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Formulation and evaluation of oral microparticulate ... Formulation and evaluation o… · Eudragit ® FS 30 D was gen- ... Briefly, overnight coating of 159 the lysate (100 g/well)

G

J

F1

SQ1

M2

3

a

G4

5b6

7

a8

9

A10

R11

A12

A

13

K14

M15

O16

O17

M18

I19

S20

121

22

U23

s24

i25

[26

f27

a28

a29

p30

31

t32

a33

0h

ARTICLE IN PRESS Model

VAC 13243 1–7

Vaccine xxx (2012) xxx– xxx

Contents lists available at SciVerse ScienceDirect

Vaccine

jou rn al h om epa ge: www.elsev ier .com/ locate /vacc ine

ormulation and evaluation of oral microparticulate ovarian cancer vaccines

uprita A. Tawdea, Lipika Chablania, Archana Akalkotkara, Cherilyn D’Souzaa, Periasamy Selvarajb,artin J. D’Souzaa,∗

Vaccine Nanotechnology Laboratory, Department of Pharmaceutical Sciences, Mercer University, College of Pharmacy and Health Sciences, 3001 Mercer University Drive, Atlanta,A 30341, USADepartment of Pathology and Laboratory Medicine, Emory University, 7309 Woodruff Memorial Research Building, 101 Woodruff Circle, Atlanta, GA 30322, USA

r t i c l e i n f o

rticle history:eceived 16 April 2012ccepted 25 May 2012vailable online xxx

eywords:icroparticlesvarian cancerral delivery-cells

D8 cellspray drying

a b s t r a c t

Ovarian cancer is the fifth most leading cause of cancer related deaths in women in the US. Cus-tomized immunotherapeutic strategies may serve as an alternative method to control the recurrenceor progression of ovarian cancer and to avoid severe adverse effects of chemotherapy. In this study, amicroparticulate vaccine using whole cell lysate of a murine ovarian cancer cell line, ID8 was preparedwith the use of a spray dryer. These particles were designed for oral delivery using enteric polymers suchas methacrylic copolymer, Eudragit® FS30D and hydroxyl propyl methyl cellulose acetate succinate.These particles were targeted for uptake via microfold cell (M-cell) in Peyer’s patches of small intestineusing M-cell targeting ligand, Aleuria aurantia lectin. The interleukins (ILs) such as IL-2 and IL-12 wereadded to the vaccine formulation to further enhance the immune response. The particles obtained wereof 1.58 ± 0.62 �m size with a charge of 12.48 ± 2.32 mV. The vaccine efficacy was evaluated by adminis-tering the particles via oral route to C57BL/6 female mice. At the end of vaccination, mice were challengedwith live tumor cells. Vaccinated mice showed significant (around six-fold) retardation of tumor volumein comparison to non-vaccinated animals for 3 weeks after the tumor challenge (p < 0.001). The serum IgGantibody levels were found to be elevated in case of vaccinated animals in comparison to non-vaccinatedgroup (p < 0.05). Analysis of IgG1 titers (indicative of Th2 response) and IgG2a titers (indicative of Th1

response) showed a mixed Th1 and Th2 immune response in case vaccine alone and Th2 response in caseof vaccine with interleukins group. Moreover, CD8+ T-cell, CD4+ T-cell and B-cell populations in differentlymphatic organs were elevated in case of vaccinated mice. Thus, whole cell lysate vaccine microparticlesformulated by spray drying could trigger humoral as well as cellular immune response when adminis-tered orally. Such vaccine could potentially be an effective treatment for patients with residual tumor orhigh tumor-relapse probability.

34

35

36

37

38

39

40

41

42

43

44

. Introduction

Ovarian cancer is the most lethal gynecological cancer in theS. Only about 10% of ovarian cancers are usually found in early

tages. Patients with epithelial tumors, which account for approx-mately 90% of ovarian cancer, generally have poor overall survival1]. First-line treatment for advanced cancer involves surgeryollowed by chemotherapy. However, cancer relapses within rel-tively short periods of time even after treatment [2]. Therefore,lternative approaches, immunotherapy is being investigated torevent relapse of cancer.

Please cite this article in press as: Tawde SA, et al. Formulation and evaluathttp://dx.doi.org/10.1016/j.vaccine.2012.05.073

Several vaccines are underway in clinical trials and most ofhem have not progressed beyond phase I/II studies. Even thoughntigen-specific response is obtained with different approaches of

∗ Corresponding author. Tel.: +1 678 547 6353; fax: +1 678 547 6423.E-mail address: dsouza [email protected] (M.J. D’Souza).

45

46

47

264-410X/$ – see front matter © 2012 Elsevier Ltd. All rights reserved.ttp://dx.doi.org/10.1016/j.vaccine.2012.05.073

© 2012 Elsevier Ltd. All rights reserved.

antigen specific immunization, there is no consistency in clinical

benefit [3,4]. In this study, we have investigated whether oral vac-

cination with microparticles containing the ovarian cancer antigens

can prevent/retard ovarian cancer growth. A murine ovarian can-

cer cell line, ID8 was used as a source of antigens as it correlates

closely to human ovarian cancer cell lines in signaling pathways

and results in development of tumor in mice models similar to

human ovarian cancer [5]. Despite of advancement in vaccine tech-

nology, the whole cell lysate vaccine still remains a very promising

approach as it can overcome the demerits associated with a sin-

gle antigen/epitope vaccine. Whole cell lysate provides a pool of

tumor-associated antigens (TAAs) which can induce both CD8+ and

CD4+ T cells [6]. Therefore, we proceeded with a whole cell lysateof ID8 cells to prepare the vaccine for this study.

ion of oral microparticulate ovarian cancer vaccines. Vaccine (2012),

Oral vaccine delivery is an attractive mode of immunization 48

because of its ease of administration, low manufacturing costs and 49

patient compliance. However, for an oral vaccine to be effective, 50

it must withstand gastro-intestinal degradation. For this purpose, 51

Page 2: Formulation and evaluation of oral microparticulate ... Formulation and evaluation o… · Eudragit ® FS 30 D was gen- ... Briefly, overnight coating of 159 the lysate (100 g/well)

ING Model

J

2 ccine

e52

a53

w54

c55

F56

c57

t58

o59

f60

f61

c62

f63

m64

t65

o66

o67

p68

i69

l70

c71

I72

r73

a74

75

f76

u77

278

279

80

U81

C82

r83

F84

e85

w86

f87

l88

p89

m90

a91

B92

r93

294

95

M96

i97

S98

p99

h100

fi101

r102

2103

104

p105

k106

o107

a108

A109

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

ARTICLEVAC 13243 1–7

S.A. Tawde et al. / Va

nteric polymers such as methacrylic copolymer Eudragit® FS 30 Dnd hydroxyl propyl methyl cellulose acetate succinate (HPMCAS)ere used. Ano et al. have reported a formulation of oral vac-

ine of inactivated Vibrio cholerae by spray drying using Eudragit®

S30D [7]. This polymer was found to be a good candidate forontrolled release of antigens and resulted in vibriocidal antibodyiters. We prepared the particles utilizing this polymer along withther excipients by using an optimized microparticulate formulaor oral vaccine delivery by spray drying [8]. To target our vaccineormulation to M-cells in the Peyer’s patches of the intestine, M-ell targeting agent, Aleuria aurantia lectin (AAL) was used in theormulation [9,10]. In addition, inclusion of immunostimulatory

olecules such as IL-2 and IL-12 was evaluated in order to enhancehe overall potency of the formulated vaccines [11,12]. It has beenbserved that specific immune response by tumor cell vaccines isften impaired due to deficiency of co-stimulatory signals. Vaccinesrepared with cancer cells secreting IL-2 or IL-12 have resulted

n better immune response when compared to non-secreting cellines. IL-2 has been efficiently used in immunotherapy against can-ers [13]. Moreover, higher anti-tumor effects were reported whenL-2 was delivered as an adjuvant in a slow-releasing depot formather than a free form [11,13]. Incorporation of IL-2 in particleddresses this issue seen with free form of IL-2.

In the present study, we demonstrate the efficacy of oral vaccineormulations which was evaluated in vivo in mouse tumor model,sing the ID8 murine ovarian cancer cell line as a solid tumor model.

. Materials and methods

.1. Materials

ID8 cell line was kindly provided by Dr. Katherine Roby, Kansasniversity Medical Center, Kansas City, KS. Six to eight week-old57BL/6 female mice were purchased from Charles River Labo-atories, Wilmington, MA. HPMCAS was purchased from AQOAT,MC Biopolymers, Philadelphia, PA. Eudragit® FS 30 D was gen-rously gifted by Evonik industries, Parsipanny, NJ. Mouse plasmaas obtained from Biochemed, Winchester, VA. AAL was obtained

rom Vector Labs, Inc., Burlingame, CA. Recombinant murine inter-eukins, IL-2 (5 × 106 units/mg) and IL-12 (1 × 107 units/mg) wereurchased from Peprotech Inc., Rocky Hill, NJ. Flow cytometry cellarkers were purchased from eBioscience, San Diego, CA. Goat

nti-mouse HRP-IgG and anti-IgG subtypes were purchased fromethyl Laboratories, Montgomery, TX and Sigma, St. Louis, MOespectively. All other materials used were of analytical grade.

.2. Preparation of whole cell lysate

The murine ovarian cancer ID8 cells were cultured in Dulbecco’sodified Eagles Medium (DMEM) supplemented with 5 �g/ml

nsulin, 5 �g/ml transferrin and 5 ng/ml sodium selenite (Sigma,t. Louis, MO) [14]. The confluent cells were washed with coldhosphate buffered saline (PBS). The flasks were then treated withypotonic buffer (10 mM Tris and 10 mM NaCl) and subjected tove 15 min freeze–thaw cycles at temperatures of −80 ◦C and 37 ◦Cespectively to obtain the cell lysate [15,16].

.3. Characterization of the lysate

The protein content of lysate was analyzed using Bio-Rad DCTM

rotein assay. The lysate was screened for presence of the only

Please cite this article in press as: Tawde SA, et al. Formulation and evaluathttp://dx.doi.org/10.1016/j.vaccine.2012.05.073

nown antigen, SP17 by western blot analysis [17]. Briefly, 25 �gf lysate protein was resolved by SDS-PAGE gel electrophoresisnd subjected to overnight wet-transfer on to a PVDF membrane.fter blocking, the membrane was treated with primary mouse

PRESSxxx (2012) xxx– xxx

anti-SP17 antibody (kindly provided by Dr. Chiriva-Internati, Texas

Tech University Health Science Center, Lubbock, TX), followed by

incubation with a horseradish peroxidase-conjugated secondary

antibody. Finally, the membrane was incubated with Enhanced

Chemiluminescence (abcam, Cambridge, MA), then exposed to

imaging film.

2.4. Preparation of vaccine microparticles

The vaccine formulation was prepared by spray drying tech-

nique [8–10,18–24]. Briefly, HPMCAS and Eudragit® FS 30D were

dissolved in an alkaline solution, followed by addition of chitosan

glycol. Mouse plasma was added to the polymeric solution at pH 7.0

as a source of albumin. Trehalose, tween 20 and AAL were added

to the solution, followed by addition of the lysate (5%, w/w). In

case of vaccine with interleukins formulations, 4 × 105 U of IL-2 and

8 × 105 U of IL-2 were added to this feed mixture [11,12]. This aque-

ous solution was spray dried using Buchi B-191 Mini Spray Dryer(Buchi Corporation, New Castle, DE).

2.5. Characterization of microparticles

Microparticles were visualized by using scanning electron

microscope (JEOL JSM 5800LV, Tokyo, Japan) at EM Core facility,Emory University, Atlanta. The particles were characterized for size

and charge, using laser particle counter (Spectrex PC-2000) and

Malvern zeta sizer (ZEN 1600) respectively. Loading efficiency was

determined by extracting the proteins in PBS and analyzing them

using Biorad DCTM protein assay in comparison to placebo particles.

2.6. Immunization

The immunogenicity of microparticulate vaccine was evalu-

ated using C57BL/6 female mice model. Animal experiments were

carried out as per approved protocols by Mercer University’s Insti-

tutional Committee for the care and Use of Laboratory animals.

Animals (n = 8) were administered with microparticles as one prime

dose followed by one booster after 1 week and thereafter by 8

boosters with an interval of 2 weeks. Briefly, 5 mg of microparticles

were suspended in citrate buffer (pH 4.0) and administered orally

using oral gavage. Three different formulations such as placebo,

vaccine and vaccine with interleukins were evaluated for this pur-

pose.

2.7. Tumor challenge study

One week after the last vaccination, the mice were challenged

s.c. with 1 × 107 live ID8 cells [25]. The cells were suspended in

incomplete media and injected into the right back flank of mice.

Tumor development was monitored using digital vernier calipers.

The mice were euthanized whenever the tumor ulcerated or tumors

exceeded a size of 15 mm in any of the perpendicular diame-

ters. The tumor volume (V) was determined by using the formula,

V = 1/2(length)(width)2 [26,27].

2.8. Assessment of humoral (B-cell mediated) immune response

in serum

The blood samples were collected prior to each dose of vacci-

nation. Serum was analyzed by ELISA. Briefly, overnight coating of

the lysate (100 �g/well) was done in a 96 well plate. The plate was

blocked with non-fat dry milk. After washing, the plate was incu-

ion of oral microparticulate ovarian cancer vaccines. Vaccine (2012),

bated with 1:10 dilution of serum samples. HRP-tagged secondary 162

anti-mouse goat IgG was then added to each well, and incubated 163

for 1 h. TMB substrate reagent (3,3′, 5,5′′-tetramethyl benzidine) 164

(BD OptEIATM, BD Biosciences, CA) was added and the plate was 165

Page 3: Formulation and evaluation of oral microparticulate ... Formulation and evaluation o… · Eudragit ® FS 30 D was gen- ... Briefly, overnight coating of 159 the lysate (100 g/well)

IN PRESSG Model

J

ccine xxx (2012) xxx– xxx 3

a166

o167

i168

a169

p170

o171

T172

y173

2174

l175

176

d177

e178

(179

g180

a181

1182

t183

l184

m185

a186

y187

2188

189

l190

T191

a192

b193

p194

S195

w196

e197

3198

3199

200

t201

w202

w203

t204

b205

e206

b207

o208

i209

I210

a211

w212

m213

[214

t215

a216

t217

v218

3219

220

o221

p222

Fig. 1. Characterization of lysate and microparticles prepared. (A) Western blotanalysis of ID8 lysates for SP17 with BSA as a negative control, indicating the pres-

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

ARTICLEVAC 13243 1–7

S.A. Tawde et al. / Va

gain incubated for 30 min. The reaction was stopped by additionf 4 N H2SO4. The plate was read using microplate reader (BioTeknstruments Inc., Winooski, VT) at 450 nm. In case of IgG subtypesnalysis, serum samples were analyzed in dilution of 1:50. Thelate was then incubated with goat anti-mouse IgG subtype IgG1r IgG2a, followed incubation with HRP-conjugated anti-goat IgG.hereafter, same procedure as described above for IgG titers anal-sis was followed.

.9. Determination of T-cell and B-cell based immune response inymphatic organs

A separate group of mice was vaccinated in similar way asescribed in Section 2.6. At the end of vaccination, mice wereuthanized and lymphatic organs such as the spleen, lymph nodesaxillary and inguinal) and bone marrows were harvested. The sin-le cell suspension of these organs were stimulated for 5 dayst 37 ◦C with mitomycin-treated tumor cells in a ratio 10:1 with0 U/ml of recombinant murine IL-2 [28]. At the end of 5-days,he cells were washed with Hank’s balanced salt solution andabeled with anti-mouse CD4 PE, anti-mouse CD8a FITC and anti-

ouse/human CD45R (B220) FITC (for B-cells). The cells werenalyzed for the specific cell populations by flow cytometric anal-sis using BD accuri® C6 flow cytometer.

.10. Statistical analysis

Serum IgG subtype titers and flow-cytometry results were ana-yzed using one-way analysis of variance (ANOVA) followed byukey’s multiple comparison test. Tumor volume measurementsnd serum IgGs were analyzed by two-way ANOVA followedy Bonferroni multiple comparisons. All statistical analyses wereerformed using GraphPad Prism5 (trial version 5.04, GraphPadoftware, Inc., La Jolla, CA). For each test, p value less than 0.05as considered significant. Data are expressed as mean ± standard

rror.

. Results and discussion

.1. Preparation and characterization of the lysate

The lysate obtained was a turbid extract and the total pro-ein concentration of lysate was 1.56 ± 0.5 mg/ml. The only proteinhich has been studied is the sperm protein (SP17, moleculareight 15 kD) which is expressed by ID8 cells [17]. It is one of

he cancer/testis antigens, a sub-class of TAAs. This protein haseen extensively studied and it is found to be expressed or over-xpressed in multiple myeloma and ovarian cancers. In a studyy Chiriva-Internati et al., this protein has been evaluated as anvarian cancer vaccine in murine models, where the mice weremmunized intramuscularly with SP17 in ten doses of vaccination.t has shown to be a promising approach prophylactically as wells therapeutically. Moreover, human SP17 shows 70% homologyith murine SP17 and is expressed in ovarian cancer. Thus, SP17ay be a promising antigen for immunotherapy in ovarian cancer

17]. Therefore, we performed western blot analysis for SP17 onhe lysate prepared. We could trace the presence of this proteins shown in Fig. 1(A). The presence of SP17 in the lysate ensuredhat the protein cocktail obtained was antigenic when given as aaccine.

.2. Preparation and characterization of vaccine microparticles

Please cite this article in press as: Tawde SA, et al. Formulation and evaluathttp://dx.doi.org/10.1016/j.vaccine.2012.05.073

One of the major challenges of oral vaccination is the avoidancef immune tolerance. However, this issue can be avoided by entrap-ing vaccine antigen into particles [29–31]. This can be due to the

ence of SP17 (15 kD) in the two different batches of lysates prepared. (B) Scanningelectronic microscopy photograph of microparticles obtained by spray drying tech-nology, showing crumbled, collapsed and irregular shaped microparticles.

efficient and enhanced uptake of particulate antigen by M cells in

the Peyer’s patches by phagocytosis, when compared to minimal

antigen uptake in solution form by pinocytosis. Moreover, protein

antigens taken up as particles are more potent in activating anti-

gen presenting cells (APCs) than soluble antigens [32–34]. Orally

delivered vaccines, especially particulate antigens are sampled by

M-cells in the Peyer’s patches of the intestine. Further these anti-

gens are processed by professional APCs such as dendritic cells and

macrophages that reside in the Peyer’s patches [35].

The morphological characterization of particles showed crum-

bled, collapsed and irregular shaped microparticles as shown in

Fig. 1(B). Eudragit® FS30D microparticles have shown to result in

such a shape upon spray drying by others as well [7]. The produc-

tion yield was 72.58 ± 3.41% (w/w). The particles obtained were of

1.58 ± 0.62 �m size with a charge of 12.48 ± 2.32 mV. There was

no significant change in size and charge upon loading these parti-cles with the lysate and interleukins. The loading efficiency of the

particles was found to be 92.68 ± 4.77% (w/w).

3.3. Immunization suppresses tumor growth

In case of mice treated with placebo particles, the tumor devel-

oped rapidly. However, vaccinated mice showed around six-fold

tumor suppression when compared to non-vaccinated/placebo

group. The tumor volume measurements obtained are shown

in Fig. 2. However, there was no significant difference between

vaccine and vaccine with interleukin groups in terms of tumor

volumes. This can be due to the interleukin concentration used in

vaccine which was not high enough to retard tumor growth more

than that seen with vaccine alone. Higher concentration of inter-

leukins might be needed to show additional tumor suppression.

3.4. Immunization generates humoral immune response and

orally administered interleukins influence Th1/Th2 pathway

ion of oral microparticulate ovarian cancer vaccines. Vaccine (2012),

In order to determine B-cell response, the serum samples 254

collected before each dose were analyzed by ELISA. As shown in 255

Fig. 3(A), at end of vaccination, immunized mice showed elevated 256

Page 4: Formulation and evaluation of oral microparticulate ... Formulation and evaluation o… · Eudragit ® FS 30 D was gen- ... Briefly, overnight coating of 159 the lysate (100 g/well)

ARTICLE ING Model

JVAC 13243 1–7

4 S.A. Tawde et al. / Vaccine

Fig. 2. Immunization with vaccine microparticles suppresses tumor growth: Meantumor volumes for mice groups treated orally with vaccine microparticles with andwithout interleukins along with data with mice treated with placebo micropar-ticles. The tumor volume was monitored with the aid of vernier calipers on awn

I257

t258

e259

p260

a261

v262

s263

o264

(265

266

267

268

269

270

271

272

273

274

275

276

277

278

279

280

281

282

283

284

285

286

287

cells of vaccine alone (p < 0.01). On the other hand, B-cell popula- 288

Ftwp((

eekly basis. Vaccinated mice showed higher tumor suppression as compared toon-vaccinated/placebo treated mice (p < 0.0001), ****p < 0.0001.

gG titers as compared to non-vaccinated ones (p < 0.05). Micereated with vaccine microparticles with interleukins showedven further elevated response as compared to mice receivinglacebo microparticles (p < 0.0001). When serum IgG1 titers werenalyzed, there was an elevation in titers in mice treated withaccine alone when compared to placebo group (p < 0.01) as

Please cite this article in press as: Tawde SA, et al. Formulation and evaluathttp://dx.doi.org/10.1016/j.vaccine.2012.05.073

hown in Fig. 3(B). The response was even further elevated in casef vaccine with interleukins group when compared to placebop < 0.001). In case of IgG2a titers, the titers were elevated in case

ig. 3. Immunization with vaccine microparticles generates humoral immune responseiters determined by ELISA, showing higher titers at the end of final vaccination in case ohen compared to mice treated with placebo microparticles. (B) Serum IgG1 titers detelacebo treated/non-vaccinated mice (p < 0.05). Vaccine alone showed elevated titers (p

p < 0.001). (C) Serum IgG2a titers determined by ELISA, showing higher titers in case of mp < 0.01), *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

PRESSxxx (2012) xxx– xxx

of vaccine alone when compared to placebo (p < 0.01) as shown

in Fig. 3(C). Thus, both subtypes of IgGs indicate that mixed Th1

and Th2 immune response was generated in case of vaccine alone

and Th2 type response was triggered in case of vaccine with

interleukin group. The reason behind the difference in the immune

response can be attributed to IL-12 effect in the Peyer’s patches

which has been reported to be a possible strategy for targeted

manipulation of immune responses to oral vaccines. Such response

was reported by Marinaro et al., when IL-12 was given orally to

mice orally immunized with tetanus toxoid and cholera toxin and

the effect was compared with intraperitoneal administration of

IL-12. They found that oral IL-12 resulted in the Th2 response

to oral vaccine, while intraperitoneal IL-12 caused Th1 response

[12].

3.5. Immunization generates T-cell and B-cell based immune

response

Lymphatic organs such as spleen and lymph nodes were ana-

lyzed for T-cell populations. The CD8+ as well as CD4+ T-cell

populations were found to be elevated in vaccinated mice when

compared to placebo group as shown in Fig. 4. The CD4+ T-cell

population was found to be elevated in case of spleen cells of vac-

cine with interleukin group (Fig. 4(C)) when compared to spleen

ion of oral microparticulate ovarian cancer vaccines. Vaccine (2012),

tion was determined in spleen, lymph nodes and bone marrow. In 289

case of B-cell population in spleen cells (Fig. 5(A)), there were ele- 290

vated levels in vaccine with interleukins group when compared to 291

and orally administered interleukins influence Th1/Th2 pathway: (A) Serum IgGf mice receiving vaccine orally with (p < 0.0001) and without interleukins (p < 0.05)rmined by ELISA, showing higher titers in case of vaccinated mice as compared to< 0.01) and vaccine with interleukins showed even further elevation in IgG1 titersice treated with vaccine alone as compared to placebo treated/non-vaccinated mice

Page 5: Formulation and evaluation of oral microparticulate ... Formulation and evaluation o… · Eudragit ® FS 30 D was gen- ... Briefly, overnight coating of 159 the lysate (100 g/well)

ARTICLE IN PRESSG Model

JVAC 13243 1–7

S.A. Tawde et al. / Vaccine xxx (2012) xxx– xxx 5

Fig. 4. Immunization with vaccine microparticles generates CD8+ and CD4+ T-cell based immune response: CD8+ T-cells population in (A) spleen, (B) lymph nodes, CD4+T-cell population in (C) spleen, (D) lymph nodes, (E) CD8+ T-cells population in spleen determined by flow cytometry, FL1-H being the count detected by filter FL1 forFITC-tagged CD8+ T-cells (shown in gate P5). The results are summarized in (A) indicating the elevated CD8+ T-cell population in vaccinated mice when compared to non-vaccinated mice (p < 0.05), (F) CD8+ T-cells population (gate P1) in lymph nodes determined by flow cytometry. The results are summarized in (B) indicating the elevatedCD8+ T-cell population in mice receiving vaccine alone (p < 0.001) and in mice receiving vaccine with interleukins (p < 0.01) when compared to non-vaccinated mice (G) CD4+T are suv whenn ng thet

p292

i293

c294

(295

TS

-cells population (gate P7) in spleens determined by flow cytometry. The results

accine alone (p < 0.05) and in mice receiving vaccine with interleukins (p < 0.001)

odes determined by flow cytometry. The results are summarized in (D) indicatireated/non-vaccinated mice (p < 0.001), *p < 0.05, **p < 0.01, ***p < 0.001.

Please cite this article in press as: Tawde SA, et al. Formulation and evaluathttp://dx.doi.org/10.1016/j.vaccine.2012.05.073

lacebo (p < 0.001) and vaccine alone (p < 0.01). B-cell populationsn lymph node cells as shown in Fig. 5(B) were found be elevated inase of vaccinated groups when compared to non-vaccinated micep < 0.001). B-cells in bone marrows (Fig. 5(C)) were found to be

able 1ummary of results obtained from in vivo study of ovarian cancer vaccine microparticles

In vivo study Organ/tissue analyzed

Tumor volume suppression Tumor

IgG titers at the end of vaccination Serum

IgG subtype IgG1 titers (Th2 type response) Serum

IgG subtype IgG2a titers (Th1 type response) Serum

B-cell population Bone marrow

Spleen

Lymph nodes

CD8+ T-cells Spleen

Lymph nodes

CD4+ T-cells Spleen

Lymph nodes

* p < 0.05, results expressed in comparison to placebo group.** p < 0.01 results expressed in comparison to placebo group.

*** p < 0.001 results expressed in comparison to placebo group.**** p < 0.0001, results expressed in comparison to placebo group.

a p < 0.05 results expressed for comparison between vaccine and vaccine with interleukaa p < 0.01 results expressed for comparison between vaccine and vaccine with interleuk

aaa p < 0.001, results expressed for comparison between vaccine and vaccine with interle

mmarized in (C) indicating the elevated CD4+ T-cell population in mice receiving compared to non-vaccinated mice (H) CD4+ T-cells population (gate P3) in lymph

elevated CD4+ T-cell population in vaccinated mice when compared to placebo

ion of oral microparticulate ovarian cancer vaccines. Vaccine (2012),

elevated in case of vaccine with interleukins group than placebo as 296

well as vaccine alone (p < 0.001). 297

The overall results obtained are summarized in Table 1. 298

When data was analyzed for vaccinated and non-vaccinated 299

when administered orally to mice.

Placebo Oral vaccine Oral vaccine + interleukins

– **** ****

– * ****

– ** ***

– ** ,a –

– – *** ,aaa

– – *** ,aa

– *** ***

– * *

– *** **

– * *** ,aa

– *** ***

ins groups.ins groups.

ukins groups.

Page 6: Formulation and evaluation of oral microparticulate ... Formulation and evaluation o… · Eudragit ® FS 30 D was gen- ... Briefly, overnight coating of 159 the lysate (100 g/well)

ARTICLE IN PRESSG Model

JVAC 13243 1–7

6 S.A. Tawde et al. / Vaccine xxx (2012) xxx– xxx

Fig. 5. Immunization with vaccine microparticles generates B-cell based immune response: (A) B-cells population in (A) spleen, (B) lymph nodes and (C) bone marrow, (D)B-cells population in spleen determined by flow cytometry, FL1-A being the count detected by filter FL1 for FITC-tagged B-cells (shown in gate P8). The results are summarizedin (A) indicating the elevated B-cell population in mice receiving vaccine with interleukin when compared to non-vaccinated mice (p < 0.001), (E) B-cells population in lymphn ) indiv inedp inated

m300

i301

T302

i303

e304

l305

n306

l307

a308

w309

m310

c311

i312

w313

o314

v315

c316

a317

r318

319

b320

l321

e322

t323

f324

c325

c326

327

328

329

330

331

332

333

334

335

336

337

338

339

340

341

342

odes (gate P23) determined by flow cytometry. The results are summarized in (Baccinated mice (p < 0.001). (F) B-cells population (gate P23) in bone marrow determopulation in mice receiving vaccine with interleukin when compared to non-vacc

ice, humoral response study showed higher antibody titersn vaccinated mice. Vaccine alone resulted in mixed Th1 andh2 response, while vaccine with interleukins showed predom-nant Th2 response. The B-cell population was found to belevated in bone marrow and spleen cells of vaccine with inter-eukins group than vaccine alone, while it was higher in lymphodes in case of vaccine alone as well as vaccine with inter-

eukins groups when compared to placebo. We found CD8+nd CD4+ T-cells were expanded in mice treated with vaccineith and without interleukins when compared to non-vaccinatedice. However, when the comparison was done between vac-

ine with and without interleukins groups, B-cell populationsn bone marrow and spleen along with CD4+ T-cells in spleen

ere found to be elevated in case of interleukins group. Theverall stimulation of humoral and cellular response uponaccination indicates the efficacy of oral vaccine microparti-les. Moreover, the immune stimulation in terms of humoralnd cellular response obtained correlated with tumor volumeetardation.

However, the anti-tumor effect was expected to be enhancedy interleukins, which was not observed in this study. The inter-

eukin concentration used might need to be increased to result innhanced anti-tumor activity. Another reason can be the activa-

Please cite this article in press as: Tawde SA, et al. Formulation and evaluathttp://dx.doi.org/10.1016/j.vaccine.2012.05.073

ion of regulatory T-cells (T-regs) by IL-2, which can be a demeritor vaccine efficacy. It has been reported that low-dose of IL-2an result in expansion of T-regs. The overall effect of T-helperells and NK-cells versus Tregs in response to IL-2 depends on

cating the elevated B-cell population in vaccinated mice when compared to non- by flow cytometry. The results are summarized in (C) indicating the elevated B-cell

mice (p < 0.001), **p < 0.01, ***p < 0.001.

IL-2 concentration, dosing time and duration of exposure, which

ultimately determines anti-tumor activity [13].

4. Conclusion

We have demonstrated the efficacy of vaccine microparticles

containing whole cell lysate of ID8 ovarian cancer cells in retarding

tumor growth in murine models. Thus, the oral microparticulate

vaccine described provides a promising approach in terms of cost-

effectiveness, ease of production and patient compatibility. This

study was performed in prophylactic setting to check the efficacy

of particles, which forms the basis for further studies to evaluate

therapeutic efficacy of the particles in tumor bearing animals.

Acknowledgements

Authors acknowledge Dr. Ray Green, Mercer University, Atlanta,

GA for his generous help with SDS-PAGE analysis, Dr. Chiriva Maur-

izio and Leonardo Mirandola, Texas Tech University Health Sciences

Center, TX for their kind guidance regarding Western blot analy-

ion of oral microparticulate ovarian cancer vaccines. Vaccine (2012),

sis and Dirk Anderson, BD Accuri Cytometers, MI for his generous 343

help with flow cytometry data analysis. A grant support from NIH 344

(R01CA138993 to P.S.) is gratefully acknowledged. 345

Conflict of interest: The authors report no conflict of interest. 346

Page 7: Formulation and evaluation of oral microparticulate ... Formulation and evaluation o… · Eudragit ® FS 30 D was gen- ... Briefly, overnight coating of 159 the lysate (100 g/well)

ING Model

J

ccine

R347

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

[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

[ 428

429

430

431

[ 432

433

434

435

[ 436

437

438

[ 439

440

441

[ 442

443

[ 444

445

446

1992;76(1):164–8. 447

ARTICLEVAC 13243 1–7

S.A. Tawde et al. / Va

eferences

[1] Berkowitz Z, Rim SH, Peipins LA. Characteristics, survival associated with ovar-ian cancer diagnosed as first cancer and ovarian cancer diagnosed subsequentto a previous cancer. Cancer Epidemiol 2011;35(2):112–9.

[2] Stuart GC. First-line treatment regimens and the role of consolidation therapyin advanced ovarian cancer. Gynecol Oncol 2003;90(3 (Pt 2)):S8–15.

[3] Leffers N, Daemen T, Boezen HM, Melief KJ, Nijman HW. Vaccine-based clinicaltrials in ovarian cancer. Expert Rev Vaccines 2011;10(6):775–84.

[4] Thibodeaux SR, Curiel TJ. Immune therapy for ovarian cancer: promise andpitfalls. Int Rev Immunol 2011;30(2–3):102–19.

[5] Pengetnze Y, Steed M, Roby KF, Terranova PF, Taylor CC. Src tyrosine kinasepromotes survival and resistance to chemotherapeutics in a mouse ovariancancer cell line. Biochem Biophys Res Commun 2003;309(2):377–83.

[6] Chiang CL, Benencia F, Coukos G. Whole tumor antigen vaccines. SeminImmunol 2010;22(3):132–43.

[7] Ano G, Esquisabel A, Pastor M, Talavera A, Cedre B, Fernandez S, et al. A neworal vaccine candidate based on the microencapsulation by spray-drying ofinactivated Vibrio cholerae. Vaccine 2011;29(34):5758–64.

[8] Chablani L, Tawde SA, D’Souza MJ. Spray-dried microparticles: a potential vehi-cle for oral delivery of vaccines. J Microencapsul, in press.

[9] D’Souza B, Bhowmik T, Shashidharamurthy R, Oettinger C, Selvaraj P, D’SouzaM. Oral microparticulate vaccine for melanoma using M-cell targeting. J DrugTarget 2012;20(2):166–73.

10] Akande J, Yeboah KG, Addo RT, Siddig A, Oettinger CW, D’Souza MJ. Targeteddelivery of antigens to the gut-associated lymphoid tissues: 2. Ex vivo evalua-tion of lectin-labelled albumin microspheres for targeted delivery of antigensto the M-cells of the Peyer’s patches. J Microencapsul 2010;27(4):325–36.

11] Babai I, Samira S, Barenholz Y, Zakay-Rones Z, Kedar E. A novel influenza subunitvaccine composed of liposome-encapsulated haemagglutinin/neuraminidaseand IL-2 or GM-CSF. II. Induction of TH1 and TH2 responses in mice. Vaccine1999;17(9–10):1239–50.

12] Marinaro M, Boyaka PN, Finkelman FD, Kiyono H, Jackson RJ, Jirillo E, et al.Oral but not parenteral interleukin (IL)-12 redirects T helper 2 (Th2)-typeresponses to an oral vaccine without altering mucosal IgA responses. J ExpMed 1997;185(3):415–27.

13] Huang X, Ye D, Thorpe PE. Enhancing the potency of a whole-cell breast cancervaccine in mice with an antibody-IL-2 immunocytokine that targets exposedphosphatidylserine. Vaccine 2011;29(29–30):4785–93.

14] Greenaway J, Moorehead R, Shaw P, Petrik J. Epithelial–stromal interactionincreases cell proliferation, survival and tumorigenicity in a mouse model ofhuman epithelial ovarian cancer. Gynecol Oncol 2008;108(2):385–94.

15] Hashemi SM, Hassan ZM, Soudi S, Shahabi S. The effect of vaccination with thelysate of heat-shocked tumor cells on nitric oxide production in BALB/c micewith fibrosarcoma tumor. Cell Biol Int 2008;32(7):835–40.

16] Danfeng X. Induction of protective antitumor activity of tumor lysate-pulseddendritic cells vaccine in RM-1 prostate cancer model. J Med Coll PLA2009;24(18–24):18–24.

17] Chiriva-Internati M, Yu Y, Mirandola L, Jenkins MR, Chapman C, Cannon

Please cite this article in press as: Tawde SA, et al. Formulation and evaluathttp://dx.doi.org/10.1016/j.vaccine.2012.05.073

M, et al. Cancer testis antigen vaccination affords long-term protec-tion in a murine model of ovarian cancer. PLoS One 2010;5(5):e10471,http://dx.doi.org/10.1371/journal.pone.0010471.

18] Lai YH, D’Souza MJ. Microparticle transport in the human intestinal M cellmodel. J Drug Target 2008;16(1):36–42.

[

[

PRESSxxx (2012) xxx– xxx 7

19] Lai YH, D’Souza MJ. Formulation and evaluation of an oral melanoma vaccine.

J Microencapsul 2007;24(3):235–52.

20] Akalkotkar A, Tawde SA, Chablani L, D’Souza MJ. Oral delivery of particulate

prostate cancer vaccine: in vitro and in vivo evaluation. J Drug Target, in press.

21] Yeboah KG, D’Souza MJ. Evaluation of albumin microspheres as oral

delivery system for Mycobacterium tuberculosis vaccines. J Microencapsul2009;26(2):166–79.

22] Gayakwad SG, Bejugam NK, Akhavein N, Uddin NA, Oettinger CE, D’Souza MJ.

Formulation and in vitro characterization of spray-dried antisense oligonu-

cleotide to NF-kappaB encapsulated albumin microspheres. J Microencapsul2009;26(8):692–700.

23] Uddin AN, Bejugam NK, Gayakwad SG, Akther P, D’Souza MJ. Oral deliv-

ery of gastro-resistant microencapsulated typhoid vaccine. J Drug Target

2009;17(7):553–60.

24] Bhowmik T, D’Souza B, Uddin MN, D’Souza MJ. Oral delivery of microparticles

containing plasmid DNA encoding hepatitis-B surface antigen. J Drug Target,in press.

25] Courreges MC, Benencia F, Conejo-Garcia JR, Zhang L, Coukos G. Prepara-

tion of apoptotic tumor cells with replication-incompetent HSV augments

the efficacy of dendritic cell vaccines. Cancer Gene Ther 2006;13(2):182–93.

26] Benencia F, Courreges MC, Conejo-Garcia JR, Mohammed-Hadley A,

Coukos G. Direct vaccination with tumor cells killed with ICP4-deficientHSVd120 elicits effective antitumor immunity. Cancer Biol Ther 2006;5(7):

867–74.

27] Sharma RK, Srivastava AK, Yolcu ES, MacLeod KJ, Schabowsky RH, Madireddi S,

et al. SA-4-1BBL as the immunomodulatory component of a HPV-16 E7 proteinbased vaccine shows robust therapeutic efficacy in a mouse cervical cancer

model. Vaccine 2010;28(36):5794–802.

28] Lubaroff DM, Karan D, Andrews MP, Acosta A, Abouassaly C, Sharma M, et al.

Decreased cytotoxic T cell activity generated by co-administration of PSA vac-cine and CpG ODN is associated with increased tumor protection in a mouse

model of prostate cancer. Vaccine 2006;24(35–36):6155–62.

29] Shen H, Ackerman AL, Cody V, Giodini A, Hinson ER, Cresswell P, et al.

Enhanced and prolonged cross-presentation following endosomal escape ofexogenous antigens encapsulated in biodegradable nanoparticles. Immunology

2006;117(1):78–88.

30] Mann JF, Acevedo R, Campo JD, Perez O, Ferro VA. Delivery systems: a

vaccine strategy for overcoming mucosal tolerance? Expert Rev Vaccines2009;8(1):103–12.

31] Primard C, Rochereau N, Luciani E, Genin C, Delair T, Paul S, et al. Traffic of

poly(lactic acid) nanoparticulate vaccine vehicle from intestinal mucus to sub-

epithelial immune competent cells. Biomaterials 2010;31(23):6060–8.

32] O’Hagan DT, Palin KJ, Davis SS. Poly(butyl-2-cyanoacrylate) particles as adju-

vants for oral immunization. Vaccine 1989;7(3):213–6.

33] Challacombe SJ, Rahman D, Jeffery H, Davis SS, O’Hagan DT. Enhanced

secretory IgA and systemic IgG antibody responses after oral immuniza-

tion with biodegradable microparticles containing antigen. Immunology

ion of oral microparticulate ovarian cancer vaccines. Vaccine (2012),

34] O’Hagan DT, Palin K, Davis SS, Artursson P, Sjoholm I. Microparticles as poten- 448

tially orally active immunological adjuvants. Vaccine 1989;7(5):421–4. 449

35] Malik B, Goyal AK, Mangal S, Zakir F, Vyas SP. Implication of gut immunology 450

in the design of oral vaccines. Curr Mol Med 2010;10(1):47–70. 451