Accepted Manuscript Title: Development and characterisation of alginate coated low molecular weight chitosan nanoparticles as new carriers for oral vaccine delivery in mice Author: Subrata Biswas Mainak Chattopadhyay Kalyan Kumar Sen Malay Kumar Saha PII: DOI: Reference:
S0144-8617(14)01240-5 http://dx.doi.org/doi:10.1016/j.carbpol.2014.12.044 CARP 9547
To appear in: Received date: Revised date: Accepted date:
31-1-2014 9-12-2014 10-12-2014
Please cite this article as: Biswas, S., Chattopadhyay, M., Sen, K. K., and Saha, M. K.,Development and characterisation of alginate coated low molecular weight chitosan nanoparticles as new carriers for oral vaccine delivery in mice, Carbohydrate Polymers (2015), http://dx.doi.org/10.1016/j.carbpol.2014.12.044 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
5 6 7 8 9
Alginate coating onto CS LMW nanoparticles protect antigen in acidic
ip t
4
106 kDa).
condition.
Degree of toxicity was not affected significantly due to difference of MW of
cr
3
Nanoparticles were prepared by using different low MW of CS (42, 74 and
CS.
Significant correlation was observed between the immune response with CS
us
2
Highlights
MW.
an
1
10 11
Ac ce p
te
d
M
12
1 Page 1 of 32
12 13 14
Development and characterisation of alginate coated low molecular weight chitosan nanoparticles as new carriers for oral vaccine delivery in mice
15 Subrata Biswas a, Mainak Chattopadhyay b, Kalyan Kumar Sen c,*, Malay Kumar
17
Saha a
ip t
16 18 19
a
20 21
Research, Kolkata– 700010, India b Department of Pharmacology, Bengal School of Technology, Hooghly-712102, West
cr
National Institute of Cholera and Enteric Diseases, Indian Council of Medical
Bengal, India.
23
c
24
More, G.T. Road, Asansol-713301, West Bengal, India.
us
22
an
Department of Pharmaceutics, Gupta College of Technological Sciences, Ashram
25 26 Email addresses:
SB-
[email protected]
M
27
MC-
[email protected]
29
KKS-
[email protected]
30
MKS-
[email protected]
d
28
te
31 32 34 35 36 37 38 39 40 41 42 43 44 45 46
Ac ce p
33
*Corresponding author
Dr. Kalyan Kumar Sen Principal & Professor Gupta College of Technological Sciences Ashram More, G T Road, Asansol - 713301 West Bengal, India E-mail:
[email protected] Telephone No.: +91-341- 2312130 Fax No.: +91-341- 2314604
47 48 2 Page 2 of 32
ABSTRACT
50
In the present study, nanoparticles of low MW chitosan (CS) were formulated in
51
which measles antigen was entrapped and subsequently coated with sodium
52
alginate. The size and surface properties of the nanoparticle can be tuned with
53
different MW of CS. In vitro release studies showed initial burst release followed by
54
extended release, best fitted in the Makoid-Banakar model (R2 > 0.98). SDS-PAGE
55
assay revealed that alginate coating could effectively protect antigen in acidic
56
condition for at least 2 h. Cell viability was assessed using MTT assay into HT 29 cell
57
line. Formulations were orally administered to mice and immunological responses
58
were evaluated using ELISA method. Obtained results showed that measles antigen-
59
loaded CS nanoparticles induced strong immune response and significant correlation
60
was observed between the immune response with CS MW. Protecting ability of
61
antigen in gastric environment, sustained release kinetics, systemic and mucosal
62
immune responses and low cytotoxicity observed for the alginate coated
63
nanoparticles demonstrated that LMW CS could be promising platform for oral
64
vaccine delivery.
M
an
us
cr
ip t
49
d
65
Keywords: adjuvant, cytotoxicity, mucosal immunity, vaccine
67
Abbreviations: PAGE- polyacrylamide gel electrophoresis, GALT-gut associated
68
lymphoid tissue
70 71 72 73 74
Ac ce p
69
te
66
75 76 77 78 79 80 81 3 Page 3 of 32
82 83
1.
Introduction
84 It is well established that protection against pathogenic organisms occurs better with
86
the presence of mucosal antibody than with serum antibody (Baumann, 2008).
87
Compared to traditional routes of administration, oral vaccine delivery systems offer
88
several attractive advantages such as lower cost, ease of administration, higher
89
patient compliance, reducing the need for trained personnel, averting vaccine-related
90
infections correlated to the disposal and reuse of needles in systemic delivery as well
91
as higher capacity of much for mass immunizations (Fooks, 2004; Kim, Lee, & Jang,
92
2012; Kostrzak et al. 2009; Zhu, & Berzofsky, 2013). It has also been demonstrated
93
that orally administered antigens can induce both local and systemic immune
94
responses, providing a complete immune response (Neutra, & Kozlowski, 2006).
95
However, immune responses following oral administration of vaccines are usually
96
poor due to rapid degradation of antigen in the harsh gut environment and their poor
97
uptake by appropriate target sites, namely M cells located in the Peyer’s patches
98
(Czerkinsky, & Holmgren, 2009). Keeping this in mind, several studies have been
99
performed to show that by associating the vaccine with a number of particulate
d
M
an
us
cr
ip t
85
delivery systems, the degradation of the antigen in the harsh environment of the GI
101
tract is prevented and the uptake by M-cells is enhanced by several times (Delie, &
102
Blanco-Prieto, 2005; Eyles, Sharp, Williamson, Spiers, & Alpar, 1998; van der
103
Merwe, Verhoef, Verheijden, Kotze, & Junginger, 2004). In addition, biodegradable
104
particles allow a sustained release of the antigen, increasing the duration of the
105
contact between antigen and immune cells thus favouring an effective immune
106
response.
Ac ce p
107
te
100
In the past decades, chitosan (CS) has been extensively used as a carrier
108
candidate because of its excellent stability, mucoadhesive property, enhanced
109
penetration capacity across the mucosal barriers and good compatibility with vaccine
110
antigen (Borges et al., 2007; Lee et al., 2011; Yoo et al., 2010; Zaharoff, Rogers,
111
Hance, Schlom, & Greiner, 2007). The formation of CS nanoparticles is a process
112
based on the complexation of oppositely charged molecules by electrostatic
113
interactions (Csaba, Koping-Hoggard, & Alonso, 1997). Nanoparticles can be easily
114
constructed using an ionic gelation process that involves sodium tripolyphosphate as
115
a negatively charged molecule and CS as polycation (Calvo, Remunan-Lopez, Vila4 Page 4 of 32
Jato, & Alonso, 1997; Oliveria et al, 2012). The mild technique involves the mixing of
117
two aqueous solutions at ambient temperature while stirring without using sonication
118
or organic solvents. Proteins with low isoelectric point, such as BSA and measles
119
antigen, were better associated with the nanoparticles when dissolved in the alkaline
120
sodium TPP solution (Li et al., 2008). Although CS nanoparticles have numerous
121
advantages as delivery carriers for oral vaccination, their limited ability to control the
122
release of encapsulated antigens and easy solubility in acidic medium largely
123
hamper their development (Li et al., 2008). These obstacles may be overcome by
124
coating those antigen loaded particles with an acid-resistant polymer, like sodium
125
alginate (Borges et al., 2006).
us
cr
ip t
116
Sodium alginate, an anionic polysaccharide with favourable biological
127
properties, can be easily interacted with cationic CS nanoparticles to form an
128
electrolyte complex via electrostatic interactions (Du et al., 2013; Kim et al., 2002).
129
Alginate-CS polyionic complexes form through ionic gelation via interactions
130
between the cationic amine groups of CS and the anionic carboxyl groups of alginate
131
(Gazori et al., 2009). This coating procedure was performed at relatively mild
132
conditions without using any organic solvent at room temperature, enabling antigens
133
to be incorporated into the CS/alginate matrices with retention of biological activity
134
(Wee, & Gombotz, 1998). Calcium ion is used as crosslinking agent to strengthen
135
and stabilize the particles (Borges et al., 2006). Recently, Liu et al. (2013)
136
demonstrated that DNA loaded in alginate coated CS nanoparticles alginate-coated
137
CS nanoparticles containing DNA are effectively taken up and expressed by the
138
Payer’s patches. Release profiles of vaccines from these delivery systems showed
139
that burst release of loaded BSA in pH 1.2 (simulated gastric fluid) was largely
140
prevented by its entrapment in alginate-coated CS particles (Anal, Bhopatkar,
141
Tokura, Tamura, & Stevens, 2003). Moreover, Borges et al. (2006) demonstrated
142
that sodium-alginate coated nanoparticles were readily able to be taken up by rat
143
Peyer’s patches which is one of the essential features to internalize, deliver and
144
target the intact antigen to specialized immune cells from gut associated lymphoid
145
tissue (GALT). In vivo studies have shown higher antibody titre for alginate coated
146
CS nanoparticles compared to plain CS nanoparticles (Malik, Goyal, Zakir, & Vyas,
147
2011). This delivery system also acted as an effective adjuvant for hepatitis B
148
surface antigen when subcutaneously administered in a mouse model (Borges et al.,
149
2008). It has been demonstrated that transfection efficiency is closely related to the
Ac ce p
te
d
M
an
126
5 Page 5 of 32
150
MW of polymer. CS of 10 -150 kDa with a specific degree of deacetylation allows
151
maximum transgenic expression in vitro and CS nanoparticles less than 500 nm in
152
diameter are transported through an endocytotic mechanism (Godbey, Wu, & Mikos,
153
1999; Lavertu, Methot, Tran-Khanh, & Buschmann, 2006; Liu et al., 2013). Recently, there has been growing interest in low molecular weight CS (CS-
ip t
154
LMW) as a new mucosal delivery vehicle due to its greater solubility in neutral
156
aqueous solvents with low viscosity and faster degradation than higher molecular
157
weight CS counterparts (Amoozgar, Park, Lin, & Yeo, 2012; Fernandes et al., 2012;
158
Liu et al., 2013). Vila et al. (2004) reported that nanoparticles made of CS-LMW with
159
tetanus toxoid (TT) as a model protein could be produced by an ionic-cross linking
160
technique and proving to be promising carrier for nasal vaccine delivery. Following
161
intranasal administration, TT-loaded nanoparticles elicited an increasing and long-
162
lasting humoral response as compared to the soluble antigen. Plasma or serum
163
samples have traditionally been used for generation of serological data due to the
164
difficulties associated with handling whole blood. However, this approach requires
165
relatively large volumes (200 - 1000 µl) of blood in order to produce the required
166
volume of matrix for bioanalysis, which makes it difficult to generate serial profiles in
167
mice test models. Composite profiles are therefore necessary, which can result in
168
lower quality data and requires the use of a greater number of animals. Conventional
169
ELISA are formatted to be used only for determining whether a given sample
170
contains antibody at a concentration above or below a set threshold value, although
171
quantification of antibody response is prerequisite to monitor the progress after
172
immunization. In recent years, dried blood spot (DBS) assays have received
173
increasing attention as an alternative to venous blood sampling and can be adopted
174
in preclinical serological studies.
us
an
M
d
te
Ac ce p
175
cr
155
To the best of our knowledge, there is no published data whereby alginate
176
coated CS-LMW nanoparticles are explored as vehicles for the oral administration of
177
vaccine. In this study, we have tried to develop a novel oral vaccine carrier based on
178
alginate coated CS-LMW nanoparticles to meet the requirement of oral vaccine. So
179
the objectives of the studies are to develop the nanoparticle based oral vaccine
180
delivery system and the delivery system will be adopted to evaluate the
181
physicochemical properties, loading efficiency of the particles and the stability of
182
antigen loaded nanoparticles against acidic condition. Cytotoxicity will be evaluated
183
using HT 29 human epithelial cell lines for the different formulation of nanoparticles. 6 Page 6 of 32
184
Finally, the effects of molecular weight of CS (42, 74 and 106 kDa) on the ability of
185
these nanoparticles will be studied in a mouse model to elicit significant immune
186
response as measured using DBS ELISA method.
188
2.
Experimental
2.1.
Materials
ip t
187
189 190
CS-LMW of three different molecular weight (42, 74 and 106 kDa) were
192
obtained through oxidation-reduction reaction of high molecular weight CS (MW 375
193
kDa) (Sigma Aldrich, USA) using NaNO2 as described by Mao et al. (2004). Briefly,
194
varying quantities of 0.1 M NaNO2 were added to the CS solution with controlled
195
temperature at 25 ºC under magnetic stirring, and the reaction was left overnight to
196
assure completion of the degradation. The depolymerisation process of CS has been
197
previously simulated based on Box-Benkhen design experiments. With this
198
approach, the required amount of NaNO2 could be predicted in order to obtain
199
approximate molecular weight of 42, 76 and 106 kDa. The molecular weight of the
200
CS was determined using High Pressure Size Exclusion Chromatography (HPSEC).
us
an
M
Sodium tripolyphosphate (TPP), sodium alginate, CaCl2, DMEM (Dulbecco’s
d
201
cr
191
Modified Eagle’s Medium), HEPES [N-(2-hydroxyethyl piperazine-N-(2-
203
ethanosulphonic acid)], MTT (methylthiazolyl diphenyl-tetrazolium bromide)
204
reagents, goat anti-mouse IgG peroxidise conjugate and TMB/H2O2 substrate for
205
ELISA were procured from Sigma (USA). Anti measles monoclonal antibody was
206
purchased from Millipore. BCA kit was purchased from Pierce (USA). Measles
207
antigen was kindly donated by Serum Institute of India Ltd (Pune, India). 96-well flat
208
bottom polystyrene plate was purchased from Nunc (Denmark). All other reagents
209
were of analytical grade. Ultrapure water (MilliQ, Waters, USA) was used to prepare
210
all solutions and freshly prepared solutions were used in all experiments.
211
Ac ce p
te
202
Human intestinal epithelial cells (HT 29) were cultured and maintained in
212
DMEM with supplemented with 10% fetal bovine serum and 1% antibiotic-
213
antimycotic solution (Invitrogen, Carlsbad, CA). Cells were maintained in 5% CO2 at
214
37 ºC humidified incubator for 10 days.
215
Immunogenicity of the antigen was evaluated in 6 weeks old specific
216
pathogen-free male BALB/c mice. Animal care and handling were maintained
217
according to the guidelines established by the Institutional Animal Ethical Committee 7 Page 7 of 32
218
(IAEC) of Gupta College of Technological Sciences, India. The experiment protocols
219
were approved by IAEC and the experimental procedures were in accordance with
220
IAEC.
221 2.2.
Preparation of CS nanoparticles
ip t
222
CS nanoparticles were prepared by an ionotropic gelation process technique
224
using TPP as a crosslinking agent under stirring (Li et al., 2008). Briefly, 0.2% (w/v)
225
CS solution was prepared in 25 mM sodium acetate buffer (pH 5.2) and TPP was
226
dissolved in ultra-pure water in order to obtain 0.84 mg/ml solutions. Nanoparticles
227
were formed when 10 ml TPP solution was added drop wise to a 10 ml CS solution
228
and magnetically stirred at 300 rpm during 30 min. For association of measles
229
antigen with CS nanoparticle, 600 µg of measles antigen was incorporated in the
230
TPP solution. Cryoprotectant mannitol (1:1) was added to the resulting nanoparticle
231
suspension, frozen at -70 ºC for 48 h, and then lyophilized in a freeze dryer (FDU-
232
S603, Operon, Korea). Freeze-dried CS nanoparticles were redispersed in ultrapure
233
water for further use.
M
an
us
cr
223
234 2.3.
Antigen loaded CS nanoparticle suspension (20 ml, pH 5.1) was added drop-
te
236
Preparation of alginate coated CS nanoparticles
d
235
wise to sodium alginate solution (20 ml, pH 7.0) at concentration of 1 mg/ml under
238
mild stirring for 30 min at room temperature. Alginate coated CS nanoparticles were
239
redispersed into aqueous solution of CaCl2 (pH 7.0, 0.5 mmol/l) to crosslink the
240
alginate layer on the surface of antigen loaded CS nanoparticles. Cryoprotectant
241
mannitol (1:1) was added to the resulting suspension and stored at -70 ºC for 48 h
242
before freeze-drying.
243 244 245
Ac ce p
237
2.4.
Loading capacity of antigen in coated nanoparticles
Loading capacity (LC) of measles antigen loaded CS nanoparticles were was
246
calculated in an indirect way by determining the measles antigen remaining in the
247
aqueous phase using the BCA kit (Borges et al., 2006). Aliquots of the resulting
248
nanoparticle suspension were centrifuged for 20 min at 18,000 X g and the
249
supernatants were then separated from the nanoparticles. The supernatant of blank
250
CS nanoparticles was adopted as the blank to correct the absorbance reading value
8 Page 8 of 32
251
of the measles antigen loaded nanoparticles. The LC values of antigen loaded
252
nanoparticles were calculated from the following equations:
253
255 256
2.5.
The morphological characteristics of nanoparticles were examined by
cr
257
Morphological characterization, size and surface charge
ip t
254
scanning electron microscopy (JEOL JSM 5200, Japan). The particle size
259
distribution was measured by laser diffraction and zeta potential of the particle was
260
examined by zeta analyser using Zetasizer (Ver. 6.00, Malvern Instruments Ltd, UK)
261
with ultrapure water as solvent (pH 7, 25 OC). These measurements were run at
262
least three times (replicates) and were presented as mean ± SD with independent
263
particle batches.
an
us
258
266
2.6.
In vitro release studies
Approximately 20 mg dried nanoparticle samples (CS or alginate-coated CS )
d
265
M
264
CS nanoparticles and alginate coated CS nanoparticles were incubated in 3 ml of
268
phosphate buffered saline (PBS, pH 7.4) in an Eppendorf tube, and then placed in
269
an air shaker at 100 rpm at 37 OC. At predetermined time intervals, the samples
270
were centrifuged at 13,200 rpm for 20 min and the supernatant was replaced with
271
fresh release medium to reach the original volume and resuspended. The amount of
272
antigen released was determined by BCA reagents as described in section 2.4 2.3.
273
According to protocol, the amount of antigen released was expressed as percentage
274
of total antigen encapsulated in CS nanoparticles as calculated from the LC value. A
275
sample consisting of only non-loaded nanoparticles re-suspended in PBS was used
276
as a blank. In vitro release experiments were repeated three times.
Ac ce p
te
267
277 278 279
2.7. Stability study in simulated gastric fluid Different formulations of CS nanoparticles (antigen loaded CS nanoparticles
280
and alginate coated antigen loaded nanoparticles) were incubated with 0.5 ml of HCl
281
(0.01 M) in a shaker bath at 37 ºC for 2 h. Then, acid was neutralized by 0.5 ml of
282
aqueous NaOH (0.01 M) solution. These mixtures were allowed to stand for another
9 Page 9 of 32
24 h, then diluted with PBS (pH 7.4) to a final volume of 4 ml. Twenty four hours
284
later, supernatant containing released antigen was collected and analyzed by SDS-
285
polyacrylamide gel electrophoresis (SDS-PAGE) in slab gel systems employed a
286
7.5% running gel and a 5.0% stacking gel. The protein sample was prepared in
287
electrophoresis loading-buffer (60 mM Tris-HCl, pH 6.8, with 25% glycerol and 2%
288
SDS containing 0.1% Bromophenol blue solution, and h-mercaptoethanol 2-
289
mercaptoethanol) and heated for 5 min at 95 ºC. The pure antigen was used as
290
control. After electrophoresis, the protein bands were visualized by staining with
291
Coomassie Blue.
cr
ip t
283
293
2.8.
In vitro cytotoxicity assay
To investigate the potential cytotoxicity of uncoated and alginate coated CS-
an
294
us
292
antigen nanoparticles, the cell viability was analyzed using HT 29 cell lines in a MTT
296
assay. The HT 29 human epithelial cell line was selected in the present study as a
297
convenient in vitro model to assess polymer toxicity (Sergent, Paget, & Chevillard,
298
2012). The assay was performed according to the method described by Mosmann
299
(1983). HT 29 cells were seeded into 96-well microplates at a density of 3000
300
cells/cm2. The medium was removed 24 h after plating and the cells were incubated
301
for 6 h with 50 µl nanoparticle in HBSS (Nanoparticle concentrations were 0.25, 1.0
302
and 4.0 mg/ml). SDS (10mg/ml) was used as positive control and HBSS as
303
reference for 100% cell viability. After incubation for 6 h the medium was discarded,
304
the cells were washed twice with phosphate-buffered saline and 50 µl of 5 mg/ml
305
MTT solution in PBS were added to each well. The plates were incubated in a
306
humidified atmosphere of 5% CO2 at 37 ºC for another 6 h. Subsequently, the wells
307
were emptied, and the formazan crystals were dissolved by adding 100 µl dimethyl
308
sulfoxide (DMSO) to each well. The absorbance was measured in triplicate at 570
309
nm, with a background correction at 630 nm, using a microplate ELISA reader
310
(Multiscan Ex, Thermo Scientific). The MTT assay was repeated three times.
Ac ce p
te
d
M
295
311 312 313
2.9.
Immunization studies The immunogenicity of the antigen was assessed in BALB/c mice following
314
oral immunization. Mice were housed in groups of six (n = 6) animals for one week
315
before the experiments were conducted. Mice were immunized orally with various
316
formulations (Table 1) containing 20 µg of antigen (calculated from loading capacity 10 Page 10 of 32
of the nanoparticles) in 50 µl of saline solutions on days 0, 7, 14 and 28 via a 25-
318
guage 1ml hypodermic needle (Song et al., 2005). Subcutaneous immunization was
319
also carried out with measles vaccine to serve as positive control. One group of mice
320
was treated with alginate coated blank bead beads to serve as negative control and
321
measles antigen for another group was administered to another control group. All
322
animals were conscious during the administration.
ip t
317
323 Table 1
cr
324 325
2.10. Sampling of body fluids
us
326
Blood samples were drawn from the tail-vein on day 7, 14, 28, 56 post
328
administration using a lancet (Hoff, 2000). Three drops of blood were collected in the
329
protein-savour dry blood spot (DBS) card (Biorad, USA). Blood spots were allowed
330
to dry overnight at room temperature. To obtain intestinal lavage fluids, on day 28
331
and 56 three mice were anesthetized i.p. with pentobarbital, then necropsied.
332
Lengths of upper small intestine from each mouse were sectioned longitudinally, and
333
the Intestinal lavage fluids were obtained by washing the intestine three times with
334
0.5 ml of ice-cold saline containing the protease inhibitor. DBS card was kept in a
335
zip-lock bag at room temperature with desiccant and gastric lavage was stored at -
336
20 ºC until analyzed.
338 339
M
d
te
Ac ce p
337
an
327
2.11. ELISA
IgG antibody levels in each sample were determined using a DBS ELISA test.
340
Discs of approximately 3 mm diameter were punched from DBS cards, individual
341
paper discs were transferred to individual wells of a 96-well titre plates, and analytes
342
were eluted with PBS containing 0.05% (w/v) Tween 20 (PBST) buffer. First,
343
measles antigen (4 µg/ml) in carbonate buffer (pH 9.6) was added to microplates and
344
incubated at 4 ºC in a humid container. The wells were washed three times with
345
PBST. To minimize non-specific interactions, 100 µl of PBST containing 2.5% (w/v)
346
of dried skimmed milk powder was added to the wells and incubated for 1 h at 37 ºC
347
in a humid container and the plate were washed three times with PBST. Eluted
348
samples were serially diluted 5-fold starting at either a 1:10 or 1:100 dilution and
349
incubated 2 h with a known concentration of antigen, then were added to the wells
350
and the plates were incubated for 2 h at 37 ºC in a humid container and washed. 11 Page 11 of 32
Then, 100 µl of anti-mouse IgG peroxidase conjugate, diluted 1:2000 in PBSTM
352
buffer, was added into each well and plates were incubated for another 1 h at 37 ºC.
353
The plates were washed, and 50 µl of substrate was added to each well, and the
354
reaction was allowed to keep proceed in the dark for 15 min at RT room
355
temperature. 100 µl of stopping solution (1 N sulphuric acid) was added to each well
356
and plates were read at 450 nm on a microplate reader.
357
ip t
351
Intestinal lavages were sonicated on ice to extract immunoglobulins from
mucin and centrifuged (16,000 X g, 60 min, 4 ºC). Supernatants were diluted with
359
200 µl of PBS for ELISA assay of specific IgA content as described above for IgG.
360
Titre was derived as the maximum dilution of intestinal lavages giving an OD of 0.2
361
after correction for background. Reciprocal dilutions corresponding to endpoint titres
362
were determined and were presented as mean log10titre ± SEM per group.
an
us
cr
358
363 364
2.12. Statistical analysis
If another method is not explicitly stated, the data were expressed as mean
M
365
values ± standard deviation (SD). The effect of the CS molecular weight on the cell
367
viability and adjuvaniticity adjuvant capacity was assessed with the aid of SPSS 16.0
368
(SPSS Inc., Chicago, USA) using one way ANOVA with the significance level set at
369
p < 0.05. Modeling and comparison of dissolution profile and release kinetics were
370
evaluated using DDSolver.
371 372
Ac ce p
te
d
366
373
3.
374
3.1.
375
Results and discussion Formulation and characterization of CS LMW nanoparticles To achieve the nanoparticle formulation for measles antigen with CS LMW,
376
systematic preformulation studies were carried out and the composition of this
377
formulation was chosen as CS/TPP ratio of 2:0.84 and CS/sodium alginate ratio 2:1.
378
Antigen loaded nanoparticles were systematically characterized to evaluate
379
morphology, drug loading and releasing efficiency. The study of scanning electron
380
microscopic images revealed that antigen-loaded alginate-coated CS nanoparticles
381
have irregular shape with slight aggregation (Fig. 1). The particle sizes and zeta
382
potentials of antigen loaded alginate uncoated and coated CS nanoparticles of three
383
different MW are shown in Table 2. Decreasing the MW of CS resulted in a decrease
384
in nanoparticle size and increasing loading efficiency (Lee, Lee, & Jon, 2010). 12 Page 12 of 32
However, very low MW CS may have poor loading efficiency and may not form
386
stable nanoparticles (Fernandes et al., 2012). Vaccine carried with alginate coated
387
CS nanoparticles exhibits similar trends in increasing particle size compared with CS
388
nanoparticles in all three cases. The zeta potential of CS nanoparticles is an
389
important factor for the stability and mucoadhesiveness of nanoparticles. Generally
390
the higher zeta potential of particles led to more stability, whereas the lower zeta
391
potentials of the particles induce agglomeration (Gan, Wang, Cochrane, &
392
McCarron, 2005). CS molecular weight effect on encapsulation is largely the
393
consequence of how the cationic CS chain interacts with protein molecules which
394
have a more elaborate and complex 3-D structure as polypeptide chains contain
395
both positive and negatively charged functional groups, and also hydrophobic
396
regions which tend to fold inside the 3-D structure in an aqueous environment (Gan,
397
Wang, Cochrane, & McCarron, 2005). However, encapsulation efficiency of a
398
protein, tetanus toxoid, was high, irrespective of the CS molecular weight and
399
indicated that the entanglement of the protein within the CS chains is not affected by
400
their molecular weight within the range studied (Vila et al., 2004).
M
an
us
cr
ip t
385
Table 2
403
Fig. 1.
405 406
3.2
In vitro release studies
Ac ce p
404
te
402
d
401
The release profiles of antigen from uncoated and alginate coated CS
407
nanoparticles in phosphate buffer are shown in Fig. 2. CS nanoparticle formulations
408
exhibited two release phases, a rapid release over the first 3 h followed by a slow
409
release for up to 120 h. The burst release might be attributed to the fact that antigen
410
was loosely bound onto CS nanoparticles by ionic interactions which could be easily
411
desorbed in an ionic environment (Chen, Zhang, & Huang, 2007). The second phase
412
might correspond to those antigen molecules more efficiently entrapped and tightly
413
bound to the CS molecules. Antigen release was relatively rapid for the first 3 h in
414
case of lower MW of CS CS MW, which can be attributed to an easier detachment of
415
low MW CS molecules and antigen molecules located at the surface of the particles.
416
However, alginate coated CS nanoparticles could increase the stability of CS
417
nanoparticles in the PBS, resulting in extended release of antigen. The dissolution
418
data were plotted according to different models and drug release profiles of 13 Page 13 of 32
formulations F2 - F6 were compared with that of reference product (F1) to evaluate
420
the effect of process using the similarity factor (f2). The curvilinear nature of the
421
cumulative percent antigen released versus time plots depicts that the release of
422
antigen from the nanoparticles does not follow zero-order or first order kinetics
423
(Table 3). The in vitro dissolution studies suggest that drug release was governed by
424
Higuchi kinetics. However, the mathematical expression that best describes the
425
antigen release from these nanoparticles is the Makoid–Banakar model in which the
426
resultant R2 values were greater than 0.98. The Korsmeyer–Peppas release
427
exponent, n, is approximately 0.3, confirming that the release of antigen is governed
428
by a diffusion process. So it can be confirmed that antigen release is controlled by a
429
Fickian diffusion process that will also result in sustained release of antigen in blood.
430
Antigen release from nanoparticles was found to be affected by antigen-polymer
431
interaction, decreasing with higher MW of CS (Costa, & Sousa Lobo, 2003).
an
us
cr
ip t
419
432 Table 3
M
433 434
Fig. 2.
436
3.3.
Antigen released from uncoated and alginate coated CS nanoparticles in an
te
437
Stability study in simulated gastric fluid
d
435
acidic medium was analyzed by SDS-PAGE followed by Coomassie brilliant blue
439
staining. First The first prerequisite for effective oral vaccine formulation is to protect
440
antigen from the harsh conditions in the stomach after oral administration delivery.
441
According to Fig. 3, molecular weight marker was shown in Lane 6, and the measles
442
antigen incubated with PBS (pH 7.4) for 24 h exhibited three clear bands at about
443
80, 60 and 45 kDa (Lane 5). However, measles antigen pretreated with 0.01 M HCl
444
for 2 h had a faint smear (Lane 4) which indicated that the antigen underwent serious
445
degradation in an acidic medium.
446
Ac ce p
438
The SDS-PAGE data clearly indicates uncoated CS nanoparticles containing
447
measles antigen were degraded in the presence of acid. However, antigen from
448
alginate coated CS nanoparticles had clear bands at about 80, 60 and 45 kDa in
449
both the lanes (Lane 1 and Lane 2), which implied that alginate coating of CS
450
nanoparticles could effectively protect antigen from degradation in an acidic medium
451
for at least 2 h. So, the obtained alginate coated CS nanoparticles might be an
14 Page 14 of 32
452
effective oral antigenic carrier for mucosal vaccine. Keeping this in mind,
453
immunological study was performed only with alginate coated CS nanoparticles.
454 455
Fig. 3.
457
3.4.
458
ip t
456 In vitro cell viability studies
The MTT test results indicated that both alginate-coated (% cell viability 86.43 ± 4.60) and uncoated (% cell viability 82.58 ± 2.80) of comparatively higher MW of
460
CS (MW 106) nanoparticles of higher MW CS (MW 106) had minimal effects on HT-
461
29 cells even at higher concentration of 5 4 mg/ml. No significant difference in
462
cytotoxicity was observed among nanoparticles formulated with three different MW
463
(Fig. 4). These results are in good agreement with other studies where MW of CS did
464
not interfere with cell viability (Huang, Khor, & Lim, 2004; Opanasopit et al., 2007;
465
Richardson, Kolbe, & Duncan, 1999). Cytotoxicity was observed only for higher
466
concentration (5 4 mg/ml) for the formulation with CS of MW 42 kDa. Interestingly,
467
alginate coating increases cell viability, indicating that the cytotoxicity is related to the
468
charge density of the particle; toxicity increases with increasing nanoparticle positive
469
charge density (Yang, He, Duan, Kui, & Li, 2007).
470
472 473 474
Fig. 4.
Ac ce p
471
te
d
M
an
us
cr
459
3.5.
Immunological response
In the present study, we investigated the immunological response after the
475
administration of the measles antigen vaccine entrapped in alginate-coated CS LMW
476
nanoparticles was investigated. The humoral immune response was accompanied
477
by the presence of measles specific IgG and IgA in the serum and on the intestinal
478
mucosa, respectively. Sample collection for a preclinical DBS assay involves
479
spotting a small volume of blood (10-20 µl per spot) onto the special protein saver
480
card and drying subsequently to prepare DBS specimens. These cards consist of a
481
specialized matrix that lyses cells on contact, denatures proteins, and inactivates
482
bacteria and viruses. When required, a disc of 3 mm diameter is punched from this
483
card and the analytes are isolated by an elution buffer. A sensitive limiting dilution
484
analysis, comprising competitive and indirect ELISA, is carried out to quantitate the 15 Page 15 of 32
485
systemic humoral immune response and monitor the progress of antibody responses
486
in different groups of mice immunized with vaccine preparations.
487
The minimum dose of measles antigen dose (equivalent to 20 µg) was selected for the immunization study, calculated from the loading capacity of each
489
nanoparticle formulation, aimed at elucidating whether or not the MW of CS affects
490
systemic and mucosal responses. The protective anti-measles IgG titre value and
491
anti-measles IgA titre value results were found measured and results are plotted in
492
Fig. 5 and Fig. 6.
cr
ip t
488
493 3.5.1. Systemic antibody response
495
us
494
Mice were immunized orally with various formulations (Table 1) on days 0, 7, 14 and 28 and blood samples were drawn from the tail-vein on days 7, 14, 28, 56
497
post administration using a lancet. All the samples were tested in the same day with
498
same batch of reagents to minimize imprecision and bias. IgG titres were measured
499
in responder mice. The association of the measles antigen (group I-III) with the
500
alginate coated CS nanoparticles induced a strong humoral immune response that
501
was significantly higher (p < 0.05) than the mean titre found for group VI, vaccinated
502
with measles antigen orally (Fig. 5). When comparing statistically, the immunological
503
response elicited at each individual time point, the IgG levels for lower MW were
504
always significantly higher than those corresponding to the higher MW. With only
505
one boost, all the groups orally vaccinated with the antigen associated with coated
506
nanoparticles were able to show seroconversion. The formulations were found to
507
elicit responses that increased for 14 days and then levelled off. In fact, the group
508
vaccinated with the uncoated measles vaccine given orally showed a very low
509
responder number, whereas the group, with the measles antigen given
510
subcutaneously showed higher log antibody titre.
512
M
d
te
Ac ce p
511
an
496
Fig. 5.
513 514 515
3.5.2.
Mucosal anti-measles sIgA Mice were immunized orally with various formulations (Table 1) on days 0, 7,
516
14 and 28 and intestinal lavage fluids were collected on days 28 and 56 post
517
administration. The IgA anti-measles levels in the intestinal lavages indicate that oral
518
vaccination with alginate coated low MW CS nanoparticles induced stimulation of 16 Page 16 of 32
519
IgA-secreting cells of the GALT. There was a significant correlation between the IgA
520
levels produced by the nanoparticles vs. CS MW, similar to the case with humoral
521
response. In contrast, subcutaneous administration of formulation was not able to
522
induce any IgA response, not even after several booster immunizations (Fig. 6).
524
ip t
523 Fig. 6.
525
527
4.
Conclusion
cr
526
An effective vaccine formulation for oral delivery maintains the antigen in a stable form, protects antigen from enzymatic degradation and harsh conditions in the
529
stomach and intestine, adheres to the mucosal surface, ensures the antigen remains
530
in the gastro intestinal tract region long enough for the antigen to interact with the
531
lymphatic system, and efficiently stimulates innate responses, and evokes adaptive
532
immune responses that are appropriate for the target pathogen. The vaccine-loaded
533
alginate coated CS LMW nanoparticles could be prepared with suitable and
534
appropriate particle sizes, which is a very important factor in the delivery of the
535
vaccine to the induction site of mucosa-associated lymphoid tissue for proper
536
immune stimulation. Moreover, alginate coating onto the surface of nanoparticles
537
could modulate the release behaviour of the antigen and could effectively protect
538
antigen from degradation against acidic medium (pH 2) in vitro for at least 2 h. Both
539
systemic and local immune responses can be induced in a dose- and time-
540
dependent manner through vaccine-loaded CS nanoparticles. The data presented in
541
this study evince suggest that alginate-coated LMW CS nanoparticles can be used
542
widely for the oral delivery of therapeutics.
544 545 546
an
M
d
te
Ac ce p
543
us
528
Acknowledgements
The authors thank Prof Debesh Chandra Majumder and Prof Pranabesh
Chakraborty for the encouragement and support provided.
547 548 549
References
550
Amoozgar, Z., Park, J., Lin, Q., & Yeo, Y. (2012). Low molecular-weight chitosan as
551
a pH-sensitive stealth coating for tumor-specific drug delivery. Molecular
552
Pharmaceutics, 9, 1262-1270. 17 Page 17 of 32
553
Anal, A. K., Bhopatkar, D., Tokura, S., Tamura, H., & Stevens, W. F. (2003).
554
Chitosan-alginate multilayer beads for gastric passage and controlled intestinal
555
release of protein. Drug Development and Industrial Pharmacy, 29, 713-724.
556
Baumann, U. (2008). Mucosal vaccination against bacterial respiratory infections.
558
Expert Review Vaccines, 7, 1257-1276.
ip t
557
Borges, O,. Cordeiro-da-Silva, A., Romeijn, S. G., Amidi, M., de Sousa, A., Borchard, G., & Junginger, H. E. (2006). Uptake studies in rat Peyer's patches,
560
cytotoxicity and release studies of alginate coated chitosan nanoparticles for
561
mucosal vaccination. Journal of Controlled Release, 114, 348-358.
cr
559
Borges, O., Silva, M., de Sousa, A., Borchard, G., Junginger, H. E., & Cordeiro-da-
563
Silva, A. (2008). Alginate coated chitosan nanoparticles are an effective
564
subcutaneous adjuvant for hepatitis B surface antigen. International
565
Immunopharmacology, 8, 1773-1780.
an
us
562
Borges, O., Tavares, J., de Sousa, A., Borchard, G., Junginger, H. E., & Cordeiro-
567
da-Silva, A. (2007). Evaluation of the immune response following a short oral
568
vaccination schedule with hepatitis B antigen encapsulated into alginate-coated
569
chitosan nanoparticles. European Journal of Pharmaceutical Sciences, 32, 278-
570
290.
d
M
566
Calvo, P., Remunan-Lopez, C., Vila-Jato, J. L., & Alonso, M. J. (1997). Chitosan and
572
chitosan/ethylene oxide-propylene oxide block copolymer nanoparticles as
573
novel carriers for proteins and vaccines. Pharmaceutical Research, 14, 1431-
Ac ce p
574
te
571
1436.
575
Chen, F., Zhang, Z. R., & Huang, Y. (2007). Evaluation and modification of N-
576
trimethyl chitosan chloride nanoparticles as protein carriers. International
577 578 579 580 581
Journal of Pharmaceutics, 336, 166-173.
Costa, P., & Sousa Lobo, J. M. (2003). Evaluation of mathematical models describing drug release from estradiol transdermal systems. Drug Development and Industrial Pharmacy, 29, 89-97. Csaba, N., Koping-Hoggard, M., & Alonso, M. J. (2009). Ionically crosslinked
582
chitosan/tripolyphosphate nanoparticles for oligonucleotide and plasmid DNA
583
delivery. International Journal of Pharmaceutics, 382, 205-214.
584 585
Czerkinsky, C., & Holmgren, J. (2009). Enteric vaccines for the developing world: a challenge for mucosal immunology. Mucosal Immunology, 2, 284-287.
18 Page 18 of 32
586 587
Delie, F., & Blanco-Prieto, M. J. Polymeric particulates to improve oral bioavailability of peptide drugs. Molecules, 10, 65-80. Du, C., Zhao, J., Fei, J., Gao, L., Cui, W., Yang, Y., & Li, J. (2013). Alginate-Based
589
Microcapsules with a Molecule Recognition Linker and Photosensitizer for the
590
Combined Cancer Treatment. Chemistry – An Asian Journal, 8, 736-742.
591
ip t
588
Eyles, J. E., Sharp, G. J., Williamson, E. D., Spiers, I. D., & Alpar, H. O. (1998). Intra nasal administration of poly-lactic acid microsphere co-encapsulated Yersinia
593
pestis subunits confers protection from pneumonic plague in the mouse.
594
Vaccine, 16, 698-707.
Fernandes, J. C., Qiu, X., Winnik, F. M., Benderdour, M., Zhang, X., Dai, K., & Shi,
us
595
cr
592
Q. (2012). Low molecular weight chitosan conjugated with folate for siRNA
597
delivery in vitro: optimization studies. International Journal of Nanomedicine, 7,
598
5833-5845.
600 601
Fooks, A. R. (2004). Development of oral vaccines for human use. Current Opinion in Molecular Therapeutics, 2, 80-86.
M
599
an
596
Gan, Q., Wang, T., Cochrane, C., & McCarron, P. (2005). Modulation of surface charge, particle size and morphological properties of chitosan-TPP
603
nanoparticles intended for gene delivery. Colloids and Surface B Biointerfaces,
604
44, 65-73.
607 608 609 610 611 612 613
te
606
Gazori, T., Khoshayand, M. R., Azizi, E., Yazdizade, P., Nomani, A., & Haririan, I. (2009). Evaluation of Alginate/Chitosan nanoparticles as antisense delivery
Ac ce p
605
d
602
vector: Formulation, optimization and in vitro characterization. Carbohydrate Polymers, 77, 599-606.
Godbey, W. T., Wu, K. K., & Mikos, A. G. (1999). Size matters: molecular weight affects the efficiency of poly(ethylenimine) as a gene delivery vehicle. Journal of Biomedical Material Research, 45, 268-275.
Hoff, J. (2000). Methods of blood collection in the lab mouse. Laboratory Animals, 29, 49–53.
614
Huang, M., Khor, E., & Lim, L. Y. (2004). Uptake and cytotoxicity of chitosan
615
molecules and nanoparticles: effects of molecular weight and degree of
616
deacetylation. Pharmaceutical Research, 21, 344-353.
617 618
Kim, B., Bowersock, T., Griebel, P., Kidane, A., Babiuk, L.A., Sanchez, M., AttahPoku, S., Kaushik, R. S., & Mutwiri, G. K. (2002). Mucosal immune responses
19 Page 19 of 32
619
following oral immunization with rotavirus antigens encapsulated in alginate
620
microspheres. Journal of Controlled Release, 85, 191-202. Kim, S. H., Lee, K. Y., & Jang, Y. S. (2012). Mucosal Immune System and M Cell-
622
targeting Strategies for Oral Mucosal Vaccination. Immune Network, 12, 165-
623
175.
ip t
621
Kostrzak, A., Cervantes Gonzalez, M., Guetard, D., Nagaraju, D. B., Wain-Hobson,
625
S., Tepfer, D., Pniewski, T., & Sala, M. (2009). Oral administration of low
626
doses of plant-based HBsAg induced antigen-specific IgAs and IgGs in mice,
627
without increasing levels of regulatory T cells. Vaccine, 27, 4798-4807.
Lavertu, M., Methot, S., Tran-Khanh, N., & Buschmann, M. D. (2006). High efficiency
us
628
cr
624
gene transfer using chitosan/DNA nanoparticles with specific combinations of
630
molecular weight and degree of deacetylation. Biomaterials 27: 4815-24
631
an
629
Lee, E., Lee, J., & Jon, S. (2010). A novel approach to oral delivery of insulin by conjugating with low molecular weight chitosan. Bioconjugate Chemistry, 21,
633
1720-1723.
M
632
Lee, S. J., Koo, H., Jeong, H., Huh, M. S., Choi, Y., Jeong, S. Y., Byun, Y., Choi, K.,
635
Kim, K., & Kwon. I. C. (2011). Comparative study of photosensitizer loaded and
636
conjugated glycol chitosan nanoparticles for cancer therapy. Journal of
637
Controlled Release, 152, 21-29.
640 641 642 643 644 645 646 647
te
639
Li. X., Kong. X., Shi. S., Zheng. X., Guo, G., Wei, Y. & Qian, Z. (2008). Preparation of alginate coated chitosan microparticles for vaccine delivery. BMC
Ac ce p
638
d
634
Biotechnology, 8, 89.
Liu, Z., Lv, D., Liu, S., Gong, J., Wang, D., Xiong, M., Chen, X., Xiang, R., & Tan, X. (2013). Alginic acid-coated chitosan nanoparticles loaded with legumain DNA vaccine: effect against breast cancer in mice. PLoS One 8: e60190
Malik, B., Goyal, A. K., Zakir, F., & Vyas, S. P. (2011). Surface engineered nanoparticles for oral immunization. Journal of Biomedical Nanotechnology, 7,
132-134. Mao, S., Shuai, X., Unger, F., Simon, M., Bi, D., & Kissel, T. (2004). The
648
depolymerization of chitosan: effects on physicochemical and biological
649
properties. International Journal of Pharmaceutics, 281, 45-54.
650
Mosmann, T. (1983). Rapid colorimetric assay for cellular growth and survival:
651
application to proliferation and cytotoxicity assays. Journal of Immunological
652
Methods, 65, 55-63. 20 Page 20 of 32
653 654
Neutra, M. R., & Kozlowski, P. A. (2006). Mucosal vaccines: the promise and the challenge. Nature Reviews Immunology, 6, 148-158. Oliveira, C. R., Rezende, C. M., Silva, M. R. Pego, A. P., Borges, O., & Goes, A. M.
656
(2012). A new strategy based on SmRho protein loaded chitosan nanoparticles
657
as a candidate oral vaccine against schistosomiasis. PLoS Neglected Tropical
658
Diseases, 6, e1894.
659
ip t
655
Opanasopit, P., Aumklad, P., Kowapradit, J., Ngawhiranpat, T., ApirakaraMWong, A., Rojanarata, T., & Puttipipatkhachorn, S. (2007). Effect of salt forms and
661
molecular weight of chitosans on in vitro permeability enhancement in intestinal
662
epithelial cells (Caco-2). Pharmaceutical Development and Technology, 12,
663
447-455.
us
cr
660
Prabakaran, M., Ho, H-T., Prabhu, N., Velumani, S., Szyporta, M., He, F., Chan, K-
665
P., Chen, L-M., Matsuoka, Y., Donis, R. O., & Kwang, J. (2009). Development
666
of Epitope-Blocking ELISA for Universal Detection of Antibodies to Human
667
H5N1 Influenza Viruses. PLoS ONE, 4, e4566.
M
an
664
Richardson, S. C., Kolbe, H. V., & Duncan, R. (1999). Potential of low molecular
669
mass chitosan as a DNA delivery system: biocompatibility, body distribution
670
and ability to complex and protect DNA. International Journal of Pharmaceutics,
671
178, 231-243.
674
te
673
Sergent, J. A., Paget, V., & Chevillard, S. (2012). Toxicity and genotoxicity of nanoSiO2 on human epithelial intestinal HT-29 cell line. The Annals of Occupational
Ac ce p
672
d
668
Hygiene, 56, 622-630.
675
Song, M. K., Vindurampulle, C. J., Capozzo, A. V., Ulmer, J., Polo, J. M., Pasetti, M.
676
F., Barry, E. M., & Levine, M. M. (2005). Characterization of immune responses
677 678
induced by intramuscular vaccination with DNA vaccines encoding measles virus hemagglutinin and/or fusion proteins. Journal of Virology, 79, 9854-9861.
679
van der Merwe, S. M., Verhoef, J. C., Verheijden, J. H., Kotze, A. F., Junginger, H.
680
E. (2004). Trimethylated chitosan as polymeric absorption enhancer for
681
improved peroral delivery of peptide drugs. European Journal of Pharmaceutics
682
and Biopharmacutics, 58, 225-235.
683
Vila, A., Sanchez, A., Janes, K., Behrens, I., Kissel, T., Vila Jato, J. L., Alonso, M. J.
684
(2004). Low molecular weight chitosan nanoparticles as new carriers for nasal
685
vaccine delivery in mice. European Journal of Pharmaceutics and
686
Biopharmaceutics, 57, 123-131. 21 Page 21 of 32
688 689 690 691
Wee, S., & Gombotz, W. R. (1998). Protein release from alginate matrices. Advanced Drug Delivery Reviews, 31, 267-285. Yang, Y., He, Q., Duan, L., Cui, Y., & Li, J. (2007). Assembled alginate/chitosan nanotubes for biological application. Biomaterials, 28, 3083-3090. Yoo, M. K., Kang, S. K., Choi, J. H., Park, I. K., Na, H. S., Lee, H. C., Kim, E. B.,
ip t
687
Lee, N. K., Nah, J. W., Choi, Y. J., & Cho, C. S. (2010). Targeted delivery of
693
chitosan nanoparticles to Peyer's patch using M cell-homing peptide selected
694
by phage display technique. Biomaterials, 31, 7738-7747.
695
cr
692
Zaharoff, D. A., Rogers, C. J., Hance, K. W., Schlom, J., & Greiner, J. W. (2007). Chitosan solution enhances both humoral and cell-mediated immune
697
responses to subcutaneous vaccination. Vaccine, 25, 2085-2094.
699
Zhu, Q., & Berzofsky, J. A. (2013). Oral vaccines: directed safe passage to the front
an
698
us
696
line of defense. Gut Microbes, 4, 246-252.
List of abbreviations used: PAGE- polyacrylamide gel electrophoresis, GALT-gut
701
associated lymphoid tissue
M
700
702
te Ac ce p
704
d
703
22 Page 22 of 32
Fig. 1. Scanning electron microscopy image of antigen loaded alginate coated CS
705
(MW 106 kDa) nanoparticle after freeze-drying process, magnification 6000X.
706
Fig. 2. In vitro release profiles of antigen from uncoated and alginate coated CS
707
nanoparticles in PBS at 37 OC in the first 120 h.
708
Fig. 3. Gel electrophoresis analysis, obtained under conditions of alginate coated
709
(Lane 1) and uncoated (Lane 2) CS (MW 106 kDa) nanoparticles, alginate coated
710
(Lane 3) CS (MW 42 kDa) nanoparticles containing antigen pre-treated with 0.01 M
711
HCl for 2 h, then sustained release in PBS for 24 h. Measles antigen pre-treated with
712
0.01 M HCl for 2 h, then incubation with PBS for another 24 h (Lane 4), measles
713
antigen incubation with PBS for 24 h (Lane 5), Molecular weight marker (Lane 6).
714
Fig. 4. Effect of (A) uncoated and (B) alginate coated CS nanoparticles at different
715
concentrations on the viability of HT 29 cells determined by MTT assay. Indicated
716
values were mean ± SD (n = 6). Statistical difference between the control groups and
717
formulations are reported as * p < 0.05, no significant difference, ns p > 0.05.
718
Fig. 5. Anti-measles IgG titres of mice immunized with different alginate-coated CS
719
LMW nanoparticle oral formulations of measles vaccine. Values are expressed as
720
antibody titres of mice on day 7, 14, 28, 56 post administration. Mean reciprocal
721
dilutions are represented as the end point titre (log10) ± SD. Titres were defined as
722
the highest dilution resulting in an absorbance value twice that of non-immune
723
plasma.
724
Fig. 6. Secretory anti measles sIgA profile after oral administration of various
725
formulations in CS LMW nanoparticles detected in individual intestine washing
726
samples of mice giving an OD of 0.1 after subtraction of background and presented
727
as geometric mean titre per group.
729 730
cr
us
an
M
d
te
Ac ce p
728
ip t
704
731
23 Page 23 of 32
731
Table 1. Measles antigen loaded vaccine formulation for immunization study
732 Table 2. Particle size, zeta potential and loading capacity of antigen loaded three
734
different molecular weight of CS-LMW nanoparticles without or with alginate coated
735
nanoparticles (n = 3, mean ± SD)
ip t
733
736
Table 3. Results of model fitting of antigen release of various nanoparticles in
738
phosphate buffer pH 7.4
cr
737
739
Table 1. Measles antigen loaded vaccine formulation for immunization study Formulation
us
740
Description
Group
AL-CS-42-MA
Measles
an
code antigen
loaded
alginate I
coated CS (MW 42 kDa) nanoparticles Measles
antigen
loaded
M
AL-CS-74-MA
alginate II
coated CS (MW 74 kDa) nanoparticles Measles
antigen
coated
CS
loaded
(MW
d
AL-CS-106-MA
106
alginate III kDa)
AL-CS-42
te
nanoparticles
Alginate coated CS (MW 42 kDa) IV
Ac ce p
nanoparticles, used as negative control
MA-SC
Measles
subcutaneously,
antigen used
given V as
positive
control
MA-ORAL 741 742
Measles antigen given orally
VI
24 Page 24 of 32
742
Table 2. Particle size, zeta potential and loading capacity of antigen loaded three
743
different molecular weight of CS-LMW nanoparticles without or with alginate coated
744
nanoparticles (n=3, mean ± SD) Mean particle Zeta
Composition
size (nm)
LMW-CS (42 kDa)- 284 ± 36
cr
Alginate/LMW-CS
432 ± 52
+1.65 ± 0.4 0.368 ± 0.06
LMW-CS (74 kDa)- 507 ± 37
+21.4 ± 1.2 0.412 ± 0.04
us
(42 kDa)-Antigen 3
+14.2 ± 0.6 0.472 ± 0.05
an
2.50 ± 0.32
Alginate/LMW-CS
1003 ± 45
-24.1 ± 0.9
0.484 ± 0.03
1.23 ± 0.18
(106 kDa)-Antigen
-10.1 ± 0.6
(106
M
LMW-CS
te
746
2.10 ± 0.26
Ac ce p
745
3.41 ± 0.88
852 ± 76
651 ± 28
kDa)-Antigen 6
1.56 ± 0.14
0.438 ± 0.08
Alginate/LMW-CS (74 kDa)-Antigen
5
2.87 ± 0.46
d
Antigen 4
capacity (%)
+31.6 ± 1.4 0.310 ± 0.08
Antigen 2
Loading
potential (mV)
1
PDI
ip t
S. No.
25 Page 25 of 32
747
phosphate buffer pH 7.4 Para
Formulation code
model
meter
NP-CS-
NP-CS-
NP-CS-
NP-AL-
NP-AL-
NP-AL-
42
74
106
CS-42
CS-74
CS-106
0.684
0.698
0.742
0.664
0.689
0.631
0.011
0.009
0.008
0.006
0.005
0.004
0.915
0.928
0.952
0.927
6.390
5.725
5.044
4.214
3.666
3.017
0.991
0.986
0.987
0.982
0.986
0.989
12.846
10.889
8.684
7.967
6.527
5.632
0.338
0.350
0.374
0.352
0.366
0.355
0.994
0.995
0.996
0.997
0.995
0.994
13.809
12.454
10.123
9.526
7.631
6.225
0.272
0.289
0.250
0.280
0.297
-0.002
-0.003
-0.003
-0.002
51.14
41.07
36.28
31.83
Korsmey erPeppas
Makoid-
te
Banakar
Ac ce p
0.295
-0.001
-0.002
Referen 66.05
cr
Higuchi
0.949
us
order
an
First
ip t
Kinetic
M
Table 3. Results of model fitting of antigen release of various nanoparticles in
d
746
0.936
ce
748 749
26 Page 26 of 32
Ac
ce
pt
ed
M
an
us
cr
i
Figure(s)
Page 27 of 32
Ac
ce
pt
ed
M
an
us
cr
i
Figure(s)
Page 28 of 32
Ac ce p
te
d
M
an
us
cr
ip t
Figure(s)
Page 29 of 32
Ac ce p
te
d
M
an
us
cr
ip t
Figure(s)
Page 30 of 32
Ac
ce
pt
ed
M
an
us
cr
i
Figure(s)
Page 31 of 32
Ac
ce
pt
ed
M
an
us
cr
i
Figure(s)
Page 32 of 32