Xanthan gum stabilized gold nanoparticles: Characterization, biocompatibility, stability and cytotoxicity

Xanthan gum stabilized gold nanoparticles: Characterization, biocompatibility, stability and cytotoxicity

Accepted Manuscript Title: Xanthan gum stabilized gold nanoparticles: Characterization, Biocompatibility, Stability and Cytotoxicity Author: Deep Pooj...

3MB Sizes 0 Downloads 56 Views

Accepted Manuscript Title: Xanthan gum stabilized gold nanoparticles: Characterization, Biocompatibility, Stability and Cytotoxicity Author: Deep Pooja Sravani Panyaram Hitesh Kulhari Shyam S Rachamalla Ramakrishna Sistla PII: DOI: Reference:

S0144-8617(14)00273-2 http://dx.doi.org/doi:10.1016/j.carbpol.2014.03.041 CARP 8703

To appear in: Received date: Revised date: Accepted date:

8-1-2014 18-2-2014 5-3-2014

Please cite this article as: Pooja, D., Panyaram, S., Kulhari, H., Rachamalla, S. S., & Sistla, R.,Xanthan gum stabilized gold nanoparticles: Characterization, Biocompatibility, Stability and Cytotoxicity, Carbohydrate Polymers (2014), http://dx.doi.org/10.1016/j.carbpol.2014.03.041 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.

1 2

Xanthan gum stabilized gold nanoparticles: Characterization, Biocompatibility, Stability and Cytotoxicity

3 Deep Pooja,a Sravani Panyaram,a Hitesh Kulhari,a,b Shyam S Rachamalla,c Ramakrishna Sistlaa,* a

Medicinal Chemistry & Pharmacology Division, bIICT-RMIT Research Centre, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.

ip t

4 5 6 7

c

8 9

cr

Faculty of Pharmacy, College of Technology, Osmania University, Hyderabad, India

us

10 11

*Corresponding author:

13

Sistla Ramakrishna, PhD

14

Principal Scientist,

15

Medicinal Chemistry & Pharmacology Division,

16

CSIR-Indian Institute of Chemical Technology,

17

Hyderabad-500007, India

18

Phone: +91-40-27193753 (office)

19

Email: [email protected]

23 24 25 26 27

M

d

te

Ac ce p

20 21 22

an

12

28

Highlights

29 30



Gold nanoparticles were prepared using xanthan gum as reducing and capping agent.

31



Different formulation and process variables were optimized. 1 Page 1 of 36

32



XG stabilized nanoparticles were non-toxic and biocompatible.

33



DOX loaded on GNP showed more therapeutic efficacy than free DOX in cancer cells.

35



Formulations displayed colloidal stability against pH and ionic strength changes.

ip t

34

cr

36 37

us

38 39

an

40 41

M

42

46 47 48 49 50 51 52

te

45

Ac ce p

44

d

43

53 54

Graphical Abstract

55

2 Page 2 of 36

ip t cr us

56

an

57 58

M

59

62

te

61

d

60

Abstract

Xanthan gum (XG) has been widely used in food, pharmaceutical and cosmetic industries. In

64

the present study, we explored the potential of XG in the synthesis of gold nanoparticle. XG

65

was used as both reducing and stabilizing agent. The effect of various formulation and

66

process variables such as temperature, reaction time, gum concentration, gum volume and

67

gold concentration, in GNP preparation was determined. The XG stabilized, rubey-red XGNP

68

were obtained with 5 ml of XG aqueous solution (1.5 mg/ml). The optimum temperature was

69

80 ºC whereas the reaction time was 3 h. The optimized nanoparticles were also investigated

70

as drug delivery carrier for doxorubicin hydrochloride. DOX loaded gold nanoparticles

71

(DXGP) were characterized by dynamic light scattering, TEM, FTIR, and DSC analysis. The

72

synthesized nanoparticle showed mean particle size of 15-20 nm and zeta potential -29.1 mV.

73

The colloidal stability of DXGP was studied under different conditions of pH, electrolytes

74

and serum. Nanoparticles were found to be stable at pH range between pH 5-9 and NaCl

75

concentration up to 0.5 M. In serum, nanoparticles showed significant stability up to 24 h.

76

During toxicity studies, nanoparticles were found biocompatible and non-toxic. Compared

Ac ce p

63

3 Page 3 of 36

77

with free DOX, DXGP displayed 3 times more cytotoxicity in A549 cells. In conclusion, this

78

study provided an insight to synthesize GNP without using harsh chemicals.

79 80

Key words: Xanthan Gum; Gold nanoparticles; Reducing agent, Doxorubicin, Cytotoxicity, Stability

ip t

81 82

cr

83

us

84 85

an

86 87 88

M

89

92

1.

Introduction

te

91

d

90

Xanthan gum (XG) is an anionic, high molecular weight, exo-polysaccharide

94

produced by aerobic fermentation of sugars by Xanthomonas campestris. Its basic chain

95

consists of a β-(1→4) linked glucose backbone with the substitution of a charged

96

trisaccharide side-chains of [β-(1→3)-mannose-α-(1→2)-glucuronic acid-β-(1→4)-mannose]

97

on alternate glucose residues (Jian, Zhu, Zhang, Sun, & Jiang, 2012). It is non-toxic,

98

hydrophilic and biodegradable bio-polymer. XG is being used in food, cosmetic and

99

pharmaceutical industries. The industrial applications of XG are based upon its exceptional

100

rheological properties. XG is soluble in both cold and hot water, hydrates quickly and

101

produces high viscosity at low concentration (Sereno, Hill, & Mitchell, 2007; Sharma,

102

Naresh, Dhuldhoya, Merchant, Merchant, 2006). In pharmaceutical industries, XG has been

Ac ce p

93

4 Page 4 of 36

reported for formulation of both solid and liquid dosage forms. In solid dosage formulation, it

104

is used as controlled release agent (Jian et al., 2012; Phaechamud and Ritthidej, 2007; Santos,

105

H., Veiga, F., Pina, M. E., & Sousa, 2005; Sinha, Mittal, Bhutani, & Kumaria, 2004) whereas

106

in liquid formulations, it is used as thickening agent, suspending agent and emulsion

107

stabilizer (Desplanques, Renou, Grisel, & Malhiac, 2012).

ip t

103

Recently, XG has also been used in the preparation and stabilization of inorganic iron

109

and palladium nanoparticles (Fan et al., 2013; Xue & Sethi, 2012; Comba , Dalmazzo,

110

Santagata, & Sethi, 2011; Vecchia, Luna, & Sethi, 2009). Nanoparticles are solid structures

111

with a size below 200 nm and have found their applications in sensing, imaging and in drug

112

and gene delivery. Gold nanoparticles (GNP) are one of the most commonly explored and

113

used nanoparticles in drug delivery because of controlled size, improved efficacy and

114

targeted delivery (Almeida, Figueroa, & Drezek; 2013; Pissuwan, Niidome, & Cortie, 2011).

115

As drug delivery carrier, GNP has been used for the delivery of both hydrophilic and

116

hydrophobic drugs (Oliveira et al., 2013; Chen et al., 2007; Gibson, Khanal, & Zubarev,

117

2007; Aryal, Grailer, Pilla, Steeber, & Gong, 2009; Prabaharan M, Grailer JJ, Pilla S, Steeber

118

DA, & Gong, 2009). But, the conventional synthesis of GNP involves the use of chemicals

119

like sodium borohydrate, tri-sodium citrate etc as reducing agent (Burygin et al., 2009;

120

Duncan, Kim, & Rotello, 2010; Khan, Jung et al., 2013; Vishakante, & Siddaramaiah, 2013;

121

Saha et al., 2007). These GNP have been found to be very unstable and form aggregates with

122

the slight change in pH and electrolyte concentration (Mirza and Shamshad, 2011; Rouhana,

123

Jaber, & Schlenoff, 2007). The GNP prepared with gellan gum showed the stability between

124

pH 4 to 8 and addition of NaCl up to 0.1M (Dhar, Reddy, Shiras, Pokharkar, & Prasad,

125

2008). Natural gums stabilize the inorganic nanoparticles by two mechanisms: first, by

126

adsorbing to the surface of nanoparticles which creates steric repulsion among the particles.

127

In second mechanism, they increase the viscosity of nanoparticle suspension, and therefore

Ac ce p

te

d

M

an

us

cr

108

5 Page 5 of 36

slow down the aggregation processes (Tiraferri, Chen, Sethi, & Elimelech, 2008; Xue &

129

Sethi, 2012; Comba & Sethi, 2009). The aim of this investigation was to synthesize the

130

environment friendly gold nanoparticles using XG as reducing agent. The nanoparticle

131

preparation was optimized for gum concentration and volume, gold concentration,

132

temperature and reaction time. Gum stabilized GNP were studied for its utility as drug

133

delivery carrier using Doxorubicin hydrochloride as model drug. Doxorubicin hydrochloride

134

is hydrophilic in nature and has been clinically used for the treatment of various cancers,

135

haematological malignancies, soft tissue sarcomas and solid tumors (Carvalho et al., 2009;

136

Laginha, Verwoert, Charrois, & Allen, 2005). Two measure limitations of DOX are non-

137

specific cytotoxicity and multi drug resistance. Multi drug resistance to DOX is due to drug

138

efflux by P-glycoproteins. Doxorubicin is substrate for P-glycoproteins which efflux DOX

139

and decrease intracellular drug level. DOX loaded GNP have shown to overcome both the

140

problems of non-specific toxicity and drug resistance (Gu, Cheng, Man, Wong, & Cheng,

141

2012). The prepared nanoparticles were also investigated for biocompatibility, stability and

142

cytotoxicity study in lung cancer cells.

143

2.

144

2.1.

cr

us

an

M

d

te

Ac ce p

145

ip t

128

Materials and methods Materials

Xanthan gum and Tetrachloroauric acid (HAuCl4) were purchased from Sigma-

146

Alderich (St Louis, MO, USA). Doxorubicin hydrochloride was received as gift sample from

147

TherDose pharma pvt ltd (Hyderabad, India). The chemical used for buffer preparations were

148

of analytical grade and were purchased from sd Fine-Chem Ltd. (Hyderabad, India). MTT (3-

149

(4, 5-dimethylthazol-2-yl)-2, 5-diphenyl tetrazolium bromide, Dulbecco’s modified eagle

150

medium (DMEM), trypsin–EDTA, fetal bovine serum (FBS) and antibiotic solution (10,000

151

U/ml penicillin, 10 mg/ml streptomycin) were purchased from Sigma-Aldrich (St Louis, MO, 6 Page 6 of 36

152

USA). Cell culture plastic wares were obtained from Tarsons Products Pvt. Ltd. (Kolkata,

153

India). All the formulations were prepared in MilliQ water.

154

2.2.

Preparation of gum solution The stock solution of gum was prepared by dissolving 500 mg of the gum in 100 ml

156

water and was stirred overnight at room temperature. The solution was centrifuged to remove

157

the insoluble materials and supernatant was lyophilized. The lyophilized dry powder was

158

dissolved in water to get desired concentration of xanthan gum.

159

2.3.

us

cr

ip t

155

an

Synthesis of gold nanoparticles

Gold nanoparticles were prepared by reducing the aqueous solution of HAuCl4 by

161

heating at 80 ºC in the presence of XG solution (1.5 mg/ml). The change in colour was

162

obtained from colourless to purple to rubey-red after 2 h. The colloidal solution was cooled at

163

room temperature and stored in amber colour vials at 4 ºC. The nanoparticle formulation

164

showed the absorption maxima at 525 nm.

te

d

M

160

166 167

Ac ce p

165

2.4.

Optimization of formulation and process variables

The effect of formulation and process variables such as gum concentration, gold

168

concentration, gold to gum volume ratio, reaction time and temperature were studied by

169

changing one parameter at a time and keeping other constant.

170

2.5.

171

2.5.1. Hydrodynamic diameter and polydispersity

Characterizations of Xanthan gum stabilized gold nanoparticles(XGNP)

172

The hydrodynamic diameter and polydispersity index of GNP were determined by

173

dynamic light scattering using Zetasizer Nano-ZS (Malvern instrument Ltd., Malvern, UK). 7 Page 7 of 36

Before measurement, the samples were diluted appropriately to get particle count rate

175

between 100-300 kcps and instrument was set up at 25 ºC with a backscattering angle of

176

173°.

177

2.5.2. Transmission electron microscopy (TEM)

ip t

174

A drop of sample was placed on carbon coated copper grid, air dried at room

179

temperature and stained with 2% uranyl acetate. The nanoparticle size measurement was

180

done using transmission electron microscope (Hitachi, H-7500) and average of 10

181

nanoparticle size was considered as the size of the sample.

182

2.5.3. Surface charge

an

us

cr

178

The electrokinetic properties of the GNP were determined by measuring the zeta

184

potential using Zetasizer Nano-ZS. The samples were diluted 10 times with MilliQ water

185

before the measurement.

186

2.5.4. Fourier transform infrared analysis

d te

An amount of 2 mg of sample was mixed with 100 mg of potassium bromide and

Ac ce p

187

M

183

188

compressed to form a pellet. The pellet was placed in pellet holder and scanned for the

189

measurement of % transmittance in the wave number range of 4000 to 450 cm-1 using FTIR

190

spectrophotometer (Perkin Elmer, USA). The spectra requisition was carried out using the

191

software Spectrum One (Perkin Elmer, USA).

192

2.6.

Doxorubicin loading to nanoparticles

193

Blank gold nanoparticles were dispersed in phosphate buffer saline and were incubated

194

with 1 mg of DOX solution (1 mg/ml) at room temperature overnight. The nanoparticles

195

dispersion was centrifuged at 15000 rpm for 30 min. DOX loading was determined by

8 Page 8 of 36

196

measuring the free drug content in supernatant. Percent drug loading was calculated as

197

follows: % DOX loading = (1-DS/DT) × 100

199

Whereas, DS is amount of DOX present in supernatant, DT = Total amount of DOX

200

loaded initially.

201

2.7.

ip t

198

cr

In vitro drug release studies

The in vitro drug release studies were performed using dialysis method. In dialysis

203

tubing, the nanoparticles equivalent to 1 mg of DOX were dispersed in 1 ml of distilled water

204

and placed in 100 ml of release medium (phosphate buffer saline pH 7.4 and sodium acetate

205

buffer pH 4.5) at 37 ºC in dark. Three millilitres of sample was withdrawn at different time

206

intervals up to 12 hours and was replaced with same volume of fresh medium. The samples

207

were analyzed for drug content using UV/VIS spectrophotometer (Perkin Elmer, Lambda

208

25, USA) at 480 nm.

209

2.8.

d

M

an

us

202

te

Hemolytic toxicity studies

The biocompatibility of prepared gold nanoparticles was determined by estimating the

211

% hemolysis, caused by nanoparticles. The assay procedure reported by Kumar et al. 2011,

212

was used with slight modification (Kumar, Paul, Sharma, 2011). Blank gold nanoparticles

213

were dispersed in normal physiological saline (0.9 %w/v NaCl). Varying concentrations (50-

214

200 µg/ml) of the dispersion were added into red blood cells suspension (2 %v/v), mixed well

215

and incubated at 37 °C. After 1 h, the samples were centrifuged at 3000 rpm for 5 min and

216

absorbance of supernatant was measured at 540 nm spectrophotometrically. The supernatant

217

absorbance of RBC suspension treated with distilled water and normal saline were taken as

218

standard and control; respectively. The % hemolysis was calculated as: % Hemolysis =

219

(Asample – Acontrol) / (Astandard - Acontrol) × 100

220

2.9.

Ac ce p

210

Cytotoxicity studies 9 Page 9 of 36

Cellular toxicity of pure DOX and DXNP was evaluated by MTT assay in A549 human

222

lung cancer cells. The assay is based on the mitochondrial reduction of yellow MTT, a

223

tetrazolium dye, to a highly coloured blue formazan product. About 10000 cells per well were

224

cultured in 96 wells plate and allowed to attach overnight. Next day, cells were incubated

225

with different concentration (0.1-10 µg/ml) of pure doxorubicin, blank gold nanoparticles

226

(positive control) and doxorubicin loaded gold nanoparticles. Cells were incubated in CO2

227

incubator at 37 °C for 48 h. The solution of each well was replaced with fresh serum free

228

media and 10 µl of MTT reagent (5 mg/ml) was added. After 4 h, the media was removed,

229

200 µl DMSO was added and incubated for 20 min. The absorbance was measured at 570 nm

230

using Spectra Max plus 384 UV-Visible plate reader. The cell viability was determined as a

231

percent based on the absorbance measured relative to the absorbance of untreated or control

232

cells. The half maximum inhibitory concentration (IC50) values were determined by non-

233

regression analysis using GraphPad Prism v. 5.03 (GraphPad Software Inc. CA).

234

2.10. Stability studies

235

2.10.1. Storage stability

cr

us

an

M

d te

Ac ce p

236

ip t

221

Doxorubicin loaded nanoparticles were stored at 4 °C and were studied for particle

237

size, zeta potential and drug content up to 1 month. Particle size and zeta potential were

238

determined by particle size analyzer and DOX content was determined using UV/VIS

239

spectrophotometer.

240

2.10.2. Effect of pH and ionic concentration of medium

241

For the determination of effect of pH on the stability, DXGP were incubated in

242

different pH medium (pH 3 to pH 11). Nanoparticles incubated in phosphate buffer saline pH

243

7.4 were taken as control. Absorbance was measured after 24 h and compared with

244

absorbance of control. 10 Page 10 of 36

The effect of ionic strength on the stability of DXNP was determined by incubating the

246

nanoparticles in deionised water containing varying concentrations (0.1-2M) of sodium

247

chloride (NaCl).

248

2.10.3. Serum Stability of DXGP

ip t

245

Serum stability of DXGP was evaluated at two different serum concentration

250

levels (10%v/v and 100% serum). Nanoparticles were dispersed in serum and particles

251

size, zeta potential and absorbance were measured at different time intervals up to 72 h.

252

2.11. Statistical analysis

an

us

cr

249

All the experiments were performed in triplicates. Data are expressed as Mean ± SD

254

(standard deviation) (n=3). The values of in vitro drug release, cytotoxicity and stability

255

studies were statistically analyzed using student t-test. Statistical significance level was set at

256

p < 0.05.

257

3.

Result and discussion

258

3.1.

Preparation and optimization of gold nanoparticles

d

te

Ac ce p

259

M

253

Gold nanoparticles were prepared by chemical reduction method using xanthan gum as

260

reducing agent. The role of gum concentration was studied by using gum solution in the

261

range of 0.5-2 mg/ml containing 10 mM of HAuCl4 concentration (Fig. 1a). The absorbance

262

in the UV spectrum which describes the reduction of GNP, was found to be directly

263

related to gum concentration. The intensity of the absorption band gradually increased and

264

the maximum intensity of absorption was attained at 1.5 mg/ml which indicated the

265

maximum gold nanoparticle concentration.

266

The effect of gold to gum volume ratio was evaluated by varying the gum volume in

267

the range of 2.5-10 ml (Fig. 1b). At lower gum volume (2.5 ml) a peak shift was observed 11 Page 11 of 36

and the absorption band shifted to 550 nm which suggested that lower gum volume was not

269

sufficient to reduce all Au ions. When the gum volume was changed from 5 to 7.5 ml

270

followed by 10 ml, there was no significant shift in the characteristic absorption band but the

271

absorbance was decreased. The maximum intensity of absorption was attained at 5 ml and

272

was considered optimum gum volume for the synthesis of XGNP. The effect of gold

273

concentration (Fig. 1c) and volume (Fig. 1d) was also studied and was found as per earlier

274

reports. A volume of 100 µl of 10 mM gold solution showed better absorbance at 525 nm.

cr

ip t

268

Reaction temperature and time are two important process parameters in the synthesis

276

on gold nanoparticles. In this study, the temperature was changed in the range of 60-90 °C

277

with 10 mM HAuCl4 concentration and 1.5 mg/ml gum solution (Fig. 2a). The maximum

278

intensity of the characteristic absorption band was attained at 80 ˚C which indicates the

279

maximum gold nanoparticle concentration. For the optimization of reaction time,

280

nanoparticle synthesis was studied up to 4 h. The absorbance was found to be increased up to

281

2 h and after that there was no change in the absorbance (Fig. 2b).

282

3.2.

an

M

d

te

Characterization of Xanthan gum stabilized gold nanoparticles The hydrodynamic diameter and polydispersity index of optimized XGNP were

Ac ce p

283

us

275

284

determined using dynamic light scattering method. The observed mean hydrodynamic

285

diameter was 41.2 ± 3.78 nm. The prepared nanoparticles showed high polydispersity (0.35-

286

0.6) which can be attributed to presence of wide range of particles. The high negative surface

287

potential (-47.2±2.59) indicated the high stability of nanoparticles and presence of gum

288

molecules on nanoparticle surface. The presence of acetyl group on O-6 position and pyruvic

289

acid linked with mannose sugar are responsible for the negative charge. After DOX loading,

290

the surface charge was decreased to -29.1±2.78 mV indicating the interaction between

291

positively charged amine group of DOX and negative acidic group of gum. The nanoparticle

292

surface morphology and actual particle size were determined by transmission electron 12 Page 12 of 36

293

microscopy. Figure 3 showed that nanoparticles were spherical in shape and 15-20 nm in

294

size. The chemical interactions between DOX and nanoparticles were studied by FTIR

296

analysis (Fig. 4). In XG spectra, the vibration peak of hydrogen bonded −OH and C–H

297

stretching were observed at 3416 and 2902 cm−1, respectively. The vibration peak of the

298

acetal was found at 1051.8 cm−1 whereas C=O stretching peak was found at 1728.4 cm−1. The

299

FITR spectra of blank nanoparticles exhibited all the characteristic peaks of XG at 3398,

300

1728 and 1042 cm−1. The peak at 1254.2 cm−1 presented the C-O-C group. The DXGP

301

showed principal IR peaks of both XG and DOX showing the non-covalent binding of DOX

302

to nanoparticles.

an

us

cr

ip t

295

DSC is a useful technique to explain the thermal stability and phase transitions. Figure

304

5 illustrates the DSC thermograms of pure DOX, native XG and DXGP. DSC scan of XG

305

exhibited endothermic transition at 125 °C. The endothermic peak of DOX appeared at its

306

melting point 195 °C. This peak was disappeared in DXGP spectra indicating that DOX was

307

not precipitated on nanoparticle surface but was present as molecular or amorphous

308

dispersion.

309

3.3.

d

te

Ac ce p

310

M

303

DOX loading

The observed doxorubicin loading was found to be 71.4 ± 2.53%. The high drug

311

loading can be attributed to presence of XG on the nanoparticle surface. Xanthan gum

312

contains negatively charged moieties such as pyruvic acid which interacts with positively

313

charged amine group of doxorubicin.

314

3.4.

In vitro drug release studies

13 Page 13 of 36

In vitro drug release from the nanoparticles was studied in PBS and SAB buffer (Fig.

316

6). The nanoparticles showed complete drug release within 10 h. Up to 4 h, there was no

317

significant difference (p > 0.05) in doxorubicin release in both the medium but at the end

318

DOX release was higher in SAB (98.1%) as compared to PBS (83.6%). The faster release of

319

DOX in SAB can be attributed to protonation of amine groups of DOX molecules which

320

increases hydrophilicity and solubility at lower pH. The protonation of amine group also

321

leads to decreased interaction between DOX and GNP (Minati et al., 2012). The PBS pH 7.4

322

is a simulated condition for blood plasma whereas SAB pH 4.5 is for the internal

323

environment of cancer cell. Thus the slower release of DOX in PBS and faster release in SAB

324

would help to release the maximum amount of drug in tumour and hence to enhance the

325

therapeutic efficacy of the formulation.

326

3.5.

M

an

us

cr

ip t

315

Hemolytic toxicity

Nanoparticles are reported to cause oxidative-stress induced hemolysis (Teodora,

328

2013). Therefore, the suitability for intravenous administration of nanoparticles is generally

329

determined by hemolytic study. In this study, dose-dependent hemolytic toxicity of xanthan

330

gum stabilized gold nanoparticle was carried out. The prepared nanoparticles did not show

331

hemolysis up to 200 µg/ml.

332

3.6.

te

Ac ce p

333

d

327

Cytotoxicity studies

Anti-proliferative activity of doxorubicin loaded xanthan gum stabilized gold

334

nanoparticles (DXGP) and free DOX was studied in A549 human lung cancer cells. The cell

335

viability was determined by MTT assay after 48 h of incubation with different concentrations

336

(Fig. 7). Blank xanthan stabilized gold nanoparticles (BXNP) were taken as positive control

337

and did not decrease the cell viability at any test concentration. It suggested that the prepared

338

nanoparticles were non-toxic and biocompatible. Drug loaded nanoparticles showed 14 Page 14 of 36

concentration-dependent cytotoxicity. The cell viability was decreased with the increase in

340

DOX concentration. The IC50 value for pure DOX and DXGP was 2.46 and 0.79 µg/ml,

341

respectively. The lower IC50 value of DXGP indicated the more cytotoxicity which can be

342

attributed to enhanced cellular uptake and hence increased intracellular drug concentration.

343

However, the exact mechanism and role of carbohydrate moieties present on nanoparticle in

344

cellular uptake require detailed investigation. One possible reason may be due to the

345

presence of mannose sugars in the composition of xanthan gum which was used for the

346

stabilization of GNP. It is documented that mannose receptors are up regulated in

347

human lung cancer cells A549 (Basuki et al., 2014; El-Boubbou et al., 2010). Basuki et al

348

reported the improvement in cell uptake of mannose functionalized iron nanoparticles

349

in human lung cancer cells A549. Hence, the presence of mannose in xanthan gum

350

might have contributed to increase in uptake of drug through receptor-mediated

351

endocytosis pathway.

d Stability studies

Ac ce p

354

3.7.

te

352 353

M

an

us

cr

ip t

339

The colloidal stability of DXGP was studied for both physical and chemical

355

stabilities. The change in particle size and zeta potential are shown in Fig. 8. No significant

356

(p>0.05) changes in particle size and zeta potential were observed up to 15 days. But,

357

chemical degradation of DXNP was faster than physical instability. The DOX content was

358

less than 90% within 24 hours. However, the DOX degradation was slower with DXGP in

359

comparison to pure DOX. The observed drug degradation rate constant (K) was 8.28 × 10-3

360

and 5.07 × 10-3 day-1 for pure DOX and DXGP (Fig. 8c). Therefore, decease in drug content

361

was due to light sensitivity of DOX itself not because of formulation.

15 Page 15 of 36

DXGP showed a wide range of pH stability (pH 5-pH 9). The observed absorbance of

363

DXGP at 480 nm was 1.109, 0.899, 0.908, 0.927 and 0.776 for pH 3, 5, 7, 9 and pH 11

364

respectively (Fig. 9a). The absorbance of control sample was 0.931. The results suggested

365

that XG or carbohydrate capping of nanoparticles helped to increase the colloidal stability of

366

gold nanoparticles. The decrease in absorbance at pH 11 may due to gel formation properties

367

of XG at higher pH levels (pH > 10) (Sharma et al., 2006).

cr

ip t

362

In other study, DXGP were treated with different NaCl concentration to determine the

369

stability of nanoparticles in varying ionic concentrations. Figure 9b showed the absorbance

370

pattern of DXGP after 24 h of incubation with different NaCl concentrations. The control

371

group was incubated in deionised water without NaCl. The DXGP were found to be stable up

372

to 0.5M NaCl concentration.

M

an

us

368

The stability of DXGP was further evaluated at two serum conditions-diluted

374

(10% v/v serum) and undiluted or 100% serum. Figure 10a showed the particle

375

diameter of DXGP exposed to serum, after different time period. In both the conditions,

376

particle size was increased continuously up to 72 h. But, the increase in particle

377

diameter was more in undiluted serum than diluted serum. After 2 h, particle size was

378

increased by 7.1 nm in 10% serum wile it increased by 31.5 nm in 100% serum. From

379

the figure 10a it is clear that DXGP were stable up to 24 h in both the conditions but

380

after that size of nanoparticle was increased tremendously. The increase in particle size

381

can be explained by the adsorption of serum proteins or other components on

382

nanoparticle surface. (Yang, Zhang, Wang, White, & Jiang, 2014; Zang & Zang, 2010).

383

The nanoparticle dispersion in distilled water had a high negative zeta potential of -30.2

384

mV and thus the electrostatic interactions between electronegative nanoparticles and

385

positive serum components can also not be ruled out. This possibility was well

386

supported by the zeta potential values observed after addition in to serum (Fig. 10b).

Ac ce p

te

d

373

16 Page 16 of 36

Zeta potential of nanoparticle was increased (less negative values) dramatically due to

388

charge neutralization. After 24 h, surface charge of DXGP was -18.2 and -14.5 mV in

389

10% and 100% serum, respectively. Higher zeta potential in 100% serum indicated the

390

more interactions between nanoparticles and serum proteins leading to higher particle

391

size.

ip t

387

The change in absorption pattern of DXGP with time was also observed over the

393

range of 350-750 nm. Interestingly, the absorption pattern in diluted serum (Fig. 10c)

394

was similar to that of spectra observed throughout the study but new absorption

395

spectrum was observed in 100% serum (Fig. 10d). It may be due to, as earlier

396

mentioned, adsorbed or electro-statically interacted serum components. However, no

397

change in absorption spectra was observed up to 24 h in both serum conditions. Overall,

398

DXGP were stable physically and chemically up to 24 h even under the extreme

399

conditions of serum.

400

402

4.

Conclusions

Ac ce p

401

te

d

M

an

us

cr

392

The applications of gold nanoparticles as multifunctional vehicle (drug delivery carrier,

403

imaging) are increasing because of ease of synthesis, biocompatibility and opportunity for

404

surface conjugation. In this investigation, gold nanoparticles were synthesized using natural

405

and biocompatible Xanthan gum as reducing and capping agent. Xanthan gum at very low

406

concentration of 1.5 mg/ml converted the ionic gold to neutral gold particles with a particle

407

size of 15-20 nm. Nanoparticles were found to be non-toxic and biocompatible in hemolysis

408

study. Doxorubicin was loaded to the nanoparticles through non-covalent interaction. The

409

prepared carbohydrate-rich gold nanoparticles showed high drug loading, good colloidal

410

stability and enhanced cytotoxicity in A549 lung cancer cells. High physical stability of gum 17 Page 17 of 36

stabilized nanoparticles can be attributed to steric repulsion among the particles provided by

412

same charge of gum moieties present on the nanoparticle surface. Receptor-mediated

413

endocytosis of nanoparticles could be a reason for better anticancer activity of DXGP than

414

free drug. Presence of mannose in xanthan gum might have contributed this effect.

415

Hence, natural gum based approach could be a better method for the synthesis of gold

416

nanoparticles.

cr

ip t

411

Acknowledgements

an

418

us

417

The research work was financially supported by Council of Scientific and Industrial

420

Research (CSIR) grant under project Advanced Drug Delivery (ADD-CSC 0302).

421

Doxorubicin hydrochloride was kindly provided by Therdose Pharma. HK thanks to the Head

422

of IICT-RMIT research centre for providing the Junior Research Fellowship. DP is awarded

423

Senior Research Fellowship by CSIR, New Delhi. Authors are also thankful to the Director,

424

CSIR-Indian Institute of Chemical Technology, Hyderabad for providing the necessary

425

facilities.

426

References

427

Almeida, J. P., Figueroa, E. R., & Drezek, R. A. (2013) Gold nanoparticle mediated cancer

428

immunotherapy. Nanomedicine: Nanotechnology, Biology and Medicine, doi:

d

te

Ac ce p

429

M

419

10.1016/j.nano.2013.09.011.

430

Aryal, S., Grailer, J. J., Pilla, S., Steeber, D. A., Gong, S. (2009). Doxorubicin conjugated

431

gold nanoparticles as water-soluble and pH-responsive anticancer drug nanocarriers.

432

Journal of Material Chemistry, 19, 7879-7884.

433

Basuki, J. S., Esser, L., Duong, H. T. T., Zhang, Q., Wilson, P., Whittaker, M. R.,

434

Haddleton, D. M., Boyer, C., & Davis, T. P. (2014). Magnetic nanoparticles with

18 Page 18 of 36

435

diblock glycopolymer shells give lectin concentration-dependent MRI signals and

436

selective cell uptake. Chemical Science, 5, 715-726. Burygin, G. L., Khlebtsov, B. N., Shantrokha, A. N., Dykman, L. A., Bogatyrev, V. A., &

438

Khlebtsov, N. G. (2009). On the Enhanced Antibacterial Activity of Antibiotics Mixed

439

with Gold Nanoparticles. Nanoscale Research Letters, 4(8), 794-801.

ip t

437

Carvalho, C., Santos, R. X., Cardoso, S., Correia, S., Oliveira, P. J., Santos, M. S., &

441

Moreira, P. I. (2009). Doxorubicin: the good, the bad and the ugly effect. Current

442

Medicinal Chemistry, 16(25), 3267-3285.

cr

440

Chen, Y. H., Tsai, C. Y., Huang, P. Y., Chang, M. Y., Cheng, P. C., Chou, C. H., Chen, D.

444

H., Wang, C. R., Shiau, A. L., Wu. C. L. (2007). Methotrexate conjugated to gold

445

nanoparticles inhibits tumor growth in a syngeneic lung tumor model. Molecular

446

Pharmaceutics, 4 (5), 713–722.

an

us

443

Comba, S., & Sethi, R. (2009). Stabilization of highly concentrated suspensions of iron

448

nanoparticles using shear-thinning gels of xanthan gum. Water Research, 43(15), 3717–

449

3726.

d

M

447

Comba, S., Dalmazzo, D., Santagata, E., & Sethi, R. (2011) Rheological characterization of

451

xanthan suspensions of nanoscale iron for injection in porous media. Journal of

452

Hazardous Matererials, 185(2–3), 598–605.

Ac ce p

te

450

453

Desplanques, S., Renou, F., Grisel, M., & Malhiac, C. (2012) Impact of chemical

454

composition of xanthan and acacia gums on the emulsification and stability of oil-in-

455

water emulsions. Food Hydrocolloids, 27, 401-410.

456

Dhar, S., Reddy, E. M., Shiras, A., Pokharkar, V., & Prasad, B. L. (2008). Natural gum

457

reduced/stabilized gold nanoparticles for drug delivery formulations. Chemistry- A

458

European Journal, 14(33), 10244-10250.

459

Duncan, B., Kim, C., & Rotello, V. M. (2010). Gold nanoparticle platforms as drug and

460

biomacromolecule delivery systems. Journal of Controlled Release, 148, 122–127.

461

El-Boubbou K, Zhu DC, Vasileiou C, Borhan B, Prosperi D, Li W, & Huang X. (2010).

462

Magnetic glyco-nanoparticles: a tool to detect, differentiate, and unlock the glyco-

19 Page 19 of 36

463

codes of cancer via magnetic resonance imaging. Journal of American Chemical

464

Society, 132(12), 4490-4499. Fan G, Cang L, Qin W, Zhou C, Gomes H. I., & Zhou D. (2013) Surfactants-enhanced

466

electrokinetic transport of xanthan gum stabilized nanoPd/Fe for the remediation of

467

PCBs contaminated soils. Separation and Purification Technology, 114, 64–72.

469

Gibson, J. D., Khanal, B. P., & Zubarev, E. R. (2007). Paclitaxel-functionalized gold nanoparticles. Journal of American Chemical Society, 129(37), 11653-11661.

cr

468

ip t

465

Gu, Y. J., Cheng, J., Man, C. W., Wong, W. T., & Cheng, S. H. (2012). Gold-doxorubicin

471

nanoconjugates for overcoming multidrug resistance. Nanomedicine: Nanotechnology,

472

Biology and Medicine, 8(2), 204-211.

us

470

Jian, H., Zhu, L., Zhang, W., Sun, D., & Jiang, J. (2012). Galactomannan (from Gleditsia

474

sinensis Lam.) and xanthan gum matrix tablets for controlled delivery of theophylline:

475

In vitro drug release and swelling behaviour. Carbohydrate Polymers, 87, 2176– 2182.

476

Jung, J., Park, S., Hong, S., Ha, M. W., Park, H. G., Park, Y., Lee, H. J., & Park, Y. (2013).

477

Synthesis of gold nanoparticles with glycosides: synthetic trends based on the structures

478

of

479

http://dx.doi.org/10.1016/j.carres.2013.12.012.

and

aglycones.

Carbohydrate

Research,

te

glycones

d

M

an

473

Khan, M. S., Vishakantea, D., & Siddaramaiah, H. (2013), Gold nanoparticles: A paradigm

481

shift in biomedical applications. Advances in Colloid and Interface Science, 199–200,

482 483 484 485

Ac ce p

480

44–58.

Kumar, K. P., Paul, W., & Sharma, C. P. (2011). Green synthesis of gold nanoparticles with Zingiber officinale extract: Characterization and blood compatibility.

Process

Biochemistry, 46, 2007–2013.

486

Laginha, K. M., Verwoert, S., Charrois, G. J. R., & Allen, T. M. (2005). Determination of

487

doxorubicin levels in whole tumor and tumor nuclei in murine breast cancer tumors.

488

Clinical Cancer Research, 11, 6944–6949.

489 490

Minati, L., Antonini, V., Torrengo, S., Serra, M. D., Boustta, M., Leclercq, X., Migliaresi, C., Vert, M., & Speranza, G. (2012).

Sustained in vitro release and cell uptake of

20 Page 20 of 36

491

doxorubicin adsorbed onto gold nanoparticles and covered by a polyelectrolyte

492

complex layer. International Journal of Pharmaceutics, 438(1-2), 45-52. Mirza, A. Z., & Shamshad, H. (2011). Preparation and characterization of doxorubicin

494

functionalized gold nanoparticles. European Journal of Medicinal Chemistry, 46(5),

495

1857-1860.

ip t

493

Oliveira, R., Zhao, P., Li, N., Maria, L. C. S., Vergnaud, J., Ruiz, J., Astruc, D., & Barratt, G.

497

(2013). Synthesis and in vitro studies of gold nanoparticles loaded with docetaxel.

498

Advances in Colloid and Interface Science, 199–200, 44–58.

cr

496

Phaechamud, T., & Ritthidej, G. C. (2007) Sustained-release from layered matrix system

500

comprising chitosan and xanthan gum. Drug Development and Industrial Pharmacy,

501

33, 595–605.

an

us

499

Pissuwan, D., Niidome, T., & Cortie, M. B. (2011). The forthcoming applications of gold

503

nanoparticles in drug and gene delivery systems. Journal of Controlled Release, 149(1),

504

65-71.

M

502

Prabaharan, M., Grailer, J. J., Pilla, S., Steeber, D. A., Gong, S. (2009). Gold nanoparticles

506

with a monolayer of doxorubicin-conjugated amphiphilic block copolymer for tumor-

507

targeted drug delivery. Biomaterials, 30(30), 6065-6075.

509

te

Rouhana, L. L., Jaber, J. A., & Schlenoff, J. B. (2007). Aggregation-resistant water-soluble

Ac ce p

508

d

505

gold nanoparticles. Langmuir, 23, 12799-12801.

510

Saha, B., Bhattacharya, J., Mukherjee, A., Ghosh, A. P., Santra, C. R., Dasgupta, A. K., &

511

Karmakar, P. (2007). In Vitro Structural and Functional Evaluation of Gold

512

Nanoparticles Conjugated Antibiotics. Nanoscale Research Letters, 2(12), 614–622.

513

Santos, H., Veiga, F., Pina, M. E., & Sousa, J. J. (2005). Compaction compression and drug

514

release properties of diclofenac sodium and ibuprofen pellets comprising xanthan gum

515

as a sustained release agent. International Journal of Pharmaceutics, 295(1–2), 15–27.

516

Sereno, N. M., Hill, S. E., & Mitchell, J. R. (2007). Impact of the extrusion process on

517

xanthan gum behaviour. Carbohydrate Research, 342(10), 1333-1342.

518

Sharma, B. R., Naresh, L., Dhuldhoya, N. C., Merchant, S. U., & Merchant, U. C. (2006)

519

Xanthan Gum - A Boon to Food Industry. Food promotion chronicle, 1(5), 27-30. 21 Page 21 of 36

520

Sinha, V. R., Mittal, B. R., Bhutani, K. K., & Kumaria, R. (2004). Colonic drug delivery of 5-

521

fluorouracil: An in vitro evaluation. International Journal of Pharmaceutics, 269(1),

522

101–108.

524

Teodora, M. (2013). Hemolysis as expression of nanoparticles-induced cytotoxicity in red blood cells. Biotechnology, Molecular Biology and Nanomedicine, 1(1), 7-12.

ip t

523

Tiraferri, A., Chen, K. L., Sethi R., & Elimelech, M. (2008). Reduced aggregation and

526

sedimentation of zero-valent iron nanoparticles in the presence of guar gum. Journal of

527

Colloid and Interface Science, 324(1–2), 71–79

cr

525

Vecchia, E. D., Luna, M., & Sethi, R. (2009). Transport in porous media of highly

529

concentrated iron micro- and nanoparticles in the presence of xanthan gum.

530

Environmental Science & Technology, 43(23), 8942-8947.

an

us

528

Xue, D., & Sethi, R. (2012). Viscoelastic gels of guar and xanthan gum mixtures provide

532

long-term stabilization of iron micro- and nanoparticles. Journal of Nanoparticle

533

Researc,14, 1239-1253.

M

531

Yang, W., Zhang, L., Wang, S., White, A. D., & Jiang, S. (2009). Functionalizable and

535

ultra stable nanoparticles coated with zwitterionic poly(carboxybetaine) in

536

undiluted blood serum. Biomaterials, 30:5617-5621.

538 539 540 541 542

te

Zhang, L., & Zhang, L. (2010). Lipid-polymer hybrib nanoparticles:synthesis,

Ac ce p

537

d

534

characterization and applications. Nano LIFE, 1, 163-173.

543 544 545 546 22 Page 22 of 36

547 548 549

ip t

550 551

cr

552

us

553 554

an

555

Figure legends

557 558 559

Fig. 1. Optimization of different formulation variables for the synthesis of Xanthan gum stabilized gold nanoparticles. Effect of a) Xanthan gum concentration b) gum volume c) gold volume and d) gold concentration

560 561

Fig. 2. Effect of temperature a) and reaction time b) in the synthesis of XG stabilized gold nanoparticles

562 563

Fig. 3. Transmission electron microscopy (TEM) image of DOX loaded Xanthan gum stabilized gold nanoparticles (DXGP)

564

Fig. 4. FTIR Spectra of a) pure Dox b) Native XG c) Blank GNP and d) Dox loaded GNP

565 566

Fig. 5. Differential scanning thermogram of a) pure DOX b) Dox loaded xanthan gum stabilized nanoparticles and c) Native xanthan gum

567 568

Fig. 6. In vitro drug release from DXNP in sodium acetate buffer pH 4.5 and phosphate buffer saline pH 7.4

569 570

Fig. 7. Percent cell viability of A549 human lung cancer cells after 48 h of treatment with free

571

Fig. 8. Storage stability of DXGP: a) particles diameter b) zeta potential and c) DOX content

572

Fig. 9. Effect of pH and ionic strength on the stability of DXGP

573 574

Fig. 10. Serum stability of DXGP a) particle size b) zeta potential c) absorption spectra in diluted (10%) serum and d) absorption spectra in undiluted (100%) serum

Ac ce p

te

d

M

556

DOX, BGNP and DXNP (Mean ± SD, n=3)

23 Page 23 of 36

575

Figures

577 579

ip t

581 583

cr

585 587

us

589 591

595 596

M

a)

597

606 608 610 612 614

te

604

Ac ce p

602

d

598 600

an

593

616 617 618

b)

619 620 24 Page 24 of 36

622 624 626

ip t

628 630

cr

632 634

us

636 638

642 643

M

c)

645

655 657 659 661 663

te

653

Ac ce p

651

d

647 649

an

640

664 665

d)

666 667 668

Fig. 1. Optimization of different formulation variables for the synthesis of Xanthan gum stabilized gold nanoparticles. Effect of a) Xanthan gum concentration b) gum volume c) gold volume and d) gold concentration

669 25 Page 25 of 36

671 673 675 677

ip t

679 681

cr

683

us

685 687

690

a)

M

691 692

702 704 706 708 710

te

700

Ac ce p

698

d

694 696

an

689

711 712

b)

713 714

Fig. 2. Effect of temperature a) and reaction time b) in the synthesis of XG stabilized gold nanoparticles

715 716 26 Page 26 of 36

ip t cr us

718 719

an

717

Fig. 3. Transmission electron microscopy (TEM) image of DOX loaded Xanthan gum stabilized gold nanoparticles (DXGP)

M

720 721

725 726

te

724

Ac ce p

723

d

722

27 Page 27 of 36

a) 911.53 870.17 1523.92

3525.46

1729.37

1212.41 1235.55

2931.91 3336.70 1617.36 1582.51

1413.68 1384.31

b)

466.13 583.32

969.48 1114.22 1072.51 1005.97

1285.36

991.01

2255.89 1984.06 2165.66 2079.45

789.59

2902.02 1728.49

1403.97 1279.69 1614.79 1051.82

cr

c)

ip t

611.81

3416.32

%T

688.27

761.54 804.62

653.41

1728.57

1254.28

1042.75

3398.11

d)

1384.48

us

1622.31

2025.45 1582.64 1615.61 2896.48

3525.48

an

1524.87

1730.21 3325.30

728 729

3600

3200

1413.94

2800

2400

2000

734 735 736 737 738

1200

1000

800

600

450.0

Fig. 4. FTIR Spectra of a) pure Dox b) Native XG c) Blank GNP and d) Dox loaded GNP

d te

733

1400

Ac ce p

732

1600

cm-1

730 731

1800

M

727

4000.0

2934.15

731.16 465.91 969.69 804.16 688.06 536.88 991.36 870.16 721.63 600.45 1005.37 761.60 583.10 1072.42 911.75 709.22 950.20 846.07 1114.33

1211.94 1235.28

739 740

28 Page 28 of 36

ip t cr us

741

743 744

an

742

Fig. 5. Differential scanning thermogram of a) pure DOX b) Dox loaded xanthan gum stabilized nanoparticles and c) Native xanthan gum

M

745 746

750 751 752 753

te

749

Ac ce p

748

d

747

29 Page 29 of 36

ip t cr us

754

Fig. 6. In vitro drug release from DXNP in sodium acetate buffer pH 4.5 and phosphate buffer saline pH 7.4

an

755 756

M

757 758

762 763 764 765 766 767

te

761

Ac ce p

760

d

759

768 769 770 771 772

30 Page 30 of 36

an

us

cr

ip t

773

774

Fig. 7. Percent cell viability of A549 human lung cancer cells after 48 h of treatment with free DOX, BGNP and DXNP (Mean ± SD, n=3)

M

775 776 777

781 782 783 784 785 786

te

780

Ac ce p

779

d

778

31 Page 31 of 36

ip t cr us

787 788

an

a)

790 791 792

Ac ce p

te

d

M

789

b)

32 Page 32 of 36

ip t cr us

793

c)

795

Fig. 8. Storage stability of DXGP: a) particles diameter b) zeta potential and c) DOX content

M

796

801 802 803 804 805

te

800

Ac ce p

799

d

797 798

an

794

33 Page 33 of 36

ip t cr us

807 808

a)

M

809

an

806

811

Ac ce p

te

d

810

812

b)

813

Fig. 9. a) Effect of pH and b) ionic strength on the stability of DXGP

814 815 816 34 Page 34 of 36

ip t cr us

817 818

an

a)

819

821 822 823

Ac ce p

te

d

M

820

b)

824 825 826 827

35 Page 35 of 36

ip t cr us

829

c)

830

832 833 834

Ac ce p

te

d

M

831

an

828

Fig. 10. Serum stability of DXGP a) particle size b) zeta potential c) absorption spectra in diluted (10%) serum and d) absorption spectra in undiluted (100%) serum

835 836 837 838

36 Page 36 of 36