Preparation of liposomes encapsulated Epirubicin-gold nanoparticles

0 downloads 0 Views 2MB Size Report
Apr 6, 2018 - Keywords: Epirubicin, drug delivery, liposomes, gold nanoparticles, .... range of biomedical applications in view of their stability, nanosize, bio ...
Biomedical Physics & Engineering Express

ACCEPTED MANUSCRIPT

Preparation of liposomes encapsulated Epirubicin-gold nanoparticles for Tumor specific delivery and release To cite this article before publication: Selvaraj Kunjiappan et al 2018 Biomed. Phys. Eng. Express in press https://doi.org/10.1088/20571976/aac9ec

Manuscript version: Accepted Manuscript Accepted Manuscript is “the version of the article accepted for publication including all changes made as a result of the peer review process, and which may also include the addition to the article by IOP Publishing of a header, an article ID, a cover sheet and/or an ‘Accepted Manuscript’ watermark, but excluding any other editing, typesetting or other changes made by IOP Publishing and/or its licensors” This Accepted Manuscript is © 2018 IOP Publishing Ltd.

During the embargo period (the 12 month period from the publication of the Version of Record of this article), the Accepted Manuscript is fully protected by copyright and cannot be reused or reposted elsewhere. As the Version of Record of this article is going to be / has been published on a subscription basis, this Accepted Manuscript is available for reuse under a CC BY-NC-ND 3.0 licence after the 12 month embargo period. After the embargo period, everyone is permitted to use copy and redistribute this article for non-commercial purposes only, provided that they adhere to all the terms of the licence https://creativecommons.org/licences/by-nc-nd/3.0 Although reasonable endeavours have been taken to obtain all necessary permissions from third parties to include their copyrighted content within this article, their full citation and copyright line may not be present in this Accepted Manuscript version. Before using any content from this article, please refer to the Version of Record on IOPscience once published for full citation and copyright details, as permissions will likely be required. All third party content is fully copyright protected, unless specifically stated otherwise in the figure caption in the Version of Record. View the article online for updates and enhancements.

This content was downloaded from IP address 130.64.11.153 on 04/06/2018 at 19:02

Page 1 of 35

Preparation of liposomes encapsulated Epirubicin-gold nanoparticles for Tumor specific

2

delivery and release

3

Selvaraj Kunjiappan1*, Theivendran Panneerselvam2, Balasubramanian Somasundaram1,

4

Sankarganesh Arunachalam1 Murugesan Sankaranarayanan3 and Pavadai Parasuraman4

us cri

5 6 7

1

8

Krishnankoil-626126, India

9

2

pt

1

Sir CV Raman-KS Krishnan International Research Center, Kalasalingam University,

an

Department of Pharmaceutical Chemistry, Karavali College of Pharmacy, Vamanjoor, Near

Reymond, Mangaluru-575028, India

11

3

Department of Pharmacy, Birla Institute of Technology and Science, Pilani-333031, India

12

4

Department of Pharmacy, M S Ramaiah University of Applied Sciences, Gnanagangothri

13

Campus, Bengaluru-560054, India.

dM

10

14 15 16

*

17

Dr. Selvaraj Kunjiappan,

18

Research Faculty,

19

Sir CV Raman-KS Krishnan International Research Center,

20

Kalasalingam University,

21

Krishnankoil-626126, India

22

Email: [email protected]

23

Ph: +919994972108

ce

pte

Address to correspondence

Ac

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

AUTHOR SUBMITTED MANUSCRIPT - BPEX-101073.R1

1

AUTHOR SUBMITTED MANUSCRIPT - BPEX-101073.R1

Abstract

25

Liposome/metal nanoparticle conjugates are found to be a promising carrier tool for effectively

26

delivering wide variety of drugs in a targeted site and without arguing toxic side effects of

27

existing anticancer drugs. In this scenario, the present work was performed to choose Epirubicin-

28

HCl (EPI) as model anticancer drug to formulate citrate stabilized EPI-gold nanoparticles (EPI-

29

GNPs). Further, the biocompatible EPI-GNPs incorporated with lipid bilayer of liposomes

30

formulated to exhibit effective drug delivery system. The biosynthesized EPI-GNPs and its

31

incorporation in a lipid bilayer of liposomes was confirmed by UV-visible spectrophotometer,

32

drug encapsulation, loading capacity, pH stability and drug releasing capacity were studied.

33

Simultaneously, drug and carrier interaction, physical nature, stability, morphological and

34

particle size characteristics were also studied by FTIR, XRD, SEM, TEM and Zeta size analyzer.

35

The obtained characterization data confirmed the properties of EPI-GNPs encapsulated

36

liposomes. The in vitro cytotoxic screening of EPI-GNPs encapsulated liposomes arrest the

37

proliferation of MCF-7 breast cancer cell lines, at 0.1 µg/mL of liposomes displayed 52% of cell

38

death at 24 h. The EPI-GNPs encapsulated liposomes induced time and dose dependent apoptosis

39

compared with untreated control cancer cells.

42 43 44 45

us cri

an

dM

pte

41

Keywords: Epirubicin, drug delivery, liposomes, gold nanoparticles, breast cancer, cytotoxicity

ce

40

pt

24

Ac

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 2 of 35

46

2

Page 3 of 35

1. Introduction

48

Breast cancer is the most widely recognized malignancy in females and it is a leading cause for

49

death rate in women (14% of all cancer deaths) worldwide [1]. However, cancer cells proliferate

50

at much faster rate than the normal cells. Breast cancer tends to spread metastasis primarily to

51

lymph nodes, liver, lungs, bones and brain [2]. Therefore, breast cancer has become major health

52

concern and proper treatment of breast cancer has nowadays become a necessity. Recently, a

53

number of recognized chemotherapeutic agents are in market for effective breast cancer therapy,

54

but unfortunately, they have inability to selectively deliver the drugs to tumor targets, shorter life

55

time and aggressively producing undesirable toxic side effects to normal tissues [3]. Therefore,

56

there is a pressing need to develop alternative way of drug delivery system to deliver a sufficient

57

dose of drug to the disease location with less invasive but more efficient cancer treatments by the

58

currently available chemotherapeutic agents. The most promising drug delivery system would be

59

to exploit nanotechnology based biodegradable carrier material to explore the treatment of cancer

60

at its molecular level. Presently polymers, proteins, liposomes, solid lipid nanoparticles, and

61

metal nanoparticle-conjugated biodegradable systems are helpful for therapeutic purposes [4].

62

Among the above carrier systems, liposomes with metal nanoparticle conjugate have many

63

advantages in the field of drug delivery systems because of their nanosize, high surface to

64

volume ratio, less severe adverse reaction with enhanced cancer treatment [5]. Liposomes are the

65

simplest artificial biological cell, and are well known to reduce toxicity related with many

66

compounds including antitumor drugs, antiviral drugs and antibiotics [6]. Further, it is currently

67

being introduced into clinical use and several more are under clinical trials or pre-clinical

68

development [7]. Liposomes are submicronic spherical vesicular structure consisting of mono or

69

bilayer of phospholipids/cholesterol having a hydrophobic tail and a hydrophilic head, which can

ce

pte

dM

an

us cri

pt

47

Ac

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

AUTHOR SUBMITTED MANUSCRIPT - BPEX-101073.R1

3

AUTHOR SUBMITTED MANUSCRIPT - BPEX-101073.R1

be encapsulated with both hydrophilic and hydrophobic drugs depending on the preparation

71

method [8].

72

In the recent decade, gold nanoparticles (GNPs) have been remarkably being used in a wide

73

range of biomedical applications in view of their stability, nanosize, bio compatibility and have

74

displayed unique and tunable surface plasmon resonance (SPR) [9]. These points of interest

75

empower the GNPs to target tumor all the more effectively without severe accumulation in

76

reticuloendothelial system, making them exceptionally encouraging for cancer diagnosis and

77

treatment [10, 11]. Further, GNPs have demonstrated high affinities with highly selective

78

targeting properties to either tissues or cells and passively aggregate at tumor sites because of

79

upgraded permeability and retention (EPR) effects [12]. It is also flexible to surface functionalize

80

with various biomolecules such as protein, DNA, algae, enzymes, and plant-derived bioactive

81

compounds [13, 14]. Therefore, GNPs are currently the subject of exceptional research for

82

potential clinical applications.

83

The hybrid GNPs/liposomal conjugate can be prepared by GNPs encapsulation or surface

84

coating with lipids, which is a useful non-covalent approach to stabilize surface chemistry for

85

prolonged plasma circulation improving pharmacokinetic profile and reducing clearance by

86

circumventing reticuloendothelial system (RES) uptake [15]. Epirubicin-HCl (EPI) is an

87

anthracycline antineoplastic agent that acts by DNA intercalation and inhibiting topoisomerase

88

II, which further interfere with DNA, RNA and protein synthesis [16]. EPI is a 4’-epimer of

89

Doxorubicin (DOX) and differs in the orientation of the C-4 hydroxyl group on the sugar that has

90

several beneficial effects over DOX including improved efficacy, lower toxicity, rapid clearance

91

from plasma resulting in less cumulative dose, and thus can successfully replace DOX in breast

92

cancer treatment [17, 18]. But the use of free EPI is rather limited due to the severe side effects

ce

pte

dM

an

us cri

pt

70

Ac

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 4 of 35

4

Page 5 of 35

and multidrug resistance [19]. Hence, the investigation reported here is aimed to enhance the

94

therapeutic effects of EPI and reduce its toxic side effects by synthesizing GNPs and

95

encapsulating them in a lipid bilayer on nanoparticles. In the present study, a novel strategy was

96

employed to exploit the use of sodium citrate dihyrate as a reducing and stabilizing agent for the

97

biosynthesis of GNPs and subsequently attached with EPI. The stabilized EPI-GNPs were

98

encapsulated in liposomes by conventional thin film hydration method. Then, the formulated

99

GNPs/liposomal conjugate were characterized by analytical techniques to evaluate their size,

100

size-distribution, morphology, stability, encapsulation efficiency, and drug releasing capacity.

101

Further, in vitro anticancer activity was evaluated against breast cancer cell lines (MCF-7).

102

2. Materials and Methods

103

2.1. Materials

104

The chemicals, Hydrogen tetracholoroaureate (III) hydrates (HAuCl4.3H2O (99.9%)), Sodium

105

citrate dihydrate (99%), 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-diphenyltetrazolium bromide (MTT),

106

fetal bovine serum (FBS), Dichloro-dihydro-fluorescein diacetate (DCFH-DA), Dulbecco’s

107

Modified Eagles Medium (DMEM) and the solvents, analytical grade ethanol (95%), chloroform

108

(98%) and toluene (99.7%) were procured from Himedia Laboratories Pvt., Ltd., Mumbai, India.

109

Distearoyl phosphatidylcholine (DSPC) with >99% purity was obtained from Lipoid (Germany)

110

and used without further purification. Cholesterol was purchased from Sigma Aldrich Company

111

(St. Louis, USA). All other chemicals and solvents used in this study were of analytical grade

112

and obtained from Merck, Mumbai, India. Epirubicin-HCl (4′-epidoxorubicin hydrochloride;

113

(EPI)) was received as kind gift from Alkem Laboratories Ltd., Mumbai, India. Glassware were

114

washed with dilute nitric acid and thoroughly rinsed with double distilled water and dried in hot

ce

pte

dM

an

us cri

pt

93

Ac

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

AUTHOR SUBMITTED MANUSCRIPT - BPEX-101073.R1

5

AUTHOR SUBMITTED MANUSCRIPT - BPEX-101073.R1

air oven with before use. MCF-7 cell line was obtained from National Centre for Cell Science

116

(NCCS, Pune, India).

117

2.2. Synthesis of GNPs

118

Citrate-stabilized GNPs were synthesized by a modified procedure similar to that of Frens, 1973

119

[20]. In short, 10 mL of sodium citrate di hydrate (0.01 M) was added to 90 mL of aqueous

120

solution of 1 × 10−4 M HAuCl4∙3H2O and the solution was kept under magnetic stirring at 60°C

121

for 30 min. This procedure results in a ruby red solution containing monodisperse GNPs. The

122

formation of GNPs was monitored with UV-visible spectrophotometer. GNPs were obtained by

123

centrifugation at 12000 × g for 30 min and washed several times with de-ionized water by

124

centrifugation and re-dispersion in water to remove excess gold.

125

2.3. Preparation of EPI-GNPs

126

2 mg of EPI was added to GNPs dispersion, obtained as described above, resulting in a final EPI

127

concentration of 10−4 M in solution. The mixture of EPI and GNPs dispersion was incubated for

128

24 h at room temperature and then centrifuged at 10000 × g for 15 min. The obtained pellet after

129

centrifugation was separated from the supernatant and re-dispersed in de-ionized water prior to

130

further characterization.

131

2.4. Preparation of Liposome encapsulated EPI-GNPs

132

Liposomes encapsulating EPI-GNPs were formulated by the method of Rengan et al. 2014 [21].

133

Briefly, a mixture of lipids DSPC and cholesterol were dissolved in a mixture of chloroform:

134

methanol (2:1), followed by removal of chloroform and methanol using a rotary evaporator

135

(Buchi rotavap). The EPI-GNPs solution (1.0 mL) was then added to the dried cationic lipid film

136

and the mixture was incubated at room temperature for 4 h with intermittent mixing, resulting in

137

a final lipid concentration of 10 mM. To achieve a homogeneous mixture of liposomes, the

ce

pte

dM

an

us cri

pt

115

Ac

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 35

6

Page 7 of 35

suspension was sonicated for 5 min. Liposomes were annealed for 1 h at 60 °C in a water bath

139

and cooled to room temperature.

140

2.5. Determination of drug encapsulation efficiency and loading capacity

141

The EPI encapsulation and loading capacity of the formulated liposomes was measured

142

according to the method described by David et al. 2015 [22]. In short, EPI concentration in the

143

supernatant after the centrifugation of the formulated liposomes solution was detected using the

144

UV-visible spectrophotometer. The resultant supernatant was examined under UV-Visible

145

spectrophotometer which yielded an absorbance at 490 nm for EPI. Calibrated standard plots and

146

encapsulation efficiency and drug loading capacity were estimated using the absorbance values

147

and converted to quantity of EPI using the equation as given below:

148

Encapsulation efficiency (%)= (Total drug - Free drug)/Total drug × 100

149

Drug Loading capacity (%)= (Total drug - Free drug)/Nanoparticle weight × 100

150

2.6. In vitro drug release study

151

In vitro drug release characteristics of liposomes was studied by dialysis method [23]. In short,

152

free drug (EPI) was separated from liposomes (50 mg) by passing through a sephadex column of

153

dialysis bag and the dialysis bag was then immersed in beaker containing 10 mL of PBS (0.01

154

M, pH 7.4) or 10 mL of PBS (0.01 M, pH 5.0) and was subjected to separation after

155

centrifugation. After completion of experiment (as the experiment was performed in duplicate),

156

the liposomal suspension was equally divided into 10 mL and transferred to two tubes kept in a

157

shaker at 150 rpm and temperature 37 °C. Samples were withdrawn at scheduled time points,

158

the released medium was completely removed and replaced with an equal volume of fresh

159

medium. DMSO was used to dissolve the collected resultant supernatant, further the

ce

pte

dM

an

us cri

pt

138

Ac

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

AUTHOR SUBMITTED MANUSCRIPT - BPEX-101073.R1

7

AUTHOR SUBMITTED MANUSCRIPT - BPEX-101073.R1

centrifugation of this solution at 14000 × g at 25 °C ensured collection of the drugs which was

161

used for UV-visible spectrophotometric analysis.

162

%DRC = (A0 - A1) × 100/A0

163

Where, A0 = absorbance of the control; A1 = absorbance of the sample.

164

2.7. Stability studies

165

In vitro stability of EPI-GNPs encapsulated liposomes were evaluated in various physiological

166

medium (10% NaCl, 0.5% BSA) and in phosphate buffer solutions at room temperature with

167

different pH values of 1.5, 3, 5, 6, 7.4 and 9 in order to investigate pH responsive behavior of

168

liposomes. Briefly, 1 mL of EPI-GNPs encapsulated liposomes suspension was mixed with 1 mL

169

each of the physiological medium and buffer solutions and its maximum absorbance was

170

recorded after 24 h using the UV-Visible spectrophotometer.

171

2.8. Physicochemical properties of EPI-GNPs encapsulated liposomes

172

2.8.1. Fourier transform infrared spectroscopy (FTIR)

173

Infrared spectra of free EPI, EPI-GNPs and EPI-GNPs encapsulated liposomes were recorded on

174

Shimadzu IR Tracer-100, FTIR spectrophotometer. A small amount (≈1) of free EPI, EPI-GNPs

175

and EPI-GNPs encapsulated liposomes (lyophilized powder) were mixed with IR grade

176

Potassium Bromide (KBr) to prepare a round disc using a small hydraulic press to study FTIR

177

characteristic spectrum in the spectral range of 4000-400 cm-1 with resolution of 4 cm-1.

178

2.8.2. Powder X-ray Diffraction analysis

179

The purity and crystalline properties of plain EPI, EPI-GNPs and EPI-GNPs encapsulated

180

liposomes were studied by Powder XRD. The XRD scanning was done at a voltage of 20 keV

181

and a current of 30 mA with Cu Kα 1 radiation (l = 0.1542) in a two-theta (degree) configuration

182

by using A BRUKER D 8 Advance ECO XRD system with SSD160 1 D Detector.

ce

pte

dM

an

us cri

pt

160

Ac

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 35

8

Page 9 of 35

2.8.3. Particle characterization

184

The surface charge and sizes of the freshly prepared EPI-GNPs encapsulated liposomes were

185

measured by Zetasizer Nano ZS ver.7.03 (Malvern Instrument, Malvern, Worcestershire, United

186

Kingdom).

187

2.8.4. SEM analysis

188

Scanning Electron microscopic study was carried out to study and compare the morphological

189

characteristics of EPI-GNPs and EPI-GNPs encapsulated liposomes. A sample of EPI-GNPs and

190

EPI-GNPs encapsulated liposomes were placed in a 200 mesh copper grid and observed it for

191

particles shape and size using SEM detector attached to scanning electron microscopy (Carl

192

Zeiss EVO 18).

193

2.8.5. TEM analysis

194

The morphological shape and size of EPI-GNPs and EPI-GNPs encapsulated liposomes were

195

studied using transmission electron microscope (TEM-JEOL model 2100) operated at an

196

acceleration voltage of 200 KV. For this purpose, EPI-GNPs encapsulated liposomes was re-

197

dispersed with 1 mL distilled water. Few drops of dispersed nanoparticles in suspension was

198

placed on the carbon coated copper grid, which was air-dried at 60 °C for 5 min.

199

2.9. Cytotoxicity studies

200

The potential cytotoxicity of EPI-GNPs encapsulated liposomes was assessed by MTT assay

201

[24]. Shortly, breast cancer cell lines (MCF-7 cell) were seeded in to 96-multiwell plate at

202

approximately 1 × 105 cells/well at 37 °C in a 5% CO2 atmosphere for 24 h before treatment,

203

allowing for cell attachment to the wall of the plate. Cells were treated with different

204

concentrations of EPI-GNPs encapsulated liposomes (0.003, 0.006, 0.01, 0.05, 0.1, 0.2 and 0.3

205

μg/mL) for 24 h incubation. The respective amounts of DMSO in PBS buffer instead of EPI-

ce

pte

dM

an

us cri

pt

183

Ac

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

AUTHOR SUBMITTED MANUSCRIPT - BPEX-101073.R1

9

AUTHOR SUBMITTED MANUSCRIPT - BPEX-101073.R1

GNPs encapsulated liposomes was used as control. The medium was removed after 24 h of

207

treatment and cells were washed with phosphate-buffered saline (PBS, 0.01 M, pH 7.4). This

208

was followed by addition of 200 μL (5 mg/mL) of 0.5% 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-

209

diphenyl-tetrazolium bromide (MTT) prepared in serum free medium solution to each well and

210

incubated for 4 h at 37 °C in a 5% CO2. Thus, the obtained cells were fixed, washed and stained

211

with MTT. Acetic acid was used to remove excess stain while Tris-EDTA buffer was used to

212

remove any attached cells. The color intensity was measured in a microplate reader at 564 nm.

213

The ratio of the absorption of treated cells to absorption of non-treated cells expressed in

214

percentage resulted in percentage of death cells. Thus, every treatment condition was repeated in

215

triplicate and IC50 of EPI-GNPs encapsulated liposomes obtained were used for treatment for all

216

further studies.

217

2.10. Analysis of apoptotic cells by Acridine Orange/Ethidium bromide (AO/EB) double

an

us cri

staining

dM

218

pt

206

Apoptosis of the cancer cells were investigated by fluorescence microscopy after staining with

220

AO/EB [25]. Briefly, MCF-7 cancer cells were seeded on a cover slip in a 24-well plate (1×105

221

cells/well), and addition of appropriate amount of EPI-GNPs encapsulated liposomes of IC50

222

value and incubated. After being cultured for 72 h, 20 μL of trypsin was added to each well.

223

Cover slip removed from 24-well plate and washed with 1×PBS buffer and treated with dual

224

fluorescent stain 10 µL/mL containing acridine orange (10 mg/mL) and ethidium bromide (10

225

mg/mL) and incubated again 30 min. After incubation, unbound dyes were washed with 1 × PBS

226

buffer. The morphology of apoptotic cells was examined under fluorescence microscope and

227

representative fields were captured at 40× magnification. Dual AO/EB staining method was

228

repeated 3 times at least.

ce

pte

219

Ac

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 35

10

Page 11 of 35

2.11. Apoptotic analysis by Annexin V-FITC and Propidium Iodide

230

Cell apoptotic activity of EPI-GNPs encapsulated liposomes was assessed by Nouri and

231

Yazdanparast (2011) method with slight modification [26]. MCF-7 cancer cells were treated with

232

EPI-GNPs encapsulated liposomes (IC50 value) for 24 h and 48 h. After stipulated time of

233

treatment, the cells were harvested and washed twice with 1 × PBS and re-suspended in 100 µL

234

binding buffer (10 mM HEPES/NaOH of pH 7.4, 140 mM NaCl and 2.5 mM CaCl2). Next,

235

Annexin V-FITC (10 µL) was added to the cells followed by the addition of 10 µL of PI (50

236

µg/mL in PBS). The samples were incubated for 10 min in the dark at 4 °C and then analyzed by

237

flow cytometry.

238

2.12. ROS generation assay

239

Intracellular ROS generation in control and treated MCF-7 cancer cells were monitored by

240

measuring changes in fluorescence resulting from intracellular probe oxidation [27]. The cancer

241

cells seeded in 6-well culture plate (1×105 cells/well) and incubated for 24h. After incubation,

242

the medium was replaced with 10% FBS supplemented with IC50 value of EPI-GNPs

243

encapsulated liposomes, and again incubated for 24 and 48 h. The cells were washed twice with

244

1× PBS and 20 μM DCFH-DA was added and incubated for another 10 min. The DCFH-DA was

245

removed, and then 100 μM H2O2 was added into the cells and incubated for 45 min. The change

246

in the fluorescence was monitored by fluorescence spectrophotometer at λex=475 nm, λem=525

247

nm.

248

2.13. Statistical analysis

249

Experiments for each sample were performed three times, and obtained values were presented as

250

mean ± standard deviation (SD). Data were analyzed by one-way analysis of variance (ANOVA)

251

and Student-t test. A value of p