Liposome Encapsulation of Oncolytic Virus M1 To Reduce

Oncolytic viral therapy is an attractive novel strategy for cancer therapy. As a natural alphavirus, oncolytic virus M1 is able to infect and kill var...
0 downloads 0 Views 2MB Size
Subscriber access provided by Iowa State University | Library

Article

Liposome encapsulation of oncolytic virus M1 to reduce immunogenicity and immune clearance in vivo Yalong Wang, Huizhi Huang, Haijuan Zou, Xuyan Tian, jun hu, Pengxin Qiu, Haiyan Hu, and Guangmei Yan Mol. Pharmaceutics, Just Accepted Manuscript • DOI: 10.1021/acs.molpharmaceut.8b01046 • Publication Date (Web): 03 Jan 2019 Downloaded from http://pubs.acs.org on January 3, 2019

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 30 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

Molecular Pharmaceutics

1

Liposome encapsulation of oncolytic virus M1 to

2

reduce immunogenicity and immune clearance in

3

vivo

4

Yalong Wang,†,# Huizhi Huang,†,# Haijuan Zou,†,§ Xuyan Tian,‡ Jun Hu,‡ Pengxin Qiu,‡ Haiyan

5

Hu,†,* and Guangmei Yan‡

6



7

Guangzhou Higher Education Mega Center, Guangzhou 510006, P.R. China

8



9

Zhongshan II Road 74, Guangzhou 510080, P.R. China

School of Pharmaceutical Sciences, Sun Yat-sen University, Waihuan East Road 132,

Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University,

10

§

Department of Pharmacy, the Seventh Affiliated Hospital of Sun Yat-sen University,

11

Shenzhen 518107, P.R. China

12

KEYWORDS: Oncolytic virus M1; Liposome; Immune clearance; Immunogenicity

13 14 15 16

ACS Paragon Plus Environment

1

Molecular Pharmaceutics 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 30

17 18

Abstract Graphic

19

ABSTRACT: Oncolytic viral therapy is an attractive novel strategy for cancer therapy. As a

20

natural alphavirus, oncolytic virus M1 is able to infect and kill various zinc finger antiviral protein

21

(ZAP)-deficient tumor cells selectively, while leaving normal cells undamaged. However, M1 can

22

trigger the production of neutralizing antibodies that dramatically weaken its antitumor effect. In

23

order to attenuate immunogenicity of therapeutic M1 virus, we encapsulated it into liposomes

24

(referred to as M-LPO) using the thin-film hydration method. The effect of anti-M1 neutralizing

25

antibody on M-LPO was examined in LoVo and Hep 3B cell lines. In the absence of neutralizing

26

antibodies, treating cells with naked M1, blank liposomes (LPO), M-LPO or a simple mixture of

27

M1 and liposomes (LPO+M1) inhibited cell growth. In the presence of neutralizing antibodies,

28

only M-LPO inhibited cell growth. After intravenous administration, M-LPO reduced the

29

production of M1-neutralizing antibody and the corresponding immune response. Analysis of M-

30

LPO uptake by cells was examined by confocal microscopy using M1 labelled with FITC and

31

liposomal shells labelled with RhB. The results suggest that M1 may be released from liposomes

32

before or after M-LPO internalization. Taken together, our results suggest that encapsulating

33

oncolytic virus M1 in liposomes may reduce intrinsic viral immunogenicity for improved

34

anticancer therapy.

35

ACS Paragon Plus Environment

2

Page 3 of 30 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

Molecular Pharmaceutics

36

INTRODUCTION

37

Viral therapy is quite promising for the treatment of cancer when the virus selectively replicates

38

in tumor cells rather than normal cells, which significantly improves the safety and efficacy of

39

tumor therapy.1-3 In effect, this amounts to selective targeting of tumor cells. Examples of viruses

40

that naturally target cancer cells are autonomously replicating H1 parvovirus,4 reovirus,5, 6 and

41

Newcastle disease virus.7, 8 Even more viruses can be adapted or engineered to target cancer cells,

42

including adenovirus from which the E1B-55K gene has been knocked out9, 10 herpes simplex virus

43

from which the ICP 34.5 gene has been knocked out11, 12 and vesicular stomatitis virus carrying a

44

modification in the IFN-β gene.13

45

Naturally occurring M1, a Getah-like alphavirus isolated from culicine mosquitoes and abundant

46

on the Chinese island of Hainan,14 may be promising for selective targeting of cancer cells. Its

47

significant antitumor activity15 is selective for cancer cells deficient in zinc-finger antiviral

48

proteins (ZAPs), including Hep 3B cells, LoVo cells, C-33A cells, and Huh-7 cells16. In addition,

49

compounds that mimic the mitochondria-derived activator of caspases potentiate the selective

50

oncolytic effect of M1 in cancer cells, enhancing M1 replication and the bystander killing effect.17

51

However, the use of M1 for anti-cancer therapy is inhibited by its high immunogenicity:

52

intravenous administration triggers production of neutralizing antibodies, leading to a strong

53

immune response that inactivates the virus and eliminates the therapeutic effect18, 19 To counteract

54

this inactivation, researchers have encapsulated oncolytic viruses in fragments of plasma

55

membrane.20-23 Alternatively, oncolytic virus can be wrapped in liposomes, which are attractive

56

because they offer a large hydrophilic lumen for packaging diverse cargo and protecting it from

57

cellular and humoral responses.24-27 To date, studies of liposomal encapsulation of such viruses

ACS Paragon Plus Environment

3

Molecular Pharmaceutics 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 30

58

have been limited to non-enveloped adenovirus. It is unclear whether this strategy can work for

59

M1, which is already enclosed within a phospholipid layer.

60

In this study, liposome-encapsulated M1 (M-LPO) were prepared to protect M1 from neutralizing

61

antibodies in vitro as well as reduce M1 immunogenicity in vivo. The physicochemical and

62

morphological properties of M-LPO were investigated. Moreover, the effects of anti-M1

63

neutralizing antibody on M-LPO and naked M1 were examined in LoVo and Hep 3B cell lines in

64

vitro. And the antigenicity of M-LPO, compared to M1, was evaluated after intravenous

65

administration. Further, analysis of M-LPO uptake by cells was performed by confocal microscopy

66

using M1 labelled with FITC and liposomal shells labelled with RhB.

67

MATERIALS AND METHODS

68

Materials. Cell lines were purchased from the American Type Culture Collection, the Shanghai

69

Institute of Cell Biology, and the Guangzhou Institute of Biomedicine and Health. Oncolytic virus

70

M1 was provided by Guangmei Yan research group of Sun Yat-sen University (Guangzhou,

71

China). Soybean lecithin (S100) and egg yolk lecithin (PC-98T) was obtained from Lipoid GmbH

72

(Germany) and A.V.T. (Shanghai, China) respectively. Cholesterol was purchased from

73

Guangzhou QiHua Medical Equipment Co., Ltd (Guangzhou, China). Cells were cultured in

74

DMEM supplemented with 10% (v/v) FBS and 1% penicillin/streptomycin (Life Technologies).

75

M1 was grown in Vero cells in SFM (Life Technologies). Virus titer in plaque-forming units (pfu)

76

was determined by TCID50 assay using BHK-21 cells. Rhodamine DHPE was purchased from

77

Life Technologies, and FITC was purchased from MP Biomedicals.

ACS Paragon Plus Environment

4

Page 5 of 30 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

Molecular Pharmaceutics

78

Preparation of liposome-encapsulated M1. M1 was purified by ultrafiltration in a centrifuge

79

column with molecular weight cut-off of 50 kDa with three spins of 20 min at 2500 rpm. Then M1

80

was diluted in 0.1M phosphate-buffered saline (PBS, pH 7.4) to a suitable titer.

81

Liposome-encapsulated M1 was prepared using the thin-film hydration method.28 Soybean lecithin

82

and cholesterol (4:1, w/w) were dissolved in mixed organic solvent [CH2Cl2 : ethyl alcohol (1:2,

83

v/v)]. Organic solvent was removed by vacuum rotary evaporation at 100 rpm at 45 ℃, yielding a

84

dry lipid film. The film was hydrated using 1  108 pfu/mL M1 in 0.1 M PBS (pH 7.4) and vortexed

85

for 5 min at room temperature. The suspension was extruded 15 times through polycarbonate

86

membranes of 200 nm pore size (Millipore, Bedford, MA, USA) and then passed through a

87

Cellufine Sulfate column to separate out naked M1. The isolated M-LPO was sterile-filtered

88

through a 0.45 m membrane (Millipore). In addition, M-LPO prepared from egg yolk lecithin

89

was also studied. The separation operation for each sample was repeated three times for the titer

90

detection.

91

As a negative control, blank liposomes (LPO) were prepared as above, except that the films were

92

hydrated in PBS without M1. Besides the thin-film hydration method, two other methods had been

93

developed and compared. They are calcium-induced phase change method29 and co-incubation

94

method. The detailed preparation methods could been seen in the Supporting Information (Section

95

2).

96

Characterization. Samples of LPO, M-LPO or M1 were transferred to a clean hydrophobic

97

surface, and a copper grid was carefully inverted onto the drop and allowed to soak for 90 s. The

98

excess liquid was removed with filter paper, and the grid was negatively stained for 90 s with 1%

ACS Paragon Plus Environment

5

Molecular Pharmaceutics 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

99

Page 6 of 30

phosphotungstic acid. Grids were air-dried and then observed under a JEM-2100F transmission

100

electron microscopy (TEM) (JEOL, Japan).

101

Average size and zeta potential of M1, LPO and M-LPO were measured using a laser-based

102

analyzer (Zeta sizer Nano ZS90, Malvern, UK).

103

Encapsulation efficiency (EE) of M-LPO was calculated using the formula EE% = Wa / Wtotal 

104

100%, where Wa and Wtotal are the amount of protein in M-LPO after or before passage through

105

the Cellufine Sulfate column, respectively. Protein concentration was estimated using through the

106

microBCA kit (120 mL, Cwbiotech, China) based on absorbance at 570 nm. The detailed

107

determination process of encapsulation efficiency could be found in Supporting Information

108

(Section 3).

109

Cell viability assay. Cells were seeded into 96-well plates at a density of 5 × 103 cells per well

110

and then incubated for 24 h in an atmosphere of 5% CO2 and 95% air at 37 ℃. The medium was

111

replaced with DMEM containing varying concentrations of M1, LPO, a simple mixture of LPO

112

and M1 (LPO+M1), or M-LPO. At 4 h later, the medium was replaced with complete DMEM and

113

the cells were incubated for another 48 h. Surviving cells were quantified using the colorimetric

114

MTT assay based on the formula Survival % = (A490 for the treated cells/A490 for the control cells)

115

× 100%, where A490 is the absorbance at 490 nm. Tests were performed in triplicate.

116

Preparation of M1-neutralizing antibody. Sprague-Dawley rats were injected intravenously

117

with 5  107 pfu M1 once every day for 3 days in the tail, then they received a second intravenous

118

injection with 5  107 pfu M1 in the same way. At 7 days after the second immunization, blood

119

was taken, allowed to sit for 30 min, and centrifuged at 3000 rpm for 10 min. The serum

120

supernatant containing M1-neutralizing antibody was stored at -20 ℃. Besides, the serum was

ACS Paragon Plus Environment

6

Page 7 of 30 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

Molecular Pharmaceutics

121

repackaged and stored at -20 ℃, thus avoiding the repeatedly freeze-thaw resulting in the

122

inactivation of neutralizing antibody.

123

Virus neutralization in vitro. Appropriate dilutions of neutralizing antibody were determined

124

using the method of fixed virus dilution. Neutralizing antibody was diluted 64, 128, 256, and 512

125

times in DMEM. These dilutions were mixed with the same volume of M1 at multiplicity of

126

infection (MOI) of 0.1, and the mixture was incubated at 37 ℃ for 1 h. The mixture was added to

127

LoVo cells, and viability was measured. The negative control (NCtrl) cells were treated with

128

DMEM only; the positive control (PCtrl) cells, with M1 at MOI 0.1; and the treated control (TCtrl)

129

cells, with M1 at MOI 0.1 that had been incubated with DMEM at 37 ℃ for 1 h.

130

Immune neutralization in vitro. DMEM, M1, LPO, LPO+M1 or M-LPO was mixed with

131

neutralizing antibody and incubated at 37 ℃ for 1 h to achieve complete immunity. Then the

132

mixtures were added to LoVo or Hep 3B cultures and cell viability was measured. As a negative

133

control, cells were exposed to M1, LPO, LPO+M1 or M-LPO without pre-incubation with

134

neutralizing antibody. M1 was present at an MOI of 0.1 in cultures exposed to virus.

135

Antigenicity of M-LPO in vivo. Six-week-old female Balb/c mice were randomly divided into

136

four groups of six mice each, then immunized intravenously via tail injection with saline, M1, LPO

137

or M-LPO. One week later, the mice received a second tail injection of the same dose. Two weeks

138

after the second injection, blood was taken, allowed to rest for 30 min, and centrifuged at 3000

139

rpm for 10 min. The supernatant was mixed with an equal volume of naked M1 and incubated at

140

37 ℃ for 1 h. M1 titer was then determined.

ACS Paragon Plus Environment

7

Molecular Pharmaceutics 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 30

141

Uptake of M-LPO by LoVo cells. Uptake of M-LPO was analyzed by confocal microscopy using

142

doubly labelled M-LPO with M1 labelled with FITC and liposomal shells labelled with Rhodamine

143

DHPE (FM1-RhLPO).

144

To prepare FITC-labelled M1 (FM1), M1 was mixed with an equal volume of FITC solution (0.1

145

mg/ml) in 0.5 M bicarbonate buffer (pH 9.0) and left at room temperature for 1 h in the dark. The

146

mixture was passed through a Sephadex G-50 column washed in PBS to remove unconjugated

147

FITC. The effluent fraction containing labelled virus was collected and sterile-filtered through a

148

0.45 m Millipore filter.30, 31

149

During production of the thin film for liposome preparation, the 1% soybean lecithin was replaced

150

by Rhodamine DHPE, and the film was hydrated using FM1. In this way, doubly labelled FM1-

151

RhLPO was prepared.

152

Localization of FM1-RhLPO after uptake into LoVo cells was analyzed using laser scanning

153

confocal microscopy (LSM710, Zeiss, Germany). LoVo cells were seeded into confocal dishes

154

(catalog no. 801002, 15 mm diameter, Nest) at a density of 1 × 105 cells per well and incubated

155

for 24 h. Then cells were treated for 4 h with FM1 or FM1-RhLPO with M1 at an MOI of 0.1.

156

Negative control cells were treated with DMEM. Cells were washed six times in PBS, fixed for

157

30 min in 4% paraformaldehyde, stained for 25 min with DAPI (10 g /mL) and washed three

158

times in PBS. Free DAPI was washed away using PBS, then fluorescence images were observed

159

under a laser scanning confocal microscope.

160

Statistical analysis. All data are presented as the mean ± SD of at least three determinations.

161

Differences between treatment groups were assessed for significance using ANOVA.

ACS Paragon Plus Environment

8

Page 9 of 30 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

Molecular Pharmaceutics

162

RESULTS AND DISSCUSSIONS

163

Selection of M-LPO preparation method. Most studies of virus wrapped in liposome have used

164

non-enveloped adenovirus-associated viruses. To encapsulate these viruses into liposomes, a

165

method based on calcium-induced phase change was usually used. We speculated that this method

166

may not be optimal for the enveloped M1 virus, because Ca2+ affects the stability of its

167

phospholipid layer. To our knowledge, liposome encapsulation of M1 has never been reported.

168

Prior to screen the preparation method, soybean lecithin and egg yolk lecithin were compared as

169

lipid materials to prepare liposomes encapsulating M1. The particle size of M-LPO made from egg

170

yolk lecithin significantly increased and the sample turned into opalescent turbid on the third day

171

at 4℃. In view of the instability of the liposomes made from egg yolk lecithin, soybean lecithin

172

was finally selected for the subsequent research (data not shown).

173

For optimization of M-LPO preparation, we tested three methods: thin-film hydration, calcium-

174

induced phase change, and co-incubation. As shown in table 1, the highest titer of M-LPO was

175

obtained using the thin-film hydration method. M-LPO that was prepared by co-incubation showed

176

nearly undetectable antitumor activity, suggesting that naked M1 cannot passively enter the

177

liposome. Thin-film hydration is simpler than the method based on calcium-induced phase change,

178

and it gives a higher M-LPO titer. Therefore, the thin-film hydration method was used to prepare

179

M-LPO for further study.

180

Table 1. M-LPO titers obtained with different methods

Method

Titer (pfu/mL)±SD

ACS Paragon Plus Environment

9

Molecular Pharmaceutics 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

Thin-film hydration

1.8×105±2.8×104

Calcium-induced phase change

4.2×104±6.6×103

Co-incubation

8.2×102±0

Page 10 of 30

181 182

Characterization. Abstract graphic shows a schematic representation of how M-LPO is designed

183

to evade immune inactivation to act on cancer cells. M-LPO and naked M1 accumulate at the

184

tumor site passively due to the enhanced permeability and retention (EPR) effect. After

185

intravenous M1 injection, the body produces M1-neutralizing antibodies, but the phospholipid

186

bilayer of the liposome of M-LPO prevents the antibodies from binding to epitopes on M1. As a

187

result, M1 accumulates in tumors in an active form, resulting in cancer cell apoptosis.

188

TEM revealed homogeneous morphology of naked M1, LPO, and M-LPO in Figure 1. Naked M1

189

appeared spherical or nearly so, consistent with its known structure. The outline of M1 indicated

190

that the average particle size of M1 was about from 65 to 80nm. It can also be seen from Figure

191

(M-LPO) that M1 could be encapsulated into liposomes with one virus per liposome in average.

192

As seen in Table 2, the particle sizes of M-LPO and LPO were similar, 155.9 nm and 167.4 nm,

193

respectively, which may be attributed to 15 extrusions of suspension through polycarbonate

194

membranes of 200 nm pore size. It was also found that those particle sizes were about 1.6 times

195

larger than that of M1. These sizes suggest that the encapsulated virus can be efficiently transported

196

across vessel walls into tumor tissues, where it can accumulate through the EPR effect. Presumably

197

the outer phospholipid bilayer of the liposome should protect M1 from inactivation by antibodies

198

in the blood.

199

ACS Paragon Plus Environment

10

Page 11 of 30 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

Molecular Pharmaceutics

200 201

Figure 1. TEM images of blank liposome (LPO), naked M1 (M1) and liposome-encapsulated M1

202

(M-LPO). White hollow arrows indicate M1 membrane, and white solid arrow indicate liposome

203

membrane.

204

Table 2. Particle Sizes and Zeta Potentials of LPO, M1 and M-LPO

Sample

Particle Size (nm)

SD (nm)

PDIa

SD

Zeta Potential (mV)

SD (mV)

M1

99.4

1.75

0.188

0.072

-12.3

0.56

LPO

167.4

1.25

0.108

0.022

-10.0

0.43

M-LPO

155.9

2.49

0.101

0.006

-14.1

0.26

205

aPDI:

206

The EE% of M-LPO was 28.50%±2.85% (n=3) based on assay of viral protein, while the titer of

207

M-LPO was 5.65×106 ±1.83×106 pfu/mL (n=3) based on the TCID50 assay. The relatively low

208

EE% may be because M1 has a molecular weight in the megadaltons, so it is more difficult to

209

encapsulate in liposomes.

210

Antibody-mediated inactivation of M1. M1-neutralizing antibody was prepared in rats, then the

211

optimal dilution was determined using the fixed virus dilution method with LoVo cells. Survival

212

rates were not significantly different between PCtrl and TCtrl cultures (Figure 2), suggesting that

213

treating virus for 1 h at 37 ℃ negligibly affected its activity. These data further showed that 64-

polydispersity index.

ACS Paragon Plus Environment

11

Molecular Pharmaceutics 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 12 of 30

214

fold dilution of antibody nearly entirely inactivated M1, leading to similar virus activity as in NCtrl

215

cultures. As expected, higher dilutions resulted in higher viral activity, with 512-fold dilution

216

giving viral activity close to the TCtrl. These results indicate that the neutralizing antibody in

217

serum affected M1 activity, and 64-fold dilution was used in further experiments to investigate the

218

protective effect of liposomes on M1.

219 220

Figure 2. Virus neutralization test. All the groups except positive control group (PCtrl) were

221

treated with neutralizing antibody for 1 h at 37 ℃. The sample of negative control group (NCtrl)

222

was DMEM; that of PCtrl was M1 untreated; that of treated control group (TCtrl) was M1 treated.

223

The value in abscissa was on behalf of the dilution ratio of antibody. ns, no significant difference;

224

***, P < 0.001.

225

M-LPO protects M1 from immune neutralization in vitro. We compared the effect of M1 or

226

M-LPO on the viability of M1-sensitive LoVo and Hep 3B cancer cells. M1 markedly induced cell

227

death: viability after treatment with M1 at MOI of 1.0 was 6.9% for LoVo cells and 3.8% for Hep

228

3B cells. Next we compared the viability of these cells after exposure to M1 at an MOI of 0.1 in

229

the form of naked M1, LPO+M1 or M-LPO.

ACS Paragon Plus Environment

12

Page 13 of 30 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

Molecular Pharmaceutics

230

In these first experiments, the virus was not previously treated with neutralizing antibody (Non-

231

immunized, Figure 3). Control cultures were treated with pure DMEM or LPO. Treating cells with

232

non-neutralized M1 led to 38.1% survival in the case of LoVo cells or 43.3% in the case of Hep

233

3B cells. This is consistent with the known sensitivity of both cell lines to M1. The survival rates

234

when LoVo or Hep 3B cells were incubated with LPO+M1 were lower by 10.9% or 15.7%,

235

respectively, than when they were incubated with M1. This may mean that M1 internalization by

236

tumor cells may be enhanced in the presence of blank liposomes, which should be examined in

237

future work. Compared with control cultures, the survival rates of LoVo or Hep 3B cells incubated

238

with M-LPO declined significantly, suggesting that encapsulating M1 in liposomes does not mask

239

its antitumor activity. They were a little higher by 9.3% or 11.3%, respectively, than when they

240

were incubated with M1, which may be due to the outer phospholipid layer may delay virus

241

biosynthesis, assembly and release.

242

These experiments were repeated after previously treating the virus with neutralizing antibody

243

(Immunized, Figure 3). The survival rate was above 95% with M1, indicating that our conditions

244

were adequate to inactivate the naked virus nearly completely. Similar inactivation was observed

245

with LPO+M1, suggesting that virus sensitivity to antibody inactivation is unaffected by the

246

presence of blank liposomes and that virus does not passively enter into liposomes to become

247

protected from inactivation. In contrast, the survival rate was only 62.0% for LoVo cells and 56.8%

248

for Hep 3B cells after treatment with neutralized M-LPO, suggesting that the liposomes efficiently

249

blocked antibody inactivation of M1.

250

In short, M-LPO showed antitumor efficacy in vitro similar to that of naked M1 in the absence of

251

prior antibody neutralization. In the presence of such neutralization, naked M1 was completely

252

inactivated, while M-LPO largely retained its ability to kill M1-sensitive Hep 3B and LoVo cells.

ACS Paragon Plus Environment

13

Molecular Pharmaceutics 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 14 of 30

253 254

Figure 3. Antitumor activity of M-LPO in LoVo cells (A) and Hep 3B cells (B) with (Immunized)

255

or without (Non-immunized) neutralizing antibody. ns, no significant difference; ***, P < 0.001.

256 257

M-LPO reduces M1 immunogenicity in vivo. Next we examined the immune response of M-

258

LPO in vivo. Mice were injected intravenously with naked M1, blank liposomes or M-LPO. Two

259

weeks after the second injection, serum was harvested and titers of M1-neutralizing antibody were

260

determined using an indirect method, since the method of detecting the quantity of M1 virus

261

neutralizing antibody is still in building. In this indirect approach, we mixed the serum from treated

262

mice with a purified preparation of naked M1 of known titer. We reasoned that higher titer of M1-

263

neutralizing antibodies in the serum should reduce the titer of M1. As expected, serum from M1-

264

treated mice led to the lowest titer in the purified M1 preparation in Figure 4. The apparent titer in

265

serum from LPO-treated mice was similar to that in serum from control animals, suggesting that

266

blank liposomes on their own do not stimulate production of M1-neutralizing antibody. Titer in

267

serum from M-LPO-treated mice appeared to be lower than that in serum from M1-treated animals.

268

These results are consistent with the idea that the outer phospholipid bilayer in M-LPO can mask

269

epitopes on M1, triggering lower production of M1-neutralizing antibodies.

ACS Paragon Plus Environment

14

Page 15 of 30 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

Molecular Pharmaceutics

270 271

Figure 4. Titer of M1 when incubated with the serum of mice. Those mice were respectively

272

immunized by saline (Ctrl), naked M1 (M1), blank liposome (LPO) or liposome-encapsulated M1

273

(M-LPO). **, P < 0.01; ***, P < 0.001.

274

Analysis of M-LPO uptake into cancer cells. To ensure the most reliable analysis of M-LPO

275

uptake into target cells, we adopted a double labelling strategy in which, for the first time to our

276

knowledge, M1 was conjugated to FITC. This was achieved by incubating M1 in alkaline solution,

277

such that a covalent bond formed between the thiamine moiety in FITC and the amino group of

278

lysines in M1. Free FITC was removed on a Sephadex G-50 column (Figure 5). Free FITC eluted

279

in 10-25 mL: these fractions showed maximum fluorescence but failed to infect BHK cells.

280

Fractions eluting at 3-5 mL showed high viral titer and fluorescence, identifying them as FITC-

281

labelled M1.

ACS Paragon Plus Environment

15

Molecular Pharmaceutics 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 16 of 30

282 283

Figure 5. Elution profile of FITC-labelled M1. Each drip’s titer and fluorescence intensity were

284

detected.

285

FM1 was encapsulated into liposomes previously labelled with Rhodamine DHPE (RhLPO), and

286

this formulation was added to LoVo cells for 4 h, after which cells were analyzed for uptake of

287

FM1-RhLPO based on green signal from FM1 and red signal from the RhLPO (Figure 6). The

288

intact complexes of liposome-encapsulated M1 gave yellow signal due to colocalization of the two

289

labelled components. Cell nuclei appeared blue because of DAPI staining. Cells treated with naked

290

FM1 showed fluorescence scattered around nuclei, suggesting that FM1 can be taken up by LoVo

291

cells in the same manner as M1. Cells treated with FM1-RhLPO showed some green signal in the

292

overlay channel, suggesting rupture of some liposomes before their internalization, liberating FM1

293

to be internalized into tumor cells. As expected, there was also yellow signal in the overlay

294

channel, suggesting that a proportion of the liposomes enter tumor cells intact, subsequently

295

releasing FM1 after uptake. In other words, our data suggest that M-LPO may release its M1 cargo

296

before or after internalization, and that the liposome coating does not alter M1 internalization.

297

Further experiments should examine the specific internalization pathways involved.

ACS Paragon Plus Environment

16

Page 17 of 30 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

Molecular Pharmaceutics

298 299

Figure 6. Confocal microscopy images of LoVo cells. Blue channel: cell nucleus stained by DAPI;

300

Green channel: M1 virus labelled by FITC; Red channel: liposome containing Rhodamine DHPE.

301

CONCLUSIONS

302

While oncolytic viruses show potential as anti-cancer treatments, they can elicit neutralizing

303

immune responses. Here we demonstrate that encapsulating the M1 alphavirus in liposomes

304

strongly reduces its immunogenicity and may preserve its anti-tumor efficacy in M1-sensitive

305

cancers. Our results suggest that the outer phospholipid coating on M1 effectively blocks the

306

binding of M1-neutralizing antibodies to the virus without affecting the ability of the virus to be

307

internalized into target cells. Our experiments with doubly-labelled M-LPO suggests that M1 can

308

be liberated from the liposomes before or after uptake by cancer cells. These experiments are, to

ACS Paragon Plus Environment

17

Molecular Pharmaceutics 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 18 of 30

309

our knowledge, the first report of liposome encapsulation of M1 and the first report of M1-FITC

310

conjugation for post-uptake analysis. Our findings justify further work into liposome-encapsulated

311

M1 as a promising anti-cancer therapy. Since liposomes contain hydrophilic and hydrophobic

312

regions that can encapsulate small molecules, our platform may be useful for combining M1 and

313

small molecules for synergistic effects.

314

ASSOCIATED CONTENT

315

Supporting Information.

316

The following files are available free of charge.

317

The separation of naked M1 and liposome-encapsulated M1 using a Cellufine Sulfate column; the

318

preparation of liposome-encapsulated M1 by calcium-induced phase change method and co-

319

incubation method; the determination of encapsulation efficiency by detecting the total protein of

320

M1 (PDF)

321

AUTHOR INFORMATION

322

Corresponding Author

323

* Tel: +86 20 39336119. E-mail: [email protected].

324

Author Contributions

325

# These authors contributed equally to this work.

326

Notes

327

The authors declare no competing financial interest.

328

ACKNOWLEDGMENT

ACS Paragon Plus Environment

18

Page 19 of 30 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

Molecular Pharmaceutics

329

We thank Prof. Wenbo Zhu, Dr. Kai Li, Dr. Yuan Lin and Dr. Jiangkai Liang for technical

330

assistance with operation of M1. This work was funded by the National Natural Science

331

Foundation of China (Grants 81473154, 81573447 and 81603127); Fundamental Research Funds

332

for central universities (Grants 18ykzd08); Natural Science Foundation of Guangdong Province,

333

China (Grants 2016A030310160 and 2016A030310146); Science and Technology Planning

334

Project of Guangdong Province, China (Grant 20160909); Research and Development Project of

335

Applied Science and Technology of Guangdong Province, China (Grant 2016B020237004) and

336

Science and Technology Planning Project of Guangdong Province, China (Grant

337

2015B020211003).

338

ABBREVIATIONS

339

ZAP, zinc finger antiviral protein; M-LPO, liposome-encapsulated M1; FM1, FITC-labelled M1;

340

FM1-RhLPO, doubly labelled M-LPO with M1 labelled by FITC and liposomal shells labelled by

341

Rhodamine DHPE; LPO, blank liposomes; TEM, transmission electron microscopy; EE,

342

Encapsulation efficiency; MOI, multiplicity of infection.

343

REFERENCES

344

(1) Liu, T. C.; Galanis, E.; Kirn, D. Clinical trial results with oncolytic virotherapy: a century of

345

promise, a decade of progress. Nat. Clin. Pract. Oncol. 2007, 4 (2), 101-117.

346

(2) Miest, T. S.; Cattaneo, R. New viruses for cancer therapy: meeting clinical needs. Nat. Rev.

347

Microbiol. 2014, 12 (1), 23-34.

348

(3) Russell, S. J.; Peng, K. W.; Bell, J. C. Oncolytic virotherapy. Nat. Biotechnol. 2012, 30 (7),

349

658-670.

350

(4) Moehler, M.; Blechacz, B.; Weiskopf, N.; Zeidler, M.; Stremmel, W.; Rommelaere, J.; Galle,

351

P. R.; Cornelis, J. J. Effective infection, apoptotic cell killing and gene transfer of human hepatoma

ACS Paragon Plus Environment

19

Molecular Pharmaceutics 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 20 of 30

352

cells but not primary hepatocytes by parvovirus H1 and derived vectors. Cancer Gene Ther. 2001,

353

8 (3), 158-167.

354

(5) Morris, D. G.; Feng, X. L.; DiFrancesco, L. M.; Fonseca, K.; Forsyth, P. A.; Paterson, A. H.;

355

Coffey, M. C.; Thompson, B. REO-001: A phase I trial of percutaneous intralesional

356

administration of reovirus type 3 dearing (Reolysin (R)) in patients with advanced solid tumors.

357

Invest. New Drugs 2013, 31 (3), 696-706.

358

(6) Garant, K. A.; Shmulevitz, M.; Pan, L.; Daigle, R. M.; Ahn, D. G.; Gujar, S. A.; Lee, P. W. K.

359

Oncolytic reovirus induces intracellular redistribution of Ras to promote apoptosis and progeny

360

virus release. Oncogene 2016, 35 (6), 771-782.

361

(7) Pecora, A. L.; Rizvi, N.; Cohen, G. I.; Meropol, N. J.; Sterman, D.; Marshall, J. L.; Goldberg,

362

S.; Gross, P.; O'Neil, J. D.; Groene, W. S.; Roberts, M. S.; Rabin, H.; Bamat, M. K.; Lorence, R.

363

M. Phase I trial of intravenous administration of PV701, an oncolytic virus, in patients with

364

advanced solid cancers. J. Clin. Oncol. 2002, 20 (9), 2251-66.

365

(8) Fournier, P.; Bian, H.; Szeberenyi, J.; Schirrmacher, V. Analysis of three properties of

366

Newcastle disease virus for fighting cancer: tumor-selective replication, antitumor cytotoxicity,

367

and immunostimulation. Methods Mol. Biol. 2012, 797, 177-204.

368

(9) Habib, N. A.; Sarraf, C. E.; Mitry, R. R.; Havlik, R.; Nicholls, J.; Kelly, M.; Vernon, C. C.;

369

Gueret-Wardle, D.; El-Masry, R.; Salama, H.; Ahmed, R.; Michail, N.; Edward, E.; Jensen, S. L.

370

E1B-deleted adenovirus (dl1520) gene therapy for patients with primary and secondary liver

371

tumors. Hum. Gene Ther. 2001, 12 (3), 219-26.

372

(10) Yu, W.; Fang, H. Clinical trials with oncolytic adenovirus in China. Curr. Cancer Drug

373

Targets 2007, 7 (2), 141-8.

ACS Paragon Plus Environment

20

Page 21 of 30 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

Molecular Pharmaceutics

374

(11) Harrow, S.; Papanastassiou, V.; Harland, J.; Mabbs, R.; Petty, R.; Fraser, M.; Hadley, D.;

375

Patterson, J.; Brown, S. M.; Rampling, R. HSV1716 injection into the brain adjacent to tumour

376

following surgical resection of high-grade glioma: safety data and long-term survival. Gene Ther.

377

2004, 11 (22), 1648-1658.

378

(12) Parker, J. N.; Zheng, X. J.; Luckett, W.; Markert, J. M.; Cassady, K. A. Strategies for the

379

Rapid Construction of Conditionally-Replicating HSV-1 Vectors Expressing Foreign Genes as

380

Anticancer Therapeutic Agents. Mol. Pharmaceutics 2011, 8 (1), 44-49.

381

(13) Willmon, C. L.; Saloura, V.; Fridlender, Z. G.; Wongthida, P.; Diaz, R. M.; Thompson, J.;

382

Kottke, T.; Federspiel, M.; Barber, G.; Albelda, S. M.; Vile, R. G. Expression of IFN-beta

383

Enhances Both Efficacy and Safety of Oncolytic Vesicular Stomatitis Virus for Therapy of

384

Mesothelioma. Cancer Res. 2009, 69 (19), 7713-7720.

385

(14) Wen, J. S.; Zhao, W. Z.; Liu, J. W.; Zhou, H.; Tao, J. P.; Yan, H. J.; Liang, Y.; Zhou, J. J.;

386

Jiang, L. F. Genomic analysis of a Chinese isolate of Getah-like virus and its phylogenetic

387

relationship with other Alphaviruses. Virus Genes 2007, 35 (3), 597-603.

388

(15) Hu, J.; Cai, X. F.; Yan, G. Alphavirus M1 induces apoptosis of malignant glioma cells via

389

downregulation and nucleolar translocation of p21WAF1/CIP1 protein. Cell cycle 2009, 8 (20),

390

3328-39.

391

(16) Lin, Y.; Zhang, H. P.; Liang, J. K.; Li, K.; Zhu, W. B.; Fu, L. W.; Wang, F.; Zheng, X. K.;

392

Shi, H. J.; Wu, S. H.; Xiao, X.; Chen, L. J.; Tang, L. P.; Yan, M.; Yang, X. X.; Tan, Y. Q.; Qiu, P.

393

X.; Huang, Y. J.; Yin, W.; Su, X. W.; Hu, H. Y.; Hu, J.; Yan, G. M. Identification and

394

characterization of alphavirus M1 as a selective oncolytic virus targeting ZAP-defective human

395

cancers. Proc. Natl. Acad. Sci. U. S. A. 2014, 111 (42), 4504-4512.

ACS Paragon Plus Environment

21

Molecular Pharmaceutics 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 22 of 30

396

(17) Cai, J.; Lin, Y.; Zhang, H. P.; Liang, J. K.; Tan, Y. Q.; Cavenee, W. K.; Yan, G. M. Selective

397

replication of oncolytic virus M1 results in a bystander killing effect that is potentiated by Smac

398

mimetics. Proc. Natl. Acad. Sci. U. S. A. 2017, 114 (26), 6812-6817.

399

(18) Pesonen, S.; Kangasniemi, L.; Hemininki, A. Oncolytic Adenoviruses for the Treatment of

400

Human Cancer: Focus on Translational and Clinical Data. Mol. Pharmaceutics 2011, 8 (1), 12-28.

401

(19) Barnard, A. S. Nanohazards: Knowledge is our first defence. Nat. Mater. 2006, 5 (4), 245-

402

248.

403

(20) Ong, H. T.; Hasegawa, K.; Dietz, A. B.; Russell, S. J.; Peng, K. W. Evaluation of T cells as

404

carriers for systemic measles virotherapy in the presence of antiviral antibodies. Gene Ther. 2007,

405

14 (4), 324-333.

406

(21) Hsiao, W. C.; Sung, S. Y.; Liao, C. H.; Wu, H. C.; Hsieh, C. L. Vitamin D3-inducible

407

mesenchymal stem cell-based delivery of conditionally replicating adenoviruses effectively targets

408

renal cell carcinoma and inhibits tumor growth. Mol. Pharmaceutics 2012, 9 (5), 1396-408.

409

(22) Ran, L.; Tan, X. H.; Li, Y. C.; Zhang, H. F.; Ma, R. H.; Ji, T. T.; Dong, W. Q.; Tong, T.; Liu,

410

Y. Y.; Chen, D. G.; Yin, X. N.; Liang, X. Y.; Tang, K.; Ma, J. W.; Zhang, Y.; Cao, X. T.; Hu, Z.

411

W.; Qin, X. F.; Huang, B. Delivery of oncolytic adenovirus into the nucleus of tumorigenic cells

412

by tumor microparticles for virotherapy. Biomaterials 2016, 89, 56-66.

413

(23) Qiao, J.; Wang, H.; Kottke, T.; Diaz, R. M.; Willmon, C.; Hudacek, A.; Thompson, J.; Parato,

414

K.; Bell, J.; Naik, J.; Chester, J.; Selby, P.; Harrington, K.; Melcher, A.; Vile, R. G. Loading of

415

oncolytic vesicular stomatitis virus onto antigen-specific T cells enhances the efficacy of adoptive

416

T-cell therapy of tumors. Gene Ther. 2008, 15 (8), 604-16.

417

(24) Shikano, T.; Kasuya, H.; Sahin, T. T.; Nomura, N.; Kanzaki, A.; Misawa, M.; Nishikawa, Y.;

418

Shirota, T.; Yamada, S.; Fujii, T.; Sugimoto, H.; Kanazumi, N.; Nomoto, S.; Takeda, S.; Nakao,

ACS Paragon Plus Environment

22

Page 23 of 30 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

Molecular Pharmaceutics

419

A. High Therapeutic Potential for Systemic Delivery of a Liposome-conjugated Herpes Simplex

420

Virus. Curr. Cancer Drug Targets 2011, 11 (1), 111-122.

421

(25) Zhong, Z. R.; Han, J. F.; Wan, Y.; Zhang, Z. R.; Sun, X. Anionic Liposomes Enhance and

422

Prolong Adenovirus-Mediated Gene Expression in Airway Epithelia in Vitro and in Vivo. Mol.

423

Pharmaceutics 2011, 8 (3), 673-682.

424

(26) Wan, Y.; Han, J. F.; Fan, G. R.; Zhang, Z. R.; Gong, T.; Sun, X. Enzyme-responsive liposomes

425

modified adenoviral vectors for enhanced tumor cell transduction and reduced immunogenicity.

426

Biomaterials 2013, 34 (12), 3020-3030.

427

(27) Mendez, N.; Herrera, V.; Zhang, L. Z.; Hedjran, F.; Feuer, R.; Blair, S. L.; Trogler, W. C.;

428

Reid, T. R.; Kummel, A. C. Encapsulation of adenovirus serotype 5 in anionic lecithin liposomes

429

using a bead-based immunoprecipitation technique enhances transfection efficiency. Biomaterials

430

2014, 35 (35), 9554-9561.

431

(28) Fu, X. P.; Zhang, X. L. Delivery of herpes simplex virus vectors through liposome

432

formulation. Mol. Ther. 2001, 4 (5), 447-453.

433

(29) Zhong, Z.; Shi, S.; Han, J.; Zhang, Z.; Sun, X. Anionic liposomes increase the efficiency of

434

adenovirus-mediated gene transfer to coxsackie-adenovirus receptor deficient cells. Mol.

435

Pharmaceutics 2010, 7 (1), 105-15.

436

(30) Yoshimura, A.; Ohnishi, S. Uncoating of influenza virus in endosomes. J. Virol. 1984, 51 (2),

437

497-504.

438

(31) Nichols, J. E.; Mock, D. J.; Roberts, N. J., Jr. Use of FITC-labeled influenza virus and flow

439

cytometry to assess binding and internalization of virus by monocytes-macrophages and

440

lymphocytes. Arch. Virol. 1993, 130 (3-4), 441-55.

441

ACS Paragon Plus Environment

23

Molecular Pharmaceutics 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

Figure 1. TEM images of blank liposome (LPO), naked M1 (M1) and liposome-encapsulated M1 (M-LPO). White hollow arrows indicate M1 membrane, and white solid arrow indicate liposome membrane. 74x27mm (600 x 600 DPI)

ACS Paragon Plus Environment

Page 24 of 30

Page 25 of 30 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

Molecular Pharmaceutics

Figure 2. Virus neutralization test. All the groups except positive control group (PCtrl) were treated with neutralizing antibody for 1 h at 37 ℃. The sample of negative control group (NCtrl) was DMEM; that of PCtrl was M1 untreated; that of treated control group (TCtrl) was M1 treated. The value in abscissa was on behalf of the dilution ratio of antibody. ns, no significant difference; ***, P < 0.001. 59x48mm (600 x 600 DPI)

ACS Paragon Plus Environment

Molecular Pharmaceutics 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

Figure 3. Antitumor activity of M-LPO in LoVo cells (A) and Hep 3B cells (B) with (Immunized) or without (Non-immunized) neutralizing antibody. ns, no significant difference; ***, P < 0.001. 114x40mm (600 x 600 DPI)

ACS Paragon Plus Environment

Page 26 of 30

Page 27 of 30 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

Molecular Pharmaceutics

Figure 4. Titer of M1 when incubated with the serum of mice. Those mice were respectively immunized by saline (Ctrl), naked M1 (M1), blank liposome (LPO) or liposome-encapsulated M1 (M-LPO). **, P < 0.01; ***, P < 0.001. 74x59mm (600 x 600 DPI)

ACS Paragon Plus Environment

Molecular Pharmaceutics 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

Figure 5. Elution profile of FITC-labelled M1. Each drip’s titer and fluorescence intensity were detected.

ACS Paragon Plus Environment

Page 28 of 30

Page 29 of 30 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

Molecular Pharmaceutics

Figure 6. Confocal microscopy images of LoVo cells. Blue channel: cell nucleus stained by DAPI; Green channel: M1 virus labelled by FITC; Red channel: liposome containing Rhodamine DHPE.

ACS Paragon Plus Environment

Molecular Pharmaceutics 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

Table of Contents Graphic/Abstract Graphic

ACS Paragon Plus Environment

Page 30 of 30