Accepted Manuscript Antiviral activity of SA-2 against influenza A virus in vitro/vivo and its inhibition of RNA polymerase Jie Yu, Dechuan Wang, Jing Jin, Jun Xu, Mengwei Li, Hui Wang, Jie Dou, Changlin Zhou PII:

S0166-3542(16)30013-4

DOI:

10.1016/j.antiviral.2016.01.011

Reference:

AVR 3759

To appear in:

Antiviral Research

Received Date: 23 September 2015 Revised Date:

19 December 2015

Accepted Date: 19 January 2016

Please cite this article as: Yu, J., Wang, D., Jin, J., Xu, J., Li, M., Wang, H., Dou, J., Zhou, C., Antiviral activity of SA-2 against influenza A virus in vitro/vivo and its inhibition of RNA polymerase, Antiviral Research (2016), doi: 10.1016/j.antiviral.2016.01.011. 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.

ACCEPTED MANUSCRIPT Antiviral activity of SA-2 against influenza A virus in vitro/vivo and its inhibition of RNA polymerase

RI PT

Jie Yu a,1, Dechuan Wang b,1, Jing Jin a, Jun Xu a, Mengwei Li a, Hui Wang a, Jie Dou a, Changlin Zhou a,*

State Key Laboratory of Natural Medicines, School of Life Science and Technology,

SC

a

b

M AN U

China Pharmaceutical University, Nanjing, Jiangsu 210009, PR China; Department of Organic Chemistry, School of Science, China Pharmaceutical

University, Nanjing, Jiangsu 210009, PR China

Corresponding author:

TE D

*

Tel: +86-25-8327-1323; Fax: +86-25-8327-1323; E-mail: [email protected] Address: School of Life Science and Technology, China Pharmaceutical University,

1﹥

AC C

EP

24 Tong Jia Xiang, Nanjing 210009, PR China.

These authors contributed equally to this work.

ACCEPTED MANUSCRIPT Abstract

2

A target-free and cell-based approach was applied to evaluate the anti-influenza

3

properties of six newly synthesized benzoic acid derivatives. SA-2, the ethyl

4

4-(2-hydroxymethyl-5-oxopyrrolidin-1-yl)-3-[3-(3-methylbenzoyl)-thioureido]

5

benzoate (compound 2) was screened as a potential drug candidate. In a cytopathic

6

effect

7

oseltamivir-resistant mutant H1N1-H275Y influenza viruses in both virus-infected

8

MDCK and A549 cells, with 50% effective concentrations (EC50) in MDCK cells of

9

9.6, 19.2 and 19.8 µM respectively, and 50% cytotoxic concentration (CC50) of 444.5

10

µM, showing competitive antiviral activity with oseltamivir in vitro. Orally

11

administered SA-2 effectively protected mice infected with lethal doses of H1N1 or

12

oseltamivir-resistant strain H1N1-H275Y, conferring 70% or 50% survival at a

13

dosage of 100 mg/kg/d, reducing body weight loss, alleviating the influenza-induced

14

acute lung injury, and reducing lung virus titer. Mechanistic studies showed that SA-2

15

efficiently inhibited the activity of RNA polymerase and suppressed NP and M1

16

levels during viral biosynthesis by interfering with gene transcription without having

17

an obvious influence on virus entry and release. Based on these favourable findings,

18

SA-2, a novel anti-influenza agent, with its potent anti-influenza activity in vitro and

19

in vivo, could be a promising antiviral for the treatment of infection of influenza A

20

viruses, including oseltamivir-resistant mutants.

21

Keywords

22

Benzoic acid derivatives; Influenza A virus; Antiviral activity; Oseltamivir-resistant;

SA-2

dose

dependently

inhibited

H1N1,

H3N2

and

the

AC C

EP

TE D

M AN U

SC

assay,

RI PT

1

ACCEPTED MANUSCRIPT 23

RNA polymerase inhibition

24

1. Introduction Influenza viruses are highly contagious and cause three to five million cases of

26

severe respiratory illness and approximately 250 000 to 500 000 deaths annually

27

(Fiore et al., 2008). Influenza A viruses include many subtypes; the H1N1 and H3N2

28

subtypes infect humans, resulting in seasonal and pandemic infections. The highly

29

pathogenic avian H5N1 and H7N9 subtypes have an estimated mortality rate of 60%

30

and 30%, respectively. There is concern that these viruses could cause severe human

31

influenza pandemics in case they would acquire human-to-human transmissibility (Li

32

et al., 2014).

M AN U

SC

RI PT

25

Currently, other than vaccination, antiviral treatment is the most effective

34

therapeutic method for the prevention and treatment of human influenza. M2 channel

35

blockers (e.g., amantadine and rimantadine), inhibitors of viral RNA synthesis (e.g.,

36

ribavirin and T-705) and neuraminidase (NA) inhibitors (e.g., oseltamivir) are three

37

classes of anti-influenza virus drugs with different modes of action. However, clinical

38

use of amantadine (or rimantadine) and ribavirin has met several limitations over

39

recent decades because of the high frequency of emerging variants that are resistant

40

for M2 channel blockers and the undesirable toxic side effects of ribavirin.

AC C

EP

TE D

33

41

The current standard-of-care antivirals for influenza cases are potent influenza virus

42

NA inhibitors, such as zanamivir and oseltamivir (Schmidt, 2004; Moscona, 2005).

43

While these agents possess high affinity and specificity for a variety of influenza

44

viruses, they suffer from limitations in their efficacy due to adverse effects and drug

ACCEPTED MANUSCRIPT resistance (Thorlund et al., 2011). For oseltamivir, drug-resistant strains have

46

increasingly emerged since 2007. There were many more reported cases of

47

oseltamivir-resistant H1N1pdm09 infections with the H275Y NA mutation during

48

2011 than during the first year of the pandemic (Leang et al., 2013). The appearance

49

of drug-resistant influenza viruses caused by NA mutations limits the future utility of

50

commonly used NA inhibitors, highlighting an urgent need for new classes of

51

anti-influenza agents to combat potential human influenza pandemics.

SC

RI PT

45

BANA-206 (Atigadda et al., 1999), a benzoic acid derivative, was reported to show

53

sub-micromolar antiviral potency against influenza A virus. Acylthiourea derivatives

54

(Sun et al., 2006; Burgeson et al., 2012) were reported for possessing broad-spectrum

55

antiviral activity. Some compounds with enhanced anti-influenza activity have been

56

successfully designed and synthesized by conjugation method, such as compound

57

BTA938 (Tarbet, et al., 2014) and ZA-7-CA (Liu, et al., 2012). Here, in order to

58

achieve potential influenza virus inhibitors with better antiviral activity, a series of

59

novel benzoic acid derivatives (Fig. 1) were designed and synthesized by conjugation

60

of benzoic acid with acylthiourea based on the combination principles as well as the

61

principle of functional groups. In this study, six compounds were evaluated for

62

antiviral activity against both H1N1 (including its oseltamivir-resistant H275Y strain)

63

and H3N2 influenza A viruses in a cell culture-based assay. We further examined the

64

efficacy of ethyl 4-(2-hydroxymethyl-5-oxopyrrolidin-1-yl)-3-[3-(3-methylbenzoyl)

65

-thioureido] benzoate (C23H25N3O5S, compound 2, named SA-2) against the H1N1

66

and H3N2 subtypes and the oseltamivir-resistant strain H1N1-H275Y in vitro and in

AC C

EP

TE D

M AN U

52

ACCEPTED MANUSCRIPT vivo. Mechanistic studies were then conducted to investigate its antiviral target and

68

mode of action. Our findings demonstrate the potential of SA-2 as an alternative

69

antiviral agent against influenza A viruses, including oseltamivir-resistant mutants.

70

2. Materials and methods

71

2.1. Cells, viruses and plasmids

RI PT

67

Madin-Darby canine kidney cells (MDCK), human lung cancer cells (A549) and

73

human embryonic kidney 293T cells were obtained from the American Type Culture

74

Collection (ATCC, Manassas, VA). MDCK and 293T cell lines were cultured in

75

DMEM, while the A549 cell line was cultured in RPMI-1640 medium, supplemented

76

with 10% foetal bovine serum; all cell lines were cultured at 37°C in the presence of

77

5% CO2.

TE D

M AN U

SC

72

Influenza A viruses subtypes A/FM/1/47 (H1N1) and A/Beijing/32/92 (H3N2) and

79

an H1N1 oseltamivir-resistant mutant strain possessing a histidine-to-tyrosine

80

substitution at position 275 (H275Y) of NA, were maintained at the Department of

81

Microbiology, School of Life Science and Technology, China Pharmaceutical

82

University. The NA of the A/FM/1/47-H275Y (H1N1-H275Y) oseltamivir-resistant

83

virus was sequenced to confirm retention of the H275Y mutation. All these viruses

84

were propagated in 10-day-old embryonated chicken eggs and stored at -80°C until

85

use (Jung et al., 2010).

AC C

EP

78

86

Plasmids for expressing influenza virus (A/WSN/33) PB1, PB2, PA and NP, a

87

polymerase expressing plasmid carrying an influenza virus-like RNA encoding firefly

ACCEPTED MANUSCRIPT 88

luciferase (vNS-Luc), and pCMV β-gal plasmids (Zhang et al., 2012) were kindly

89

provided by Xin Ye, Institute of Microbiology, Chinese Academy of Sciences.

90

2.2. Compounds Oseltamivir (OS) and oseltamivir carboxylate (OC) were purchased from

92

MedChem Express (Shanghai, China). Ribavirin (RBV) was obtained from Sichuan

93

Baili Pharmaceutical Co., Ltd. (Chengdu, China). Test compounds were synthesized

94

at the School of Science, China Pharmaceutical University, according to published

95

procedures (Wang et al., 2015). In brief, ethyl aminobenzoate, as the starting material,

96

was reacted with diethyl bromomalonate. Subsequent reactions included cyclization,

97

nitration and reduction. Then the product was treated with R-isothiocyanate to obtain

98

the target compounds 1–6. The structures of the purified compounds were confirmed

99

by 1H NMR (Bruker Avance 300) and the purity was determined to be

TE D

M AN U

SC

RI PT

91

95%. Each

compound was dissolved in dimethyl sulfoxide (DMSO) to a 50 mM concentration

101

for use as stock solutions that were diluted to the required concentrations for in vitro

102

studies.

103

2.3. Determination of influenza A virus TCID50

AC C

104

EP

100

The TCID50 (50% tissue culture infectious dose) was determined by serial dilution

105

of the influenza virus added to 100 µL of MDCK cells cultured at a density of 5 × 103

106

cells/well in 96-well microplates. The infected cells were incubated at 37°C for 48 h

107

and then 10 µL of Cell Counting Kit-8 (Dojindo, Japan) reagent (Takeuchi et al., 2003)

108

was added to each well. After incubation for 2 h, absorbance at 450 nm was read on a

ACCEPTED MANUSCRIPT 109

plate reader. Influenza virus TCID50 was determined using the Reed-Müench method

110

(Reed and Müench, 1938).

111

2.4. Cytopathic effect (CPE) reduction assay MDCK cells and A549 cells (5 × 103 cells/well) were cultured in 96-well plates for

113

24 h and infected with 100 TCID50 A/FM/1/47 (H1N1) virus, the oseltamivir-resistant

114

strain H1N1-H275Y or A/Beijing/32/92 (H3N2) virus for 2 h. Then, the cells were

115

incubated in the maintenance medium (DMEM / RPMI-1640 containing 0.5% foetal

116

bovine serum) in the presence of 0.2% DMSO (a vehicle to dissolve test compounds)

117

or different concentrations of chemicals. After a 48-h incubation, the cell viability was

118

measured using Cell Counting Kit-8. The EC50 of test compounds were determined by

119

fitting the curve of percent CPE versus the compound concentrations using GraphPad

120

Prism 5.

TE D

M AN U

SC

RI PT

112

The inhibition rate of the test compounds was calculated using the following

122

equation (Ding, et al., 2014): inhibition rate (%) = [(mean optical density of test –

123

mean optical density of virus controls) / (mean optical density of cell controls – mean

124

optical density of virus controls)] × 100.

AC C

125

EP

121

The CC50 of chemicals to MDCK or A549 cells were determined by procedures

126

similar to those for EC50 determination but without virus infection. The selectivity

127

index (SI) was calculated as the ratio of CC50 to EC50.

128

2.5. Therapeutic efficacy study in mice

129

Mouse-adapted A/FM/1/47 (H1N1) and its oseltamivir-resistant mutant strain

ACCEPTED MANUSCRIPT A/FM/1/47-H275Y were used as the challenge viruses. Six-week-old male ICR mice

131

were purchased from the Laboratory Animal Center of Yangzhou University

132

(Yangzhou, Jiangsu, China). The experiment was performed as previously described

133

(Ding et al., 2014). Five mice per group were enrolled to determine the lung virus

134

titers. On day 5 post-infection, mice were euthanized and lungs collected and

135

homogenized. Lung homogenates were serially diluted and added to MDCK cells.

136

Virus titer was measured by TCID50 method and expressed as TCID50/lung (Byrn et

137

al., 2015). For oral gavage, SA-2 was first dissolved in a small quantity of DMSO,

138

and then evenly mixed with 0.5% CMC-Na solution. The final concentration of

139

DMSO was 5%. Animal experiments were conducted in accordance with protocols

140

approved by the Animal Ethics and Experimentation Committee of China

141

Pharmaceutical University.

142

2.6. Inhibitory effects of SA-2 on different stages of viral replication

TE D

M AN U

SC

RI PT

130

An experiment focused on the effect of SA-2 on different stages of viral replication

144

was carried out as previously described (Ding et al., 2014). In the first protocol

145

(pre-infection), MDCK cells were pre-incubated with different concentrations of SA-2

146

(10–80 µM) for 12 h before viral infection. In the second protocol (co-infection), the

147

virus suspension and SA-2 were added to MDCK cells simultaneously in a mixture

148

and incubated for 2 h. In both protocols, maintenance medium replaced the

149

supernatant after viral adsorption, and the cultures were then incubated for 48 h. In the

150

third protocol (post-infection), after adsorption of the influenza virus for 2 h,

151

maintenance medium containing SA-2 was added to MDCK cells, and the cultures

AC C

EP

143

ACCEPTED MANUSCRIPT 152

were incubated for 48 h. Cell viability in each well was determined using Cell

153

Counting Kit-8 and the inhibition rate was calculated.

154

2.7. Hemagglutination assay Hemagglutination titrations of the A/FM/1/47 (H1N1) and A/Beijing/32/92 (H3N2)

156

strains were first determined using standard methods (Clarke and Casals, 1958), and

157

the unit of hemagglutinin agglutination (HAU) for both strains were obtained. The

158

hemagglutination assay was then performed in the same condition with 4 HAU/well

159

of virus as previously described (Delogu et al., 2011). Negative controls contained no

160

virus, and the sedimentation of red blood cells was observed in these controls.

161

2.8. Neuraminidase (NA) inhibition assay

M AN U

SC

RI PT

155

Neuraminidase enzyme inhibition assays were carried out as previously described

163

(Kim et al., 2013). Allantoic fluid containing the A/FM/1/47 (H1N1) or

164

A/Beijing/32/92 (H3N2) influenza viruses was used. Chemicals were incubated with

165

diluted allantoic fluid for 10 min at room temperature followed by the addition of 20

166

µM fluorescent substrate 2’-(4-methylumbelliferyl)-α-D-N-acetylneuraminic acid

167

(MUNANA; Sigma). Fluorescence was read after incubation with MUNANA for 60

168

min using excitation and emission wavelengths of 360 and 450 nm, respectively.

169

2.9. Western blot analysis

AC C

EP

TE D

162

170

In order to determine the viral protein expression level, MDCK cells in 6-well

171

plates were infected with 1.5 × 103 TCID50 of the A/FM/1/47 (H1N1) influenza virus

172

and treated with SA-2 (40 µM) or ribavirin (40 µM) at different exposure periods after

ACCEPTED MANUSCRIPT infection. In the interval of for instance 2–4 h post infection (hpi), compound was

174

added at 2 h and then the medium was removed and replaced by fresh medium at 4h.

175

Before fresh medium was added, cells were washed twice with PBS. Ten hours post

176

infection, cells were lysed with 200 µL of Mammalian Protein Extraction Reagent

177

(GENEray Biotechnology). The protein lysates were then applied to immunoblotting

178

with anti-NP (Catalog No. GTX83054, GeneTex), anti-M1 (Catalog No. GTX125928,

179

GeneTex), and anti-β-actin (Catalog No. SC-47778, Santa Cruz) antibodies,

180

respectively. The blots were detected on a Bio-Imaging system and the band

181

intensities of proteins were quantified using ImageJ and normalized to β-actin levels.

182

2.10. RT-PCR and real-time qPCR analysis

M AN U

SC

RI PT

173

MDCK cells in 6-well plates were inoculated with 1.5 × 103 TCID50 of the

184

A/FM/1/47 (H1N1) influenza virus for 2 h followed by the addition of 40 µM SA-2.

185

Cells were collected 6 h post infection (Shih et al., 2010) and total RNA was extracted

186

using TRIzol. Total RNA was reverse-transcribed using the PrimeScript

187

Kit (Takara) to obtain cDNA. The RT-PCR experiment was performed as previously

188

described (Chen et al., 2011) and the amplification conditions were: 95°C (5 min); 30

189

cycles of 94°C (30 s), 54°C (30 s), and 72°C (20 s); and an extension at 72°C (10

190

min). Each PCR product was analyzed by electrophoresis on a 2% (w/v) agarose gel

191

and visualized using ethidium bromide staining.

TM

Reagent

AC C

EP

TE D

183

192

By using a 7300 qRT-PCR System (ABI) with a standard SYBR Green PCR

193

protocol (Fan et al., 2014) according to the manufacturer’s instructions, the real-time

194

qPCR amplification was performed with the following parameters: 95°C (10 min); 40

ACCEPTED MANUSCRIPT 195

cycles of 95°C (15 s) and 60°C (1 min); and 95°C (15 s), 60°C (1 min), 95°C (15 s).

196

Each sample was run in triplicate alongside the endogenous control (GAPDH) to

197

normalize reactions. The relative RNA levels of NP and M1 genes were calculated

198

based on the 2

method (Livak and Schmittgen, 2001).

RI PT

△△Ct

The primers used to detect the viral mRNAs are described below.

200

NP forward primer, 5'-CCCAGGATGTGCTCTCTGAT-3'

201

NP reverse primer, 5'- TGAAAGGGTCTATTCCGACT-3'

202

M1 forward primer, 5'-TTCTAACCGAGGTCGAAAC-3'

203

M1 reverse primer, 5'-AAGCGTCTACGCTGCAGTCC-3'

204

GAPDH forward primer, 5'-CACTCACGGCAAATTCAACGGCAC-3'

205

GAPDH reverse primer, 5'-GACTCCACGACATACTCAGCAC-3'

M AN U

2.11. Luciferase assay

TE D

206

SC

199

To measure the influence of SA-2 on influenza viral RNA polymerase activity,

208

293T cells in a 24-well plate were transfected with plasmids for expressing influenza

209

A virus (A/WSN/33 (H1N1)) PB1, PB2, PA, NP and vNS-Luc (150 ng of each) with

210

pCMV β-gal (50 ng) as an internal control for 6 h followed by the addition of SA-2.

211

After 24 h, the cell lysates were harvested and subjected to a luciferase assay (Zhang

212

et al., 2012). The luciferase activity was normalized to β-gal activity.

213

2.12. Statistical analysis

AC C

EP

207

214

All experiments were performed in triplicate. Data were analysed by one-way

215

analysis of variance (ANOVA). The results were presented as the mean ± S.D.

ACCEPTED MANUSCRIPT Significant differences between control and treated groups were determined using an

217

unpaired, two-tailed Student’s t test with significance set at P < 0.05. For survival

218

studies, Kaplan-Meier survival curves were generated and compared by the Log-rank

219

(Mantel-Cox) test using SPSS 17.0 software.

220

3. Results

221

3.1. Antiviral activity of SA-2 and its analogues against the A/FM/1/47 (H1N1) and

222

A/Beijing/32/1 (H3N2) influenza viruses in vitro

M AN U

SC

RI PT

216

A cell culture-based screening system was established for influenza viruses,

224

including A/FM/1/47 (H1N1), A/Beijing/32/92 (H3N2) and A/FM/1/47-H275Y, an

225

oseltamivir-resistant strain of H1N1. Using a CPE reduction assay, CC50 and EC50

226

values of compounds 1–6 were determined (Table 1). Oseltamivir carboxylate and

227

ribavirin, two commonly used antiviral drugs, were used as positive controls to

228

confirm the reliability of the assay.

TE D

223

Compounds 2–5 protected cells from influenza virus infection with limited

230

cytotoxicity, albeit with different potency. They were effective against both H1N1 and

231

H3N2 viruses, as well as the oseltamivir-resistant strain, A/FM/1/47-H275Y. Notably,

232

compound 2, ethyl 4-(2-hydroxymethyl-5-oxopyrrolidin-1-yl)-3-[3-(3-methylbenzoyl)

233

-thioureido] benzoate (designated SA-2), had the highest potency of all six

234

compounds, with EC50 values against H1N1 and H3N2 of 9.6 µM and 19.2 µM,

235

respectively, which were comparable to those of oseltamivir carboxylate. Further,

236

SA-2 (2) strongly inhibited the oseltamivir-resistant mutant strain H1N1-H275Y

AC C

EP

229

ACCEPTED MANUSCRIPT (EC50 = 19.8 µM) at a similar level to ribavirin, while the EC50 for oseltamivir

238

reached as high as 1177.9 µM. These results indicate that SA-2 (2) effectively inhibits

239

H1N1 and H3N2 influenza viruses in vitro, including the oseltamivir-resistant mutant.

240

3.2. SA-2 inhibited the replication of A/FM/1/47 (H1N1) and A/Beijing/32/92 (H3N2)

241

influenza viruses in cell culture

RI PT

237

Based on the results of the CPE reduction assay (Table 1), the ability of compound

243

2 (SA-2) to inhibit the replication of the virus in cell culture was explored using

244

additional antiviral assays with MDCK and A549 cells. SA-2 exhibited a considerable

245

protective effect when added to H1N1-infected MDCK cells at concentration of 80

246

µM. The reduction of the CPE was confirmed by direct microscopic observation

247

which detected far less CPE than in the DMSO control (Fig. 2A). The three strains

248

(H1N1, H1N1-H275Y, and H3N2) were then tested with OC as a control. SA-2 (5–80

249

µM) showed significant anti-influenza activity in a dose-dependent manner. The

250

inhibition rates at the final concentration of 80 µM against H1N1 and H3N2 reached

251

82.9% and 69.6%, respectively (Fig. 2B). For the oseltamivir-resistant (H1N1-H275Y)

252

strain, the inhibition rate of SA-2 at 80 µM was 85.9%, while that of oseltamivir

253

carboxylate (OC) was only 4.0%. In addition, SA-2 displayed clear anti-influenza

254

activity in MDCK cells infected with A/FM/1/47 (H1N1) and A/Beijing/32/92 (H3N2)

255

viruses at different MOIs (1, 0.5, and 0.3) (Fig. 2C and D). An obvious inhibitory

256

effect of SA-2 was observed at an MOI of 0.3 due to the lower infection dose. Similar

257

anti-influenza effects were observed in influenza virus-infected A549 cells (Fig. 2E).

AC C

EP

TE D

M AN U

SC

242

ACCEPTED MANUSCRIPT All three virus strains were inhibited by SA-2 in a dose-dependent manner, and the

259

rates of inhibition of the H1N1, H3N2 and H1N1-H275Y strains were 73.0%, 54.9%

260

and 78.1%, respectively, when SA-2 was used at a concentration of 80 µM. The

261

cytotoxicity of SA-2 in both cell lines was also examined. As shown in Fig. 2F, cell

262

viability was maintained at 100% even at a 200 µM concentration. The CC50 values of

263

SA-2 in MDCK and A549 cells were 444.5 µM and 580.3 µM, respectively.

RI PT

258

Taken together, these results indicate that SA-2 inhibits the replication of the

265

A/FM/1/47 (H1N1) and A/Beijing/32/92 (H3N2) influenza viruses, as well as the

266

oseltamivir-resistant mutant strain, in a dose-dependent manner in both MDCK and

267

A549 cell cultures, while also exhibiting limited cytotoxicity.

268

3.3. Therapeutic efficacy of SA-2 against A/FM/1/47 (H1N1) influenza virus in vivo

TE D

M AN U

SC

264

To evaluate the efficacy of SA-2 against influenza virus in vivo, a dose-response

270

study in a virus-infected mouse model was performed. Fig. 3A illustrates the

271

protocol for the experiments. On 'day 0', mice were infected intranasally with a

272

lethal dose of A/FM/1/47 (H1N1) virus. Two hours post infection, SA-2, oseltamivir

273

or saline (placebo) were administered twice daily for 5 days (days 0–4) by oral

274

gavage. Each mouse was weighed daily for 14 days. On 'day 5', five mice per group

275

were euthanized for lung pathological examinations. SA-2-treated mice were

276

significantly protected from influenza-induced lethality, whereas 100% of the mice

277

administered placebo died within 8 days (Fig. 3B). Treatment of mice with 50, 75 or

278

100 mg/kg/d SA-2 increased survival to 30%, 40%, 70%, respectively, and there was

AC C

EP

269

ACCEPTED MANUSCRIPT 279

a dose-dependent reduction in body weight loss (Fig. 3C). SA-2 treatment also led to a statistically significant reduction in influenza-induced

281

lung pathology. On day 5 post-infection, signs of extensive lung damage, such as

282

highly basophilic lining epithelium in the bronchioles, extensive cellular infiltrates

283

with neutrophils, and cell necrosis, were observed in placebo group (Fig. 3D). In

284

SA-2-treated mice, however, lung damage was relieved with increasing dosage.

285

When SA-2 was administered at 100 mg/kg/d, approximately 60% of lung sections

286

exhibited nearly normal lung architecture (similar to the ‘control’), while in the rest

287

of the section, inflammatory infiltrates could be observed, although to a lesser extent

288

than in the lungs of mice in the placebo group. Further, all the SA-2 treatment

289

groups reduced lung virus titers on day 5 post-infection (Fig. 3E). The log virus

290

titers calculated in SA-2 at 50, 75 and 100 mg/kg/day were 5.5 ± 0.3, 5.0 ± 0.2 and

291

4.1 ± 0.5, respectively. SA-2 showed significant reduction at the dosage of 100

292

mg/kg/day with log reduction of 1.8 compared to the placebo group (5.9 ± 0.3).

293

These observations were supported by the measured lung index and histological

294

scoring (Fig. 3F and G). SA-2 suppressed the influenza-induced increase in the lung

295

index and histological score in a dose-dependent manner and had a significant effect

296

at a dosage of 100 mg/kg/d (lung index: 1.2, histological score: 5.3) compared with

297

the placebo group (lung index: 2.0, histological score: 9.0).

AC C

EP

TE D

M AN U

SC

RI PT

280

298

These data indicate that SA-2 significantly enhances survival, reduces body

299

weight loss, inhibits lung consolidation and reduces lung virus titer in a

300

dose-dependent manner, which demonstrate the potent therapeutic efficacy of SA-2

ACCEPTED MANUSCRIPT in vivo against infection with the A/FM/1/47 (H1N1) influenza virus.

302

3.4. Effective protection of SA-2 on mice infected with oseltamivir-resistant

303

A/FM/1/47-H275Y virus

RI PT

301

The therapeutic efficacy of SA-2 was further explored in mice infected with the

305

oseltamivir-resistant virus A/FM/1/47-H275Y. All mice treated with placebo

306

succumbed to the disease by study day 7, and oseltamivir at a dose of 100 mg/kg/d

307

for 5 days was ineffective at protecting mice from mortality (Fig. 4A). SA-2

308

administered at 75 or 100 mg/kg/d provided 30% and 50% protection, respectively,

309

from death as well as a dose-dependent reduction in body weight loss compared with

310

the placebo (Fig. 4A and B).

M AN U

SC

304

Similarly, the lungs of infected mice were harvested on day 5 post-infection and the

312

pathological changes were examined. As shown in Fig. 4C, extensive alveolar

313

damage and marked cellular infiltrates were observed in lungs of mice belonging to

314

placebo and oseltamivir groups. In contrast, all the SA-2 treatment groups (50, 75 or

315

100 mg/kg/d) showed less inflammation and fewer lesions in lung tissue. Dosing of

316

SA-2 at 100 mg/kg/d alleviated pneumonia symptoms, and the reductions in lung

317

index and histological score were significantly greater than that observed with

318

placebo or oseltamivir (Fig. 4D and E). These results suggest that SA-2 confers

319

effective protection to mice against the oseltamivir-resistant influenza virus

320

A/FM/1/47-H275Y.

321

3.5. SA-2 exhibited no obvious influence on virus entry and NA activity of the

AC C

EP

TE D

311

ACCEPTED MANUSCRIPT 322

A/FM/1/47 (H1N1) and A/Beijing/32/92 (H3N2) influenza viruses To clarify the mechanism of activity against the influenza virus, the effect of SA-2

324

on virus entry was first investigated in a cell-based assay. The results consistently

325

indicated that the inhibition rates of both the A/FM/1/47 (H1N1) and

326

A/Beijing/32/92 (H3N2) viruses were much higher when SA-2 was added 2 h

327

post-infection (that is, after the virus entry step) than when SA-2 was added

328

'pre-infection' or 'co-infection' (Fig. 5A and B). For both virus strains, inhibition

329

rates were no greater than 30% when SA-2 was added to a final concentration of 80

330

µM before or during viral inoculation, which indicated that SA-2 had no influence

331

on viral adsorption and penetration. The results from the hemagglutination assay also

332

supported this inference. All tested concentrations of SA-2 were ineffective to

333

prevent the HA-mediated agglutination of red blood cells (Fig. 5C and D). To

334

examine whether SA-2 could affect the final step of virus progeny release from

335

infected cells, the NA inhibitory activity of SA-2 was assessed using the whole

336

influenza virion. The inhibition rates of SA-2 were less than 7.0% at all

337

concentrations tested. The results clearly showed that NA enzyme activity of H1N1

338

and H3N2 was hardly inhibited by SA-2, whereas it was suppressed by the influenza

339

NA inhibitor oseltamivir (Fig. 5E and F). All of these data indicate that SA-2 has no

340

effect on receptor binding and NA activity of the influenza virus.

341

3.6. SA-2 suppressed H1N1 influenza virus replication by inhibiting the activity of

342

RNA polymerase and interfering with NP and M1 expression during viral

AC C

EP

TE D

M AN U

SC

RI PT

323

ACCEPTED MANUSCRIPT 343

biosynthesis As part of the effort to elucidate the mechanism of action, a time-of-addition study

345

was performed by exposing influenza-infected cells to SA-2 at different parts of the

346

virus replication cycle and measuring the expression of NP and M1 proteins 10 h

347

after infection. When SA-2 was added during the viral replication stage (2–4 or 4–6

348

h after infection), both NP and M1 protein levels were significantly reduced, while

349

no inhibitory activity was observed when SA-2 was added at the adsorption stage

350

(0–2 h) or at the release stage (6 or more hours after infection) (Fig. 6A and B). The

351

expression of NP and M1 genes were then examined at 6 h post-infection by

352

RT-PCR and real-time qPCR analysis. Compared with the DMSO control, SA-2

353

significantly reduced the mRNA levels of NP and M1 genes to 52.0% and 46.9%,

354

respectively (Fig. 6C and D). These results strongly suggest that SA-2 disrupts the

355

early

356

polymerase-mediated transcription and replication step.

intermediate

stage

of

viral

replication,

particularly

the

RNA

EP

to

TE D

M AN U

SC

RI PT

344

Subsequently, a luciferase experiment was conducted, and the results showed that

358

the activity of influenza virus RNA polymerase was significantly inhibited by SA-2

359

in a dose-dependent manner (Fig. 6E). SA-2 at concentrations of 40 and 80 µM

360

resulted in 68.1% and 75.0% reductions, respectively, of luciferase activity; SA-2,

361

showed a better inhibitory effect than did ribavirin (48.9% reduction at 40 µM). In

362

summary, SA-2 inhibits influenza virus replication by reducing viral polymerase

363

activity and interfering with RNA synthesis.

AC C

357

ACCEPTED MANUSCRIPT 364

4. Discussion Outbreaks and epidemics of influenza are serious threats to human health. The

366

limited number of licensed drugs together with the continual emergence of viral

367

variants and drug-resistant mutants highlight the urgent need for antiviral agents with

368

novel mechanisms of action.

RI PT

365

Because the influenza virus produces a rapid and clear CPE followed by cell death

370

in vitro (Atkins et al., 2012), we initiated an antiviral evaluation of benzoic acid

371

derivatives using a cell-based approach. Compound 2 (SA-2) was found to be a

372

potent influenza inhibitor with limited cytotoxicity. In both virus-infected MDCK

373

and A549 cell models, SA-2 effectively inhibited the replication of the A/FM/1/47

374

(H1N1) and A/Beijing/32/92 (H3N2) influenza viruses, with EC50 values of 9.6 µM

375

and 19.2 µM, respectively, levels that are comparable to those for oseltamivir. In

376

addition, oral administration of SA-2 significantly protected mice infected with a

377

lethal dose of the A/FM/1/47 (H1N1) influenza virus, conferring 70% protection

378

from death at a dosage of 100 mg/kg/d, prolonging survival time, reducing body

379

weight loss, inhibiting lung consolidation, reducing lung virus titer, and decreasing

380

lung parameters. All these data demonstrate the potent anti-influenza activity of

381

SA-2 both in vitro and in vivo.

M AN U

TE D

EP

AC C

382

SC

369

Notably, permissive mutations enable influenza A (H1N1) viruses to acquire the

383

H275Y NA resistance mutation to oseltamivir without fitness loss, resulting in their

384

rapid global spread (Hurt, 2014; Tarbet et al., 2014). In the present study, a

385

drug-resistant mutant strain (A/FM/1/47-H275Y) selected by oseltamivir was used.

ACCEPTED MANUSCRIPT SA-2 displayed a sufficient antiviral effect to treat infection with H275Y-mutated

387

virus both in vitro and in vivo. In cell culture, the oseltamivir-resistant strain

388

A/FM/1/47-H275Y was susceptible to SA-2, with an EC50 value of 19.8 µM, while

389

that for oseltamivir was as high as 1177.9 µM. SA-2 also consistently showed

390

therapeutic

391

A/FM/1/47-H275Y in mice. Although oseltamivir at 100 mg/kg/d was ineffective at

392

protecting mice from mortality, 50% of virus-infected mice survived with alleviated

393

influenza-induced lung injury when treated with a 100 mg/kg/d dosage of SA-2.

394

These results suggest that SA-2 has effective inhibitory activity toward an

395

oseltamivir-resistant influenza virus. Moreover, considering the results we obtained

396

in mechanistic studies that indicated that SA-2 targeted RNA polymerase rather than

397

NA, this compound could potentially be used to treat infections caused by NA

398

inhibitor-resistant mutants of the influenza A virus.

against

the

oseltamivir-resistant

mutant

strain

TE D

M AN U

SC

efficacy

RI PT

386

Viruses initiate their life cycle by attaching to host cell surface receptors, entering

400

cells, uncoating their viral nucleic acids, and replicating their genomes. After new

401

copies of viral proteins and genes are synthesized, these components assemble into

402

progeny virions, which exit the cell (Watanabe et al., 2010). The complete influenza

403

virus life cycle takes approximately 8 hours. The nucleoprotein (NP) and matrix

404

protein (M1) are major structural proteins that are essential for the integrity of a

405

virus (Wang et al., 2015). Their expression levels reflect the state of virus replication.

406

Consistent with results from hemagglutination and NA inhibition assays, results

407

from a time-of-addition assay suggested that the SA-2 antiviral effect may occur at

AC C

EP

399

ACCEPTED MANUSCRIPT an early to intermediate stage, particularly the RNA polymerase-mediated

409

transcription and replication step, but not at the steps of the virus entry or release.

410

Luciferase experiment, RT-PCR and real-time qPCR analysis confirmed the

411

inference that SA-2 suppressed influenza virus replication by inhibiting RNA

412

polymerase activity and interfering with gene transcription during viral biosynthesis.

413

SA-2 at concentrations of 40 and 80 µM resulted in 68.1% and 75.0% reductions,

414

respectively, of luciferase activity, which is a better inhibitory effect on RNA

415

polymerase than that of ribavirin (48.9% reduction at 40 µM). Meanwhile, the

416

mRNA levels of the NP and M1 genes were reduced to 52.0% and 46.9%,

417

respectively at 6 h post infection.

M AN U

SC

RI PT

408

Because the drug-resistance of influenza virus is frequently caused by amino acid

419

mutations in NA (e.g., R152K, H275Y, R292K) (Yen et al., 2014), inhibitors of viral

420

RNA synthesis in the influenza virus replication cycle have unique advantages

421

compared with NA inhibitors (Furuta, et al., 2009). Since SA-2 inhibits influenza

422

virus replication by reducing the viral polymerase activity and the action of point lies

423

in the phase of RNA synthesis, SA-2 should be considered a promising drug

424

candidate for the treatment of influenza, including influenza caused by existing and

425

potential drug-resistant mutants. However, based on the existing data we have

426

obtained, it is difficult to determine whether SA-2 acts directly on the viral

427

polymerase complex or the compound reduces polymerase activity indirectly by

428

interaction with host factors such as BUB3, CTNNB1 and CAMK2B, which are

429

essential for viral RNA transcription (Konig, et al., 2010; Watanabe, et al., 2015;

AC C

EP

TE D

418

ACCEPTED MANUSCRIPT Sasaki, et al., 2014). The presumed broad-spectrum antiviral activities, further

431

mechanism study, and more preclinical development of SA-2 will be the direction of

432

our future work.

433

Acknowledgments

RI PT

430

We sincerely thank Dr Xin Ye (Institute of Microbiology, Chinese Academy of

435

Sciences) for technical assistance with the luciferase assay. This work was supported

436

by the 111 Project (grant 111-2-07), the Priority Academic Program Development of

437

Jiangsu Higher Education Institutions (PAPD) and the Fundamental Research Funds

438

for the Central Universities (grant ZL2014SK0035).

439

References

440

Atigadda, V.R., Brouillette, W.J., Duarte, F., Ali, S.M., Babu, Y.S., Bantia, S.,

441

Chand, P., Chu, N., Montgomery, J.A., Walsh, D.A., Sudbeck, E.A., Finley, J.,

442

Luo, M., Air, G.M., Laver, G.W., 1999. Potent inhibition of influenza sialidase

443

by a benzoic acid containing a 2-pyrrolidinone substituent. J. Med. Chem. 42,

444

2332-2343.

446

M AN U

TE D

EP

AC C

445

SC

434

Atkins, C., Evans, C.W., White, E.L., Noah, J.W., 2012. Screening methods for influenza antiviral drug discovery. Expert Opin. Drug Discov. 7, 429-438.

447

Burgeson, J.R., Moore, A.L., Boutilier, J.K., Cerruti, N.R., Gharaibeh, D.N.,

448

Lovejoy, C.E., Amberg, S.M., Hruby, D.E., Tyavanagimatt, S.R., Allen, R.D.,

449

Dai, D., 2012. SAR analysis of a series of acylthiourea derivatives possessing

450

broad-spectrum antiviral activity. Bioorg. Med. Chem. Lett. 22, 4263-4272.

ACCEPTED MANUSCRIPT Byrn, R.A., Jones, S.M., Bennett, H.B., Bral, C., Clark, M.P., Jacobs, M.D., Kwong,

452

A.D., Ledeboer, M.W., Leeman, J.R., McNeil, C.F., Murcko, M.A., Nezami, A.,

453

Perola, E., Rijnbrand, R., Saxena K., Tsai, A.W., Zhou, Y., Charifson, P.S., 2015.

454

Preclinical activity of VX-787, a first-in-class, orally bioavailable inhibitor of the

455

influenza virus polymerase PB2 subunit. Antimicrob. Agents Chemother. 59,

456

1569 -1582.

RI PT

451

Chen, L., Dou, J., Su, Z., Zhou, H., Wang, H., Zhou, W., Guo, Q., Zhou, C., 2011.

458

Synergistic activity of baicalein with ribavirin against influenza A (H1N1) virus

459

infections in cell culture and in mice. Antiviral Res. 91, 314-320. Clarke,

D.H.,

Casals,

J.,

M AN U

460

SC

457

1958.

Techniques

for

hemagglutination

and

hemagglutination-inhibition with arthropod-borne viruses. Am. J. Trop. Med.

462

Hyg. 7, 561-573.

TE D

461

Delogu, I., Pastorino, B., Baronti, C., Nougairède, A., Bonnet, E., de Lamballerie, X.,

464

2011. In vitro antiviral activity of arbidol against Chikungunya virus and

465

characteristics of a selected resistant mutant. Antiviral Res. 90, 99-107.

467 468 469

Ding, Y., Dou, J., Teng, Z., Yu, J., Wang, T., Lu, N., Wang, H., Zhou, C., 2014.

AC C

466

EP

463

Antiviral activity of baicalin against influenza A (H1N1/H3N2) virus in cell culture and in mice and its inhibition of neuraminidase. Arch. Virol. 159, 3269-3278.

470

Fan, H., Dong, G., Zhao, G., Liu, F., Yao, G., Zhu, Y., Hou, Y., 2014. Gender

471

differences of B cell signature in healthy subjects underlie disparities in

472

incidence and course of SLE related to estrogen. J. Immunol. Res. 2014, 814598.

ACCEPTED MANUSCRIPT Fiore, A.E., Shay, D.K., Broder, K., Iskander, J.K., Uyeki, T.M., Mootrey,

474

G., Bresee, J.S., Cox, N.S., 2008. Prevention and control of influenza:

475

recommendations of the Advisory Committee on Immunization Practices (ACIP).

476

MMWR Recomm. Rep. 57, 1-60.

RI PT

473

Furuta, Y., Takahashi, K., Shiraki, K., Sakamoto, K., Smee, D.F., Barnard, D.L.,

478

Gowen, B.B., Julander, J.G., Morrey, J.D., 2009. T-705 (favipiravir) and related

479

compounds: novel broad-spectrum inhibitors of RNA viral infections. Antiviral

480

Res. 82, 95-102.

M AN U

SC

477

Guilligay, D., Tarendeau, F., Resa-Infante, P., Coloma, R., Crepin, T., Sehr, P.,

482

Lewis, J., Ruigrok, R.W., Ortin, J., Hart, D.J., Cusack, S., 2008. The structural

483

basis for cap binding by influenza virus polymerase subunit PB2. Nat. Struct.

484

Mol. Biol. 15, 500-506.

487 488 489 490 491

viruses. Curr. Opin. Virol. 8, 22-29.

Jung, E.J., Lee, K.H., Seong, B.L., 2010. Reverse genetic platform for inactivated

EP

486

Hurt, A.C., 2014. The epidemiology and spread of drug resistant human influenza

and live-attenuated influenza vaccine. Exp. Mol. Med. 42, 116-121.

AC C

485

TE D

481

Kim, J.H., Resende, R., Wennekes, T., Chen, H.M., Bance, N., Buchini, S., Watts, A.G., Pilling,

P., Streltsov,

S., McKimm-Breschkin,

J.L.,

V.A., Petric, Niikura,

M., Liggins, M., Withers,

R., Barrett, S.G.,

2013.

492

Mechanism-based covalent neuraminidase inhibitors with broad-spectrum

493

influenza antiviral activity. Science 340, 71-75.

494

Konig, R., Stertz, S., Zhou, Y., Inoue, A., Hoffmann, H.H., Bhattacharyya, S.,

ACCEPTED MANUSCRIPT Alamares, J.G., Tscherne, D.M., Ortigoza, M.B., Liang, Y., Gao, Q., Andrews,

496

S.E., Bandyopadhyay, S., Jesus, P.D., Tu, B.P., Pache, L., Shih, C., Orth, A.,

497

Bonamy, G., Miraglia, L., Ideker, T., García-Sastre, A., Young, J.A.T., Palese, P.,

498

Shaw, M.L., Chanda, S.K., 2010. Human host factors required for influenza virus

499

replication. Nature 2010, 463, 813-817.

500

RI PT

495

Leang, S.K., Deng, Y.M., Shaw, R., Caldwell, N., Iannello, P., Komadina, N., Buchy, P., Chittaganpitch,

D.E., Fagan,

P., Gourinat,

A.C., Hammill,

502

F., Horwood, P.F., Huang, Q.S., Ip, P.K., Jennings, L., Kesson, A., Kok, T., Kool,

503

J.L., Levy, A., Lin, C., Lindsay, K., Osman, O., Papadakis, G., Rahnamal,

504

F., Rawlinson, W., Redden, C., Ridgway, J., Sam, I.C., Svobodova, S., Tandoc,

505

A., Wickramasinghe, G., Williamson, J., Wilson, N., Yusof, M.A., Kelso,

506

A., Barr, I.G., Hurt, A.C., 2013. Influenza antiviral resistance in the Asia-Pacific

507

region during 2011. Antiviral Res. 97, 206-210.

TE D

M AN U

M., Dwyer,

SC

501

Li, Q., Zhou, L., Zhou, M., Chen, Z., Li, F., Wu, H., Xiang, N., Chen, E., Tang,

509

F., Wang, D., Meng, L., Hong, Z., Tu, W., Cao, Y., Li, L., Ding, F., Liu,

510

B., Wang, M., Xie, R., Gao, R., Li, X., Bai, T., Zou, S., He, J., Hu, J., Xu,

512 513

AC C

511

EP

508

Y., Chai, C., Wang, S., Gao, Y., Jin, L., Zhang, Y., Luo, H., Yu, H., He, J., Li, Q., Wang, X., Gao, L., Pang ,X., Liu, G., Yan, Y., Yuan, H., Shu, Y., Yang, W., Wang, Y., Wu, F., Uyeki, T.M., Feng, Z., 2014. Epidemiology of human

514

infections with avian influenza A (H7N9) virus in China. New Engl. J. Med. 370,

515

520-532.

516

Liu, K.C., Fang, J.M., Jan, J.T., Cheng, T.J., Wang, S.Y., Yang, S.T., Cheng, Y.S.,

ACCEPTED MANUSCRIPT 517

Wong,

C.H.,

2012.

Enhanced

anti-influenza

agents

518

anti-inflammatory activity. J. Med. Chem. 55, 8493-8501.

conjugated

with

Livak, K.J., Schmittgen, T.D., 2001. Analysis of relative gene expression data using

520

real-time quantitative PCR and the 2 (– Delta Delta C (T)) Method. Methods 25,

521

402-408.

524 525

1363-1373.

SC

523

Moscona, A., 2005. Neuraminidase inhibitors for influenza. New Engl. J. Med. 353,

Reed, L.J., Müench, H., 1938. A simple method of estimating fifty percent endpoints. Am. J. Hyg. 27, 493-497.

M AN U

522

RI PT

519

Sasaki, Y., Kakisaka, M., Chutiwitoonchai, N., Tajima, S., Hikono, H., Saito, T.,

527

Aida, Y., 2014. Identification of a novel multiple kinase inhibitor with potent

528

antiviral activity against influenza virus by reducing viral polymerase activity.

529

Biochem. Biophys. Res. Commun. 450, 49-54.

532 533 534 535

comparative review. Drugs 64, 2031-2046.

EP

531

Schmidt, A.C., 2004. Antiviral therapy for influenza: a clinical and economic

Shih, S.R., Chu, T.Y., Reddy, G.R., Tseng, S.N., Chen, H.L., Tang, W.F., Wu,

AC C

530

TE D

526

M.S., Yeh, J.Y., Chao, Y.S., Hsu, J.T., Hsieh, H.P., Horng, J.T., 2010. Pyarzole compound BPR1P0034 with potent and selective anti-influenza virus activity. J. Biomed. Sci. 17, 13-22.

536

Sun, C., Huang, H., Feng, M., Shi, X., Zhang, X., Zhou, P., 2006. A novel class of

537

potent influenza virus inhibitors: polysubstituted acylthiourea and its fused

538

heterocycle derivatives. Bioorg. Med. Chem. Lett. 16, 162-166.

ACCEPTED MANUSCRIPT 539

Takeuchi, A., Mishina, Y., Miyaishi, O., Kojima, E., Hasegawa, T., Isobe, K., 2003.

540

Heterozygosity with respect to Zfp148 causes complete loss of fetal germ cells

541

during mouse embryogenesis. Nat. Genet. 33, 172-176. Tarbet, E.B., Hamilton, S., Vollmer, A.H., Luttick, A., Ng, W.C., Pryor, M., Hurst,

543

B.L., Crawford, S., Smee, D.F., Tucker, S.P., 2014. A zanamivir dimer with

544

prophylactic and enhanced therapeutic activity against influenza viruses. J.

545

Antimicrob. Chemother. 69, 2164-2174.

SC

RI PT

542

Thorlund, K., Awad, T., Boivin, G., Thabane, L., 2011. Systematic review of

547

influenza resistance to the neuraminidase inhibitors. BMC Infect. Dis. 11, 134.

548

Wang, D., Yu, J., Zhou, Y., Zhou, C., Ding, Y., Zhao, J., Dou, J., 2015 Mar. 25.

549

Synthesis and anti-influenza application for a series of benzoic acid thiourea

550

compounds. Chinese patent CN104447481A.

TE D

M AN U

546

Wang, W., Li, R., Deng, Y., Lu, N., Chen, H., Meng, X., Wang, W., Wang, X., Yan,

552

K., Qi, X., Zhang, X., Xin, W., Lu, Z., Li, X., Bian, T., Gao, Y., Tan, W., Ruan,

553

L., 2015. Protective efficacy of the conserved NP, PB1, and M1 proteins as

554

immunogens in DNA- and vaccinia virus-based universal influenza A virus

AC C

555

EP

551

vaccines in mice. Clin. Vaccine Immunol. 22, 618-630.

556

Watanabe, T., Kawakami, E., Shoemaker, J.E., Lopes, T.J., Matsuoka, Y., Tomita,

557

Y., Kozuka-Hata, H., Gorai, T., Kuwahara, T., Takeda, E., Nagata, A., Takano,

558

R., Kiso, M., Yamashita, M., Sakai-Tagawa, Y., Katsura, H., Nonaka, N., Fujii,

559

H., Fujii, K., Sugita, Y., Noda, T., Goto, H., Fukuyama, S., Watanabe, S.,

560

Neumann, G., Oyama, M., Kitano, H., Kawaoka, Y., 2015. Influenza virus-host

ACCEPTED MANUSCRIPT 561

interactome screen as a platform for antiviral drug development. Cell Host

562

Microbe. 16, 795-805.

564

Watanabe, T., Watanabe, S., Kawaoka, Y., 2010. Cellular networks involved in the influenza virus life cycle. Cell Host Microbe. 7, 427-439.

RI PT

563

Yen, H.L., Zhou, J., Choy, K.T., Sia, S.F., Teng, O., Ng, I.H., Fang, V.J., Hu, Y.,

566

Wang, W., Cowling, B.J., Nicholls, J.M., Guan, Y., Peiris, J.S., 2014. The

567

R292K mutation that confers resistance to neuraminidase inhibitors leads to

568

competitive fitness loss of A/Shanghai/1/2013 (H7N9) influenza virus in ferrets.

569

J. Infect. Dis. 210, 1900-1908.

M AN U

SC

565

Zhang, J., Liu, T., Tong, X., Li, G., Yan, J., Ye, X., 2012. Identification of novel

571

virus inhibitors by influenza A virus specific reporter cell based screening.

572

Antiviral Res. 93, 48-54.

TE D

570

Figure Legends

574

Fig. 1. Chemical structure of benzoic acid derivatives.

575

Fig. 2. Inhibitory effect of SA-2 on influenza A virus replication in MDCK and

576

A549 cells. (A) Microscopic images for A/FM/1/47 (H1N1) virus-infected and 80

577

µM SA-2-treated MDCK cells at 48 h post-infection. DMSO was used as a vehicle

578

control. (B) Rates of SA-2 (5–80 µM) inhibition of the A/FM/1/47 (H1N1),

579

A/Beijing/32/92

580

oseltamivir-resistant) influenza viruses in MDCK cells. Oseltamivir carboxylate (OC)

581

(10 µM) was used as a positive control. (C) Inhibitory rates of SA-2 in MDCK cells

AC C

EP

573

(H3N2)

and

A/FM/1/47-H275Y

(H1N1-H275Y;

ACCEPTED MANUSCRIPT infected with the A/FM1/1/47 (H1N1) and (D) A/Beijing/32/92 (H3N2) influenza

583

viruses at different MOIs (0.3, 0.5, and 1). (E) Rates of SA-2 (5–80 µM) inhibition

584

of the H1N1, H3N2 and H1N1-H275Y influenza viruses in A549 cells. Oseltamivir

585

carboxylate (OC) (10 µM) was used as a positive control. (F) Cellular toxicity of

586

SA-2 in MDCK and A549 cells. The results were presented as the mean ± S.D. of

587

three independent experiments, *P < 0.05; **P < 0.01; ***P < 0.001, compared with

588

oseltamivir carboxylate (OC)-treated group.

589

Fig. 3. Therapeutic efficacy of SA-2 against the A/FM/1/47 (H1N1) influenza virus

590

in mice. (A) Basic experimental protocol used to test SA-2 in mice infected with

591

influenza virus. ICR mice were intranasally infected with a mouse-adapted

592

A/FM/1/47 (H1N1) influenza virus (8 × LD50). Two hours later, ten mice per group

593

received placebo, SA-2 (50, 75, and 100 mg/kg/d), or oseltamivir (100 mg/kg/d)

594

twice daily for 5 successive days (days 0 to 4) by oral gavage. (B) Survival and (C)

595

body weight were monitored daily (n = 10). On day 5 post-infection, five mice per

596

group were euthanized for lung pathological examinations. (D) H&E staining of

597

sectioned lungs (n = 5). Some of the histopathological changes are indicated by

598

arrows in pathological sections. (E) Lung virus titers, (F) Lung indices and (G)

599

histological scores for mice infected with A/FM1/1/47 (H1N1) influenza virus (n =

600

5). Data are represented as the mean ± S.D. *P < 0.05; **P < 0.01; ***P < 0.001,

601

compared with the placebo group.

602

Fig. 4. Protection of SA-2 in mice infected with oseltamivir-resistant strain

603

A/FM/1/47-H275Y (H1N1-H275Y). Ten mice per group were intranasally infected

AC C

EP

TE D

M AN U

SC

RI PT

582

ACCEPTED MANUSCRIPT with the oseltamivir-resistant strain A/FM/1/47-H275Y (H1N1-H275Y) (8 × LD50).

605

Two hours post-infection, mice were administered placebo, SA-2 (50, 75, and 100

606

mg/kg/d), or oseltamivir (100 mg/kg/d) twice daily for 5 successive days by oral

607

gavage. (A) Survival and (B) body weight were monitored daily (n = 10). On day 5

608

post-infection, five mice per group were euthanized for lung pathological

609

examinations. (C) H&E staining of sectioned lungs (n = 5). Some of the

610

histopathological changes are indicated by arrows in pathological sections. (D) Lung

611

indices and (E) histological scores for mice infected with the A/FM/1/47-H275Y

612

influenza virus (n = 5). Data are presented as the mean ± S.D. *P < 0.05; **P < 0.01,

613

compared with the placebo group.

614

Fig. 5. SA-2 exhibited no obvious influence on virus entry and NA activity of the

615

A/FM/1/47 (H1N1) and A/Beijing/32/92 (H3N2) influenza viruses. (A) Inhibition

616

rates of SA-2 (10–80 µM) against H1N1 and (B) H3N2 influenza viruses with

617

different infection protocols in MDCK cells. (C) Hemagglutination assays of various

618

SA-2 concentrations (10, 40, and 80 µM) with 4 HAU/well of H1N1 and (D) H3N2

619

viruses. DMSO was used as a vehicle control. (E) Neuraminidase (NA) inhibition

620

assays of SA-2 (0.01–100 µM) against H1N1 and (F) H3N2 viruses. Oseltamivir

621

carboxylate (OC) (1 nM) was used as a positive control.

622

Fig. 6. SA-2 suppressed H1N1 influenza virus replication by inhibiting virus RNA

623

polymerase and interfering with NP and M1 expression during viral biosynthesis. (A)

624

MDCK cells were infected with 1.5 × 103 TCID50 of A/FM/1/47 (H1N1) influenza

625

virus. SA-2 (40 µM) or ribavirin (40 µM) was added at the indicated periods. The

AC C

EP

TE D

M AN U

SC

RI PT

604

ACCEPTED MANUSCRIPT cell lysates were harvested 10 h post-infection and subjected to western blot with NP

627

and M1 antibodies. (B) The relative amounts of NP and M1 proteins were

628

represented by band intensities normalized to β-actin. The relative intensity was

629

calculated relative to the DMSO control. (C) MDCK cells were infected with 1.5 ×

630

103 TCID50 of A/FM/1/47 (H1N1) influenza virus for 2 h to allow adsorption. Then,

631

the media was replaced with fresh media with or without 40 µM SA-2. Cells were

632

collected 6 h post-infection, and total RNA was extracted. Then, the mRNA

633

expression levels of NP and M1 were determined using specific primers in a

634

RT-PCR assay. (D) The relative RNA levels of NP and M1 genes were determined

635

in a real-time qPCR experiment. Each sample was run in triplicate normalized to

636

GAPDH. The relative RNA levels of NP and M1 genes were calculated based on the

637

2

638

with PB1, PB2, PA, NP (A/WSN/33 (H1N1)), vNS-Luc and pCMV β-gal plasmids

639

for 6 h, then SA-2 (10, 40, and 80 µM) or ribavirin (40 µM) was added. After 24 h,

640

the cell lysates were harvested for use in the luciferase assay, and their activities

641

were normalized to β-gal activity. The relative luciferase activity was calculated

642

relative to the DMSO control. The results are shown as the mean ± S.D. of three

643

independent experiments, *P < 0.05; **P < 0.01; ***P < 0.001, compared with the

644

DMSO control.

method and relative to the DMSO control. (E) 293T cells were transfected

AC C

EP

TE D

△△Ct

M AN U

SC

RI PT

626

ACCEPTED MANUSCRIPT 1

Table 1

2

Structures, cytotoxicities and anti-influenza virus activities of newly synthesized

3

benzoic acid derivatives.

Compound

CC50a (µM)

R

A/FM/1/47 (H1N1)

RI PT

EC50b (µM) for influenza virus (SIc) A/Beijing/32/92 (H3N2)

Wild-type

H275Y

>500

>160

>160

2

444.5

9.6 (46.3)

19.8 (22.4)

3

>500

72.2

4

398.2

96.2

5

>500

6

>500

Oseltamivir carboxylate

>500

Ribavirin

56.3

>160

19.2 (23.2)

SC

1

132.4

82.3

>160

85.3

128.6

141.4

142.2

>160

>160

7.6

1177.9

11.7

8.8

10.2

NDd

TE D

M AN U

73.2

4

Results were the mean values from three independent assays.

5

a

6

by 50%.

7

b

8

virus-infected cells by 50%.

9

c

Selectivity index, CC50 / EC50.

10

d

Not determined.

EP

Concentration of chemicals required to reduce the viability of normal MDCK cells

AC C

Concentration of chemicals required to improve the viability of influenza

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIPT 

Novel benzoic acid derivatives were designed and synthesized with antiviral activity against influenza A viruses.



SA-2 (compound 2) significantly inhibited H1N1 and H3N2 in cell culture with



SA-2 showed potent therapeutic efficacy against the oseltamivir-resistant strain H1N1-H275Y both in vitro and in vivo.

SA-2 efficiently inhibited the activity of RNA polymerase during viral

EP

TE D

M AN U

SC

biosynthesis by interfering with gene transcription.

AC C



RI PT

EC50 values of 9.6 and 19.8 µM respectively.

vivo and its inhibition of RNA polymerase.

A target-free and cell-based approach was applied to evaluate the anti-influenza properties of six newly synthesized benzoic acid derivatives. SA-2, t...
565B Sizes 1 Downloads 11 Views