<<

AAC Accepted Manuscript Posted Online 16 May 2016 Antimicrob. Agents Chemother. doi:10.1128/AAC.00282-16 Copyright © 2016, American Society for Microbiology. All Rights Reserved.

1 Discovery of a broad-spectrum antiviral compound that inhibits biosynthesis

2 and establishes a type 1 interferon-independent antiviral state

3

4 Dong-Hoon Chung,a,b # Jennifer E. Golden,c,* Robert S. Adcock,b Chad E. Schroeder,c

5 Yong-Kyu Chu,b Julie B. Sotsky,b Daniel E. Cramer,b Paula M. Chilton,a,† Chisu Song,d

6 Manu Anantpadma,e Robert A. Davey,e Aminul I. Prodhan,f Xinmin Yin,f Xiang Zhangf

7

8 Departments of Microbiology and Immunology a, and Center for Predictive Medicine for

9 Biodefense and Emerging Infectious Diseases b, University of Louisville, KY, USA;

10 University of Kansas Specialized Center, Lawrence, KS, USAc; Division of

11 Infectious Diseases and Northwestern HIV Translational Research Center, Department

12 of Medicine, Northwestern University Feinberg School of Medicine, IL, USAd; Texas

13 Biomedical Research Institute, San Antonio, TX, USAe; CREAM Center, University of

14 Louisville, KY, USA f.

15

16 # Address correspondence to Dong-Hoon Chung, [email protected]

17 * Current address: School of Pharmacy, 777 Highland Ave., University of Wisconsin-

18 Madison, Madison, WI, USA

19 † Current address: Christine M. Kleinert Institute for Hand & Microsurgery, 225

20 Abraham Flexner Way, Louisville, KY, USA 21 Abstract

22 Viral emergence and re-emergence underscores the importance of the developing

23 efficacious, broad-spectrum antivirals. Here we report the discovery of

24 tetrahydrobenzothiazole-based compound 1, a novel, broad-spectrum antiviral lead,

25 which was optimized from a hit compound derived from a CPE-based antiviral screen

26 using Venezuelan equine encephalitis virus. Compound 1 showed antiviral activity

27 against a broad-range of RNA viruses including alphaviruses, flaviviruses, influenza

28 virus, and ebolavirus. Mechanism of action studies with metabolomics and molecular

29 approaches revealed that the compound inhibits host pyrimidine synthesis, and

30 establishes an antiviral state by inducing a variety of interferons-stimulated genes (ISG).

31 Notably, the induction of the ISGs by compound 1 was independent of the production of

32 type 1 interferons. The antiviral activity of 1 was cell-type dependent with a robust effect

33 observed in human cell lines and no observed antiviral effect in mouse cell lines. We

34 herein disclose tetrahydrobenzothiazole 1 as a novel lead for the development of a

35 broad-spectrum, antiviral therapeutic and as a molecular probe to study the mechanism

36 of the induction of ISGs independent of type 1 interferons.

37 38 Introduction

39 Despite the economic and healthcare burden posed by viral infections, current

40 treatments for associated diseases are limited mostly to prophylactic vaccines. Only a

41 small number of viral diseases (e.g., Human immunodeficiency virus and Hepatitis C

42 virus, HCV) can be treated with virus-specific therapeutics (e.g., ) (1, 2) .

43 These agents, so-called direct acting antivirals (DAA), target viral gene products for

44 their activities. In general, DAAs are prone to develop resistant mutants and have a

45 narrow antiviral spectrum. Given the emergence of new viruses and the rapid spread of

46 emerging viral diseases to previously unaffected geographic areas, there is an urgent

47 need for the identification of agents that more efficiently target a broad range of viral

48 diseases, which DAA approaches may not be able to deliver in time.

49 While broad-spectrum antivirals may overcome these limitations, the development of

50 these agents has been hindered due to low efficacy or undesirable toxic effects, which

51 are intrinsic characteristics of most broad-spectrum antivirals. For example, has

52 been studied since 1972 and tested against many RNA viruses; however, its useful

53 antiviral spectrum is relatively narrow(3). Many RNA viruses, such as alphaviruses, are

54 not susceptible to ribavirin, and patients may not benefit from the treatment due to its

55 limited therapeutic window (4-6). T-705, a RNA-dependent RNA polymerase inhibitor,

56 was also reported with antiviral activity against a variety of RNA viruses. It is under

57 development as a therapeutic candidate; however, its potency (IC50 value) falls in the

58 few hundred micromolar range for most viruses, with the exception of influenza viruses

59 (7). 60 Previously, we reported the discovery of new anti-Venezuelan equine encephalitis virus

61 (VEEV) inhibitors from a high-throughput screening (HTS) campaign(8). VEEV is an

62 RNA virus that causes encephalitis in humans and equids, and effective therapeutics for

63 the disease have not yet been developed. We screened a library of 348,000 small

64 compounds with a cell-based measuring protection of cells from VEEV-

65 induced cytopathic effect (strain TC-83) and discovered five active compounds ('hits')

66 with EC50 values better than 15 μM. One of these hits and the resulting optimized lead,

67 ML336, turned out to be a DAA that inhibits viral RNA synthesis by targeting the amino

68 terminal domains of viral nonstructural proteins 2 and 4 (8, 9).

69 In this current study, we investigated whether the HTS had identified a broad-spectrum

70 antiviral inhibitor. Since the screen was based on a functional read-out, i.e. reduction in

71 virus-induced cell death, we hypothesized that the screen could identify a broad-

72 spectrum antiviral compound as well. Indeed, we found that one of our hit compounds,

73 CID 847035, did show antiviral effects in many cell-based antiviral assays, including

74 Marburg virus assay (http://pubchem.ncbi.nlm.nih.gov/assay/assay.cgi?aid=540276),

75 Lassa virus assay (http://pubchem.ncbi.nlm.nih.gov/assay/assay.cgi?aid=540256) and

76 RSV assay (http://pubchem.ncbi.nlm.nih.gov/assay/assay.cgi?aid=2391). Based on

77 these observations, we undertook a study aimed at the development of novel, broad-

78 spectrum antiviral inhibitors based on hit compound CID 847035 and the mechanism of

79 action underlying their activity against multiple viruses.

80 Herein we present the broad-spectrum antiviral behavior and mechanism of

81 tetrahydrobenzothiazole 1 (Figure 1), an analogue of our primary hit compound, CID 82 847035. Using metabolomics and genomics approaches, we found that compound 1

83 inhibits pyrimidine biosynthesis and establishes an antiviral state by activating genes

84 involved in the innate immunity, including -inducible gene I protein (RIG-I,

85 encoded by DDX58), interferon-induced protein with tetratricopeptide repeats 1(ISG56,

86 encoded by IFIT1) and 2'-5'-oligoadenylate synthetase-like (OASL, encoded by OASL)

87 genes. Interestingly, the antiviral status induced by compound 1 was independent of

88 type 1 interferons (IFNs) or exogenous RNA. Importantly, we also determined that

89 mouse cell lines are not capable of establishing antiviral status when treated with

90 compound 1.

91 92 Materials and Methods

93 Cells and viruses : Vero 76 (ATCC CRL-1587), BHK (ATCC CCL-10), HEp-2 (ATCC

94 CCL-23), Neuro 2A (ATCC CCL-131), SH-SY5Y(ATCC CRL-2266), MRC-5 (ATCC

95 CCL-171) and HEK 293T (ATCC CRL-3216) were obtained from ATCC and maintained

96 in Minimum Essential Medium with Earl's modification (MEM-E) containing 10% fetal

97 bovine serum (FBS) and 1X GlutaMAX (Gibco 35050-061) at 37 °C with 5% CO2.

98 MDCK (Sigma-Aldrich 84121903), RD (ATCC CCL-136) and NIH 3T3 (ATCC CRL-

99 1658) were maintained in Dulbecco's modified Eagle's medium (DMEM) with 10% FBS

100 and 2 mM L- at 37 °C with 5% CO2. TZM-bl cells were obtained through the

101 NIH AIDS Research and Reference Reagent Program from John C. Kappes, Xiaoyun

102 Wu, and Tranzyme, Inc. The TZM-bl indicator cell line, used for infectivity assays of

103 HIV-1, is a genetically engineered HeLa cell clone expressing CD4, CXCR4, CCR5, and

104 Tat-responsive firefly luciferase and Escherichia coli β-galactosidase under the control

105 of an HIV-1 long terminal repeat. TZM-bl cells were cultivated in DMEM (containing 4.5

106 g/liter , L-glutamine, and sodium pyruvate) medium with 10% fetal calf serum,

107 50 IU/ml penicillin, and 50 μg/ml at 37 °C with 5% CO2.

108 VEEV TC-83 (lyophilized vaccine from USAMRIID) and V3526 were amplified in and

109 titrated in BHK-21cells. V3526-luc was rescued from the BHK cells transfected a full-

110 length viral RNA derived from pV3526-luc as described elsewhere(10). VEEV TrD (gift

111 from Dr. R. Tesh, World Reference Center for Emerging Viruses and Arboviruses),

112 herpes simplex virus type-2 (HSV-2, gift from Dr. Steinbach, University of Louisville)

113 were grown in Vero 76 cells that were maintained in Dulbecco’s-modified essential 114 media (DMEM) with 10% FBS. Lymphocytic Choriomeningitis Virus ARM strain (LCMV-

115 ARM) was a gift from Dr. Lukashevich (University of Louisville). Chikungunya virus S-27

116 (BEIR NR-13220), West equine encephalitis virus California (WEEV California, ATCC

117 VR-70), Japanese Encephalitis virus SA14 ( BEIR NR-2335), West Nile virus NY-99

118 (gift from Dr. R. Tesh, World Reference Center for Emerging Viruses and Arboviruses),

119 Yellow Fever virus 17D (BEIR VR-1506) were grown in Vero76 cells in a virus infection

120 media (MEM-E with 10% FBS, 1X GlutaMAX and 25 mM of HEPES pH 7.3). Human

121 enterovirus D71 (strain MP4, BEIR NR-472) and human encephalomyocarditis virus MM

122 (BEIR NR-19846) were amplified in RD cells maintained in DMEM with 10% FBS.

123 To generate wild-type HIV-1 virus HEK293T cells were plated at a density of 6 x 106

124 cells/100 mm culture dish 24 hours prior to transfection. Cells were transfected with 10

125 μg of wild-type HIV-1 proviral clone, pNL4.3, using linear polyethylenimine (PEI; 25 kDa;

126 Polysciences, Inc.), as describe elsewhere (11). Culture supernatants were collected 48

127 hours after transfection and cellular debris was removed by filtration through a 0.45 μm

128 filter. Viral p24 was quantitated using standard p24 ELISA.

129

130 Dose-response studies EC50 and CC50 were evaluated in a dose-response format

131 starting from 50 µM by a 2-fold dilution, triplicates for each, in a 96-well format. For a

132 CPE-based assay, cells were seeded in white well plates at a cell density of 12,000

133 cells per well in a volume of 45 microliters and incubated in an actively humidified

134 incubator with 5.0% CO2 at 37 °C and 95% humidity for 18 hours. Test compounds

135 diluted in thirty microliters of cell culture media was added to each well. After a two- 136 hours incubation at 37 °C with a 5% CO2, a 600 pfu of virus (or cell culture media for

137 cytotoxicity assay) was added to the wells in a volume of fifteen microliters then

138 incubated two days in an actively humidified incubator with 5.0% CO2 at 37 °C and 95%

139 humidity. Cell viability was measured with 90 microliter per well of CellTiter-Glo™

140 reagent (Promega). Vero 76 cells were used for alphaviruses and RD cells were used

141 for HEV and EMCV assays. For a luciferase-tagged virus assay (i.e., V3526-luc and

142 pVSV-luc), an optimized amount of virus and cell numbers were used for each cell line

143 tested. For HEK 293T, Neuro 2A and SH-SY5Y cells, 24,000 cells and 2,400 TCID50

144 units of virus per well was used. For Vero 76 and BHK cells, 12,000 cells and 1200

145 TCID50 units of virus per well was used. For NIH3T3, 24,000 cells and 20,000 TCID50

146 units of virus per well was used. After an 18-hours incubation with virus, the plates were

147 developed with Bright-Glo™ reagent (Promega) and the luciferase activity was

148 measured as a readout for the virus replication.

149 For measurement of inhibition of Ebolavirus infection, a recombinant Ebolavirus with a

150 green fluorescent protein (GFP) gene inserted into the genome (Ebola-eGFP virus, a

151 kind gift of Dr. Heinz Feldmann) was used(12). The virus stock was generated by

152 infecting Vero E6 cells with Ebola-eGFP virus followed by pelleting the culture

153 supernatant through a 20% cushion. HeLa cells were pretreated for 2 hour with

154 two fold-dilutions of 10 to 0.005 µM of compound and incubated with virus for 24 hours

155 in the presence of the compound. Fixed cells were imaged by a fluorescent microscope

156 . Total and infected cells were counted by Cell Profiler image analysis software (Broad

157 Institute, MIT, Boston, MA), detecting nuclei stained with DAPI and virus encoded GFP 158 expression(13). This work was performed in a biosafety level 4 (BSL4) laboratory at

159 Texas Biomedical Research Institute.

160

161 Titer reduction assay To measure virus titer reduction, 12-well plates with 100,000

162 Vero 76 cells grown overnight were pre-treated with compound diluted in virus infection

163 medium at 37 °C with 5% CO2 for 8 hours, unless denoted. For virus adsorption, cell

164 plates were incubated on ice for a 15 min, and then the cell culture supernatant was

165 removed. Virus diluted at an MOI of 0.05 (or 3 for a time of addition study) in 250

166 microliter of virus infection medium was added to the cell and the virus was allowed to

167 absorb to the cells on ice for one hour. The unabsorbed virus was washed with 1 mL of

168 PBS once and the wells were replenished with virus infection medium with 5 µM of

169 ML416 or DMSO (0.25% vol/vol). The progeny virus was harvested after 26 hours for

170 VEEV, CHIKV, WEEV, HSV-2 or Influenza virus; 48 hours for JEV, YFV17D or WNV; 72

171 hours for RSV or LCMV virus. The progeny viruses in the supernatants were

172 enumerated by using either virus infection center assay or TCID50 assays. Virus

173 infection center assay was done with Vero76 cells grown confluent in 24-well plates.

174 The cells were infected with 167 µL of the serially diluted virus samples for one hour at

175 37 °C with 5% CO2. Wells were washed with PBS and replenished with virus infection

176 medium with 0.75% methylcellulose. Three or four days after virus infection, virus

177 infection centers were visualized with staining (0.2% crystal violet, 4%

178 paraformaldehyde, and 10% ). For influenza virus and HSV-2, a TCID50 assay

179 used for the titration. 180 HIV-1 Viral Infectivity Assay TZM-bl indicator cells were plated at a density of

181 10,000 cells/well in a 96 well culture plate 24 hours prior to HIV-1 infection and

182 incubated at 37 °C (5% CO2). On the day of infection, the culture medium was removed,

183 and the cells were inoculated in triplicate with 100 μL of 2-fold serial dilutions of viral

184 supernatants in culture medium containing 20 μg/mL DEAE-dextran and incubated at 37

185 °C (5% CO2). At the time of infection DMSO as a control or 10μM ML461 were added to

186 test effect of the compound on HIV-1 infection. After a 24 hours of incubation, culture

187 medium was removed from each well and replaced with 100 μL of Britelite Plus

188 luciferase assay substrate (PerkinElmer). Following 5 min of incubation at room

189 temperature, 70 μL of each cell lysate was transferred to a 96-well OptiPlate 96

190 (PerkinElmer) and luminescence was measured in a VICTOR X2 Multilabel Reader

191 (PerkinElmer). Relative infectivity was calculated by plotting luciferase activity of viral

192 particle with treated DMSO as 100 %.

193

194 Microarray

195 HEp-2 cells were treated for 18 hours either with 5 μM of CID 70698683 or with DMSO

196 for the control in cell culture medium. 100 ng total RNA was amplified and labeled

197 following the Affymetrix (Santa Clara, CA) standard protocol for their 3’IVT Plus

198 Labeling Kit, followed by hybridization to Affymetrix’ Primeview® Human Gene

199 Expression arrays. The arrays were processed following the manufacturer

200 recommended wash and stain protocol on an Affymetrix FS-450 fluidics station and

201 scanned on an Affymetrix GeneChip® 7G scanner using Command Console 4.0 . The 202 resulting .cel files were imported into Partek Genomics Suite 6.6 and transcripts were

203 normalized and summarized using RMA as normalization and background correction

204 method. A 1-way ANOVA was set up to compare the treatment of 5 μM of

205 CID:70698683 to the control. False Discovery Rate (FDR) was chosen as multiple test

206 correction for the resulting p-values.

207

208 RealTime PCR Total RNAs from cells in a 12-well plate were isolated with RNAzol ®

209 RT (Molecular Research Center, Inc) reagent as per the manufacturer’s protocol and

210 were dissolved in 50 μL of The RNA storage solution (Life Technologies). One

211 microgram of RNA samples were subjected to a cDNA synthesis with Maxima™ H

212 Minus Reverse Transcriptase (Life Technologies), random hexamers, and oligo-dT by

213 following the manufacturer’s protocol. For quantitation of gene expression, we used a

214 real-time PCR with 2(-Delta Delta C(T)) method in a total of twenty microliters per well

215 with 2 μL of 2-fold diluted cDNA mixture in a multiplex mode in conjunction with TaqMan

216 chemistry. Information on the primers and probes are described in Supplementary

217 Information (Table S2). The copy number of viral genome was quantitated as described

218 elsewhere(8). 18S rRNA (Cat. No. 4319413E, Life Technologies) and human GAPDH

219 (Cat. No. 4326317E Life Technologies) were used as the endogenous controls to

220 quantitate the relative viral and human gene RNA copy numbers, respectively. Three

221 biological replicates, each with two technical replicates, were used for the quantitation.

222 223 Interferon assays Interferon-α and -β in the cell culture supernatant were detected by

224 using LumiKine™ Xpress hIFN-α and LumiKine™ hIFN-β kits (InvivoGen) as per the

225 manufacturer’s protocol. One-day-old HEK 293T cells cultured in a 12-well plate were

226 treated with 5 µM of compound 1 or DMSO in virus infection medium for 18 hours and

227 the cell culture supernatants were harvested and cleared by centrifugation at 3000 x g

228 for 10 min. For each treatment, six replicates were used per group and three technical

229 replicates were used for the controls, HEK293-expressed human IFN-α2 and CHO-

230 expressed human IFN-β.

231 HEK-Blue™ IFN-α/β cell reporter assay (InvivoGen) was done followed by the

232 manufacturer’s protocol. One-day-old HEK-Blue™ IFN-α/β cells plated in a 96-well plate

233 were treated with two fold-dilutions of 50 to 0.4 µM of compound 1 or DMSO for 24

234 hours. The expression of SEAP under the control of ISRE9, which is activated by Type

235 1 interferons, was measured by the absorbance at 620 nm after 20 min incubation with

236 100 µL of SEAP substrate. Three replicates were used for each data points. 237 Metabolomics analysis Metabolites were extracted from cell samples using a solvent

238 mixture of , water, and (2:1.5:1 by volume). The metabolite

239 extract from each sample was then derivatized using N-methyl-N-(trimethylsilyl)

240 trifluoroacetamide (MSTFA). All derivatized samples were further analyzed on a LECO

241 Pegasus 4D GC×GC–TOF MS instrument (St. Joseph, MI). The instrument data were

242 first processed using LECO’s instrument control software ChromaTOF for peak picking

243 and tentative metabolite identification. MetPP software was used for retention index

244 matching, peak merging, peak list alignment, normalization, and statistical significance

245 test (14). The abundance test was performed using pairwise two-tail t-test with sample

246 permutation, to standardize the abundance variation of each metabolite between

247 sample groups. The presence-absence test was performed using Fisher’s exact test.

248 249 Results

250 Discovery of tetrahydrobenzothiazole 1

251 Our lead, compound 1, resulted from a rigorous medicinal chemistry optimization effort

252 focused on a hit compound (CID 847035) that was prioritized from our high-throughput

253 screen measuring the protection of Vero 76 cells from a VEEV- induced CPE (Figure 1A

254 and 1B) (8). Medicinal chemistry optimization of the this hit compound, which will be

255 described separately in due course, was executed in parallel to the development of a

256 structurally- and mechanistically-distinct anti-VEEV probe, ML336. Unlike ML336,

257 compound 1 showed a potent antiviral effect towards multiple alphaviruses with

258 cytopathic effect (CPE) EC50 values of 0.35 µM, 0.29 µM, and 0.96 µM for VEEV TC-83,

259 CHIKV S27, and WEEV, respectively (Table 1).

260 To verify the antiviral activity of compound 1 against multiple alphaviruses, we

261 employed various assays with different read-outs. In a luciferase-based anti-VEEV

262 assay (V3526-luc,Table 1), compound 1 showed promising antiviral activity with an EC50

263 of 0.17 µM. In a quantitative real-time PCR (qRT-PCR) assay with VEEV TC-83 strain

264 (Figure 1C), compound 1 (20 μM) decreased viral RNA copy numbers greater than

265 1000-fold compared to the control. Finally, we employed a titer reduction assay with

266 variety of alphaviruses to confirm the antiviral activity of compound 1. For VEEV TC-83,

267 the progeny virus production decreased by greater than 15,000-, 2,600-, or 340-fold at

268 20, 10, or 5 μM, respectively (Figure 1D). VEEV TrD and CHIKV S17 strains were more

269 sensitive to compound 1. At a concentration of 5 μM, the compound decreased the level

270 of viral replication more than 10,000-fold (4 Log) for the both viruses. From these 271 experiments, we concluded that tetrahydrobenzothiazole 1 potently inhibits multiple

272 alphaviruses.

273

274 Time of addition assay

275 To understand at what stage of virus replication compound 1 exerted its antiviral

276 activity, we employed a time of addition assay with Vero76 cells synchronously infected

277 with TC-83 at multiplicity of infection (MOI) of 3 (Figure 1E). Compound 1 was added to

278 cells at various time-points in regards to the virus replication cycle and maintained until

279 the progeny virus was harvested. The experiment showed that pre-treatment was

280 necessary in order to observe full antiviral activity. Treatment at time 0, right after the

281 virus adsorption to cells on ice, resulted in only 65% reduction in progeny virus titer;

282 however, pre-treatment for six hours resulted in ~ 400-fold reduction in virus titer.

283 Antiviral efficacy of compound 1 was dependent on the length of pre-treatment time

284 between 4 and 0 hours prior to infection. This result clearly shows that compound 1

285 requires the induction of a cellular response for its activity.

286

287 Cytotoxicity of tetrahydrobenzothiazole 1

288 The cytotoxicity of compound 1 was evaluated, as a cytotoxic compound may

289 misleadingly exhibit broad-spectrum antiviral activity. Vero76 cells were plated in 96-

290 well plates and incubated 48, 72, and 96 hours in the presence of 1 at various 291 concentrations. Compound 1 did not show apparent cytotoxicity up to 12.5 μM (Figure

292 S1 A). The CC50 values were 74.1, 31.0 and 34.6 μM after 2, 3, and 4 days exposure

293 respectively, resulting in a selective Index 50 (SI50) greater than 100 (CC50 at Day3/

294 EC50 of CHIKV = 106.9). This indicated that the agent is not toxic to cells at the effective

295 concentrations, and that the inhibition of virus replication is not be due to a non-specific

296 cytotoxicity. Cells treated with compound 1, however, looked larger and more stretched

297 compared to the control group under visual observation. The compound was further

298 tested in viability, cytotoxicity and/or apoptosis assays with HEK 293T cells for four

299 days. No significant cytotoxicity or apoptosis was induced with up to 25 μM of

300 compound 1 (Figure S1 B).

301

302 Antiviral spectrum of tetrahydrobenzothiazole 1

303 As compound 1 showed similar antiviral effects on all alphaviruses we tested, we

304 questioned whether the compound could inhibit a broader-spectrum of viruses. To

305 address this question, we tested compound 1 against various viruses including:

306 Vesicular stomatitis virus (pVSV-luc), Respiratory syncytial virus (RSV), ebolavirus

307 (EBOV), HIV-1, Japanese Encephalitis virus (JEV), Herpes simplex virus type-2 (HSV-

308 2), Yellow Fever virus 17D(YFV 17D), West Nile virus (WNV), Influenza virus (IFNV),

309 human enteroviruses 71 (EV-71), and Encephalomyocarditis virus (EMCV).

310 As shown in Table 1, compound 1 showed an antiviral effect against a broad-spectrum

311 of viruses with different sensitivity to each virus. Alphaviruses, LCMV, and IFNV were 312 most sensitive. Treatment with compound 1 (5 µM) resulted in a greater than 4 Log

313 reduction in the progeny virus titers of alphaviruses tested, and greater than 3 Log

314 reduction for IFNV and LCMV. The RSV, WNV, YFV 17D viruses were sensitive as well

315 with ~ 2 Log titer reduction; however, JEV was less sensitive than the others tested

316 (~1.3 Log reduction). HIV-1 infection was sensitive as well, with 94% reduction in viral

317 infectivity using TZM-bl reporter system measuring luciferase activity with 10 μM

318 concentration of compound 1. Interestingly, the replication of HSV-2 was not inhibited

319 by 1, resulting in no changes in progeny virus titer.

320 The compound’s antiviral activity was also assessed using a dose-response format

321 (EC50 determination). The replication of pVSV-luc or EBO-GFP was sensitive to the

322 compound treatment with EC50 values of 0.19 μM and 0.30 μM, respectively. The agent,

323 however, did not inhibit the replication of the picornaviruses efficiently, (i.e. EC50 values

324 of EV D71 and EMCV MM were 11.1 μM and > 25 μM, respectively). From these

325 experiments, we found that compound 1 is a broad-spectrum antiviral effect with

326 different sensitivities depending on virus. This result implies that the antiviral

327 mechanism of compound 1 has specificity to certain type of viruses.

328

329 Pyrimidine synthesis inhibition by tetrahydrobenzothiazole 1

330 To test if our lead compound could interfere with cellular metabolism for its antiviral

331 activity, we measured the difference in cellular metabolites using a metabolomics

332 approach. HEK 293T cells were treated either with 5 μM of compound 1 or with DMSO 333 for 18 hours and the cellular metabolites were analyzed and compared using GC-MASS

334 spectrophotometry. The treatment with compound 1 changed the abundance of cellular

335 metabolites significantly (Table S1). For example, the amounts of L- and

336 differed by 0.09- and 11-fold when compound 1-treated group was

337 compared to mock-treated cells. For some metabolites, the difference was more evident

338 (Table 2). For example, dihydroorotic acid (DHO) and were found only in

339 compound 1-treated groups. DHO and orotic acid are metabolites that are related with

340 de novo pyrimidine synthesis, suggesting that compound 1 inhibits pyrimidine synthesis

341 after the synthesis of orotic acid from DHO. Consistent with this result, free was

342 not detected in the compound 1-treated groups.

343 To validate this finding, we tested to see if the antiviral effect of compound 1 could be

344 overcome by the addition of exogenous . We measured the virus replication

345 in HEK 293T cells treated with (MPA) or compound 1 in the

346 presence of various exogenous nucleosides, and then compared the virus replication to

347 the controls (Figure 2). In MPA-treated groups, the antiviral effect of MPA was

348 completely reversed when was supplemented in the culture: from 47% to

349 105% and 17% to 62% for pVSV-luc and V3526-luc, respectively. This result was

350 consistent with our expectation that MPA inhibits -5′-monophosphate

351 dehydrogenase (IMPDH) as its antiviral mechanism, which is a key enzyme to

352 synthesize guanosine de novo. Similarly, the antiviral effect of compound 1 greatly

353 decreased when the cells were supplemented with pyrimidines, or uridine. For

354 example, while 5 μM of compound 1 decreased the replication of V3526-luc to 1.9% 355 compared to the control, the addition of cytidine or uridine restored the viral replication

356 83.4 or 77.5 % compared to the control, respectively.

357 Two pyrimidine synthesis intermediates, DHO and orotic acid, were also tested to

358 confirm the metabolomics results with the accumulation of the in the

359 compound 1-treated group. The addition of DHO did not affect the antiviral activity of

360 compound 1 at all, suggesting that compound 1 inhibits a downstream step from DHO.

361 Orotic acid showed a moderate reversion effect to compound 1 (21.9% to 55.0% and

362 1.9% to 7.1% for pVSV-luc and V3526-luc, respectively).

363

364 Induction of Innate immune genes by tetrahydrobenzothiazole 1 without virus

365 infection or type 1 interferons

366 It has been reported that the inhibition of pyrimidine biosynthesis could amplify the

367 cellular response to ssRNA via type 1 IFN system (15) ; however, we questioned this as

368 Vero 76, the primary cell line for the antiviral activity testing for compound 1 and its

369 derivatives, is known to be deficient in type 1 IFN production(16).

370 To test if compound 1 induced cellular immune response without type 1 IFN, we

371 examined the changes in host gene expression after the treatment with CID 70698683

372 without addition of ssRNA. CID70698683 is an N-phenyl, derivative of

373 compound 1 with similar antiviral effect towards multiple alphaviruses with CPE EC50

374 values of 0.60 µM, 0.66 µM, and 0.93 µM for VEEV TC-83, CHIKV S27, and WEEV,

375 respectively. Human HEp-2 cells were treated with 5 µM of CID 70698683 or DMSO for 376 18 hours and then the cellular mRNAs were subjected to a DNA microarray assay.

377 Ninety-two genes were up-regulated and one hundred forty five genes were down-

378 regulated by greater than two-fold difference (GEO accession ID: GSE72167). Among

379 these changes, certain sets of genes that are involved in the interferon pathways were

380 clearly upregulated (Figure 3), including RIG-I (4.63-fold) and OASL (3.76-fold

381 increase). We found that some ISGs (e.g., GBP2, ISG20, IFI44, IRF9 or IFIT1) were

382 also upregulated significantly (3.5, 2.85, 2.83, 2.4 or 2.3, respectively). While the ISGs

383 were upregulated, the expressions of interferons were not (0.9 ~ 1.1 fold changes);

384 rather, the expression of IFN-ε decreased in half. These findings are consistent with our

385 hypothesis that induction of those ISGs occurs without functional IFNs.

386 We further validated this finding with a real-time PCR and ELISA assay. First, we

387 sought to understand whether compound 1 simply amplified cellular innate immune

388 responses after virus infection or if the compound was able to establish an antiviral state

389 without an external interferon inducer, such as virus infection. To test this, we measured

390 the induction of innate immune response genes by compound 1 in mock or pVSV-luc

391 infected HEK 293T cells.

392 As shown in Figure 4, the induction of innate immune genes by compound 1 was

393 independent of virus infection. First, we confirmed that the treatment with compound 1

394 induced only certain genes in the interferon pathway. For example, the expressions of

395 MYD88 and OAS1 did not show any changes by compound 1; however, RIG-I, IFIT1,

396 IRF7, and OAS2 genes were upregulated more than 10-fold (solid bars in blue). More

397 interestingly, the induction of the genes by compound 1 was not amplified by virus 398 infection. Treatment of mock- or virus-infected cells (solid vs. spotty blue bars) with

399 compound 1 resulted in a same level of gene expression for the tested genes. We also

400 found that the induction of the ISGs by compound 1 was much stronger than by pVSV-

401 luc infection. pVSV-luc increased the expression of innate immune genes, such as

402 IFIH1, IFIT1, OAS2, OASL and IFNB1 by about 2~4 fold changes, indicating that HEK

403 293T cells are responsive to the infection of the virus. However, the effect was much

404 weaker compared to that of compound 1. Interestingly, compound 1 repressed the

405 expression of IFIH1 and IFNB1; whereas, these genes were induced rather than

406 repressed in the virus-infected groups (orange spotty bars).

407 The lack of induction of type 1 IFNs by compound 1 was confirmed at the cytokine level

408 with ELISA and a cell-based reporter assay. HEK 293T cells were treated with 5 μM of

409 compound 1 for 18 hours then the IFN α/β in the cell culture supernatant was detected

410 in an IFN ELISA assay (Figure 5A) or in a reporter assay (HEK-Blue™ IFN-α/β cells).

411 HEK-Blue™ IFN-α/β cells directly respond to IFN α/β and express the secreted alkaline

412 phosphatase (SEAP) via the activation of IRF9. In both assays, no measurable quantity

413 of IFNs was detected compared to the controls, indicating the lack of IFN α/β induction

414 by the treatment with compound 1. We also treated HEK-Blue™ IFN-α/β cells directly to

415 test whether compound 1 could directly activate the IFNAR and IRF9 pathway or

416 produce IFN as in an autocrine manner (Figure 5B). Treatment with compound 1 did

417 not induce SEAP activity compared to the control as well, indicating no direct activation

418 through IFNAR or any other receptor that can use IRF9. 419 In summary, these data indicate that compound 1 induces expression of certain sets of

420 innate immunity genes, creating an anti-viral state in cells in the absence of type 1 IFN

421

422 Cell line specificity of tetrahydrobenzothiazole 1

423 Since the supporting data strongly suggested that 1 inhibits pyrimidine synthesis and

424 induces the antiviral state of the host cells, we questioned if the antiviral effect and

425 mechanism of 1 is cell type-dependent. To address this question, we measured the

426 antiviral activity of 1 in several cell lines by determining an EC50 value for each. The cell

427 lines include HEK 293T (human embryonic kidney), SY-SH5S (human bone marrow

428 derived neuroblast), Vero76 (African green monkey kidney fibroblast), BHK C21

429 (hamster kidney fibroblast), Neuro2A (mouse neuroblast), and NIH3T3 (mouse

430 fibroblast).

431 The antiviral activities of compound 1 in various cell lines are summarized in Table 3 as

432 a function of EC50 value. As a control for the experiment, we used , which is

433 known to inhibit virus replication by hampering the acidification of endosome during

434 virus entry. The antiviral activities of monensin were very close to each other in all cell

435 lines tested in this experiment. The EC50 values were within a range between 0.02 and

436 0.25 for V3526-luc, and between 0.17 and 0.56 μM for pVSV-luc. This result shows that

437 endocytosis is a critical pathway for the viruses in the cell lines and monensin worked

438 equally in the cell lines. 439 In contrast to monensin, the antiviral activities of compound 1 were cell line and/or

440 species-dependent. The EC50 values of compound 1 in HEK 293T or Vero76 cells were

441 between 0.15 μM and 0.68 μM using either V3526-luc or pVSV-luc, thus implying a

442 strong antiviral activity in these cell lines. Contrary to this, no antiviral activity of

443 compound 1 was detected in the mouse cell lines (Neuro 2A and NIH 3T3) we tested

444 (EC50 > 50 μM for both viruses). Interestingly, compound 1 still showed a moderate

445 effect in BHK cells, a hamster cell line. These data clearly indicate that the antiviral

446 activity of compound 1 is cell line-dependent and much less effective in mouse cells.

447 448 Discussion

449 This report discloses the discovery and mechanistic characterization of

450 tetrahydrobenzothiazole 1, a broad-spectrum antiviral agent that inhibits pyrimidine

451 biosynthesis and induces antiviral responses. This structural series was identified from

452 an anti-VEEV HTS with a mechanism-independent readout, offering the possibility that

453 multiple antiviral agents with different mechanisms of action could be identified from the

454 same screen. In fact, our pursuit of hits from the VEEV HTS led us to develop a

455 structurally distinct inhibitor, ML336, which targets a novel domain of VEEV nsP2 and

456 highlights the domain’s importance for viral replication and as a unique antiviral target.

457 In this study, we revisited our CPE-assay hit compounds with the intent of finding a

458 mechanism of action orthogonal to that of ML336. This effort ultimately led to the

459 development of compound 1. There have been many HTS campaigns that employed

460 the same strategy with different viruses (PubChem assay IDs: 1635, 2310, 2440,

461 540278, 463114, 540249, 588723, 651637, 1074, 51831 and 504781) with each

462 resulting in the discovery of novel hit compounds. As evidenced with our study, those

463 hits may include compounds targeting diverse antiviral mechanisms as well.

464 As Lucas-Hourani, M. et al. indicated, several antiviral HTS campaigns independently

465 discovered pyrimidine synthesis inhibitors targeting DHODH as an antiviral hit (15, 17-

466 19). This fact may indicate that the inhibition of de novo pyrimidine biosynthesis can

467 elicit antiviral activity without hampering cell proliferation. In fact, our experiment

468 addressing the effect of compound 1 on growing cells showed no or minimal impact to

469 cell growth or cytotoxicity at a concentration 10-fold higher than EC50 (Figure S1). We 470 believe that the cellular pyrimidine level that initiates the activation of the ISGs is higher

471 than the concentrations affecting cell proliferation or viability. Unlike the DHODH

472 inhibitors discovered by others, however, compound 1 seems to inhibit an enzyme

473 downstream from DHODH. Our metabolomics analysis clearly showed that treatment

474 with 1 caused the accumulation of orotate as well as DHO (Table 2), which indicates an

475 inhibition at a post-orotate step (Figure S2). The accumulation of orotidin-

476 monophosphate was not monitored; therefore, we hypothesize compound 1 could inhibit

477 orotate phosphoribosyltransferase (OPRTase) activity, which is executed by a dual

478 functional enzyme, synthetase (Umps) (20). Nevertheless, the

479 addition of orotate lessened the antiviral effect of compound 1 (Figure 2B), which could

480 indicate DHODH as the inhibitory step. We believe that the inhibition of OPRTase by

481 compound 1 may not be potent enough; therefore, high concentrations of exogenous

482 orotate (substrate) could facilitate the reaction even in the presence of 1. Alternatively,

483 exogenous orotate could affect the gene expression of ISGs through an unknown

484 pathway. A follow-up study is needed to understand the detailed mechanism of

485 compound 1 in the context of target enzyme.

486 Our assay matrix showed that compound 1 has a broad antiviral activity towards most of

487 the viruses tested but the effect was virus species-dependent (Table 1). While RNA

488 viruses seemed more sensitive than DNA viruses (e.g., HSV-2) to compound 1, not all

489 RNA viruses exhibited equal responsiveness. For example, enteroviruses were resistant

490 to the treatment with compound 1. While the most of RNA viruses we tested are

491 recognized by RIG-I in the infected cells, the infection of HSV-2 and enteroviruses are

492 known to be recognized by melanoma differentiation-associated gene 5 (MDA5, 493 encoded by the IFIH1 gene) and TLR/MYD88, respectively(21, 22). Consistent with this

494 finding, MDA5 and MYD88 were not induced by the treatment with compound 1 (Figure

495 4). A detailed study will be required to elucidate the exact mechanism; however, these

496 results imply that it may be associated with RIG-I pathway.

497 We also found that JEV was less susceptible to compound 1 compared to other

498 flaviviruses tested. Assuming that the viral RNA syntheses were similar to each other

499 (based on titers of the viruses), this finding may indicate a potential difference in the

500 sensitivity to the ISGs that are activated by pyrimidine synthesis inhibition. It is known

501 that the antiviral effect of pyrimidine synthesis inhibitors is through functional ISGs

502 driven by IRF1 transcription factor (15). Therefore, the difference in sensitivity may

503 indicate the resistance to ISGs by each virus. Interestingly, some dengue virus mutants

504 that are resistant to , a DHODH inhibitor, have been reported (23). In that

505 case, the authors proposed a resistance mechanism based on enhancement of

506 polymerase activity or viral assembly; however, our study with compound 1 as a probe

507 suggests that those mutants may exert resistance by evading the ISGs expressed by

508 pyrimidine deprivation. We have attempted to isolate compound 1-resistant mutant

509 VEEV, strain TC-83 by serial passages in the presence of compound 1 for three times;

510 however, we have not detected the emergence of resistant mutants (Figure S3). A

511 future study for isolation of resistant mutants with more passages would be informative

512 to determine resistant mechanisms for alphavirus to this class of compounds, which

513 could include an enhancement of polymerase activity or evasion of ISG. 514 One of the most interesting findings in our study is the induction of ISGs by compound

515 1, including OASL, in the absence of sensitizing the cells with virus, exogenous ssRNA,

516 or interferons (Fig. 4). OASL is an antiviral gene that is known to be expressed under

517 the control of IRF-3 by interferon and ISRE upon virus infection. However, our data

518 showed that OASL could be induced more than 100-fold without exogenous RNA or

519 induction of IRF-3. In our experiment, however, IRF-3 was downregulated after the

520 treatment. Further, the infection of compound 1-pretreated cells with pVSV-luc did not

521 change the cellular innate immune response. This could be explained by the strong

522 antiviral activity of compound 1, which may prevent viral RNA accumulation to a level to

523 trigger the RIG-I/MAVS pathway. Alternatively, the result may imply that the induction of

524 ISGs by compound 1 is independent of viral infection. Nonetheless, our study could

525 explain the mechanism of the amplification of cellular innate immune response by

526 pyrimidine synthesis inhibitors reported by others (15, 24). Our results clearly show the

527 amplification of the response is due to the IFN-independent induction of ISGs (See

528 below for further discussion), which will prime or amplify the antiviral state of the cells

529 upon viral infection. Our findings confirm the previous reports and further highlight the

530 antiviral mechanism of nucleoside synthesis inhibitors in detail.

531 In fact, our study showed that the expression of the ISGs is not associated with the type

532 1 IFNs. In our gene expression assays, no type 1 IFNs genes were induced by

533 compound 1; rather, they were downregulated. Expression of interferon could be

534 transient so the lack of interferon mRNA could not rule out a potential expression at an

535 early time point in the process. To avoid this pitfall, we validated our findings using an

536 ELISA-based assay in addition to a reporter-based assay to detect the interferons 537 produced by compound 1; however, no interferon-α/β activities were detected. Our

538 argument is also supported by the antiviral activity of compound 1 in Vero 76 cells,

539 which allow a variety of virus replication due to the lack of functional type 1 IFNs

540 production systems (16). Considering the antiviral activity of compound 1 in Vero76

541 cells was similar to that in other human cell lines (Table 3), this observation strongly

542 supports the idea that the presence of interferon is not essential for the antiviral activity

543 of compound 1 or other pyrimidine synthesis inhibitors, and the expressed ISGs are not

544 involved in the production of interferons. Our results are differ from the current view on

545 the antiviral mechanism of many ISGs such as OASL, which is believed to induce type I

546 interferons by assembling RIG-I complex in the response to viral infection(25).

547 Therefore, our results clearly highlight new insights on novel antiviral mechanisms of

548 ISGs.

549 The host cell type-dependent antiviral activity was particularly interesting in many

550 aspects. The species-dependent activity could be a universal characteristic of any host-

551 targeting compound, including 1. Study of host-dependent activity would be important to

552 choose an animal model to test those compounds. Compound 1 showed the strongest

553 antiviral effect in human cells but no antiviral activity in mouse cell lines we tested. We

554 also found brequinar does not show antiviral effect in mouse cell lines(26) and Clément

555 Grandin et al. mentioned that GAC50, a potent novel DHODH inhibitor, showed antiviral activity

556 only in primate cells (27). Interestingly, our finding is consistent with the lack of antiviral

557 activity of pyrimidine synthesis inhibitors against RNA viruses in mice as reported by

558 others. Some pyrimidine synthesis inhibitors have been evaluated for their in vivo

559 antiviral activities in mouse-based animal models (except cotton rat model for RSV) (17- 560 19, 28). Those tests have failed to show antiviral efficacy even with a significant

561 decrease in the pyrimidine concentration in the animals’ sera. Due to these results,

562 pyrimidine synthesis inhibitors have not been not well accepted as an antiviral strategy.

563 Our findings, however, conversely suggest that such failure may be due to the use of

564 non-responsive animal models for the inhibitors. , a pyrimidine synthesis

565 inhibitor, has been reported to have an antiviral effect in humans, suggesting pyrimidine

566 synthesis inhibition as a potential broad-spectrum antiviral target for humans (29-31).

567 Therefore, we believe that a worthy goal includes the development of an animal model

568 for this class of inhibitors and to evaluate them for antiviral activities. A recent study

569 using rhesus macaques, however, failed to show anti-RSV effect of a pyrimidine

570 synthesis inhibitor when it was administrated post infection. It has been proposed that

571 the level of circulating pyrimidines in the serum produced through the salvage pathway

572 could be sufficient to reverse the antiviral effect, or the interference with cell-mediated

573 immunity by the inhibitors could be proviral (27). In our study, a pre-treatment was

574 necessary to maximize the antiviral effect in the IFN-deficient cell line. It is not clear,

575 however, whether a pyrimidine synthesis inhibitor would successfully mount an antiviral

576 state and amplify the IFN-driven host innate immune response even at a post-infection

577 stage in IFN-competent cells. More in vivo antiviral studies to test pyrimidine synthesis

578 inhibitors in various animal models would be informative to evaluate their potential of

579 pyrimidine biosynthesis inhibitors as antiviral therapeutics.

580 As the antiviral activity of compound 1 is based on the pyrimidine synthesis inhibition

581 and the subsequent expression of certain ISGs, it is difficult to determine whether the

582 cell type-specificity is related to pyrimidine synthesis inhibition, or to inducing the ISGs. 583 Difference in the sequences of the protein targeted by compound 1, presumably Umps,

584 could account for this phenomenon. The sequence identity between human and mouse

585 Umps is high (88.8%); however, this cannot rule out the potential difference in the

586 binding residues in Umps. A future study is necessary to identify the molecular target.

587 An alternative hypothesis is that the mouse lacks the mechanism to induce the ISGs

588 upon the pyrimidine synthesis inhibition. This is based on the observation that

589 pyrimidine synthesis inhibitors with various chemical structures have failed to

590 demonstrate antiviral activity in mice as mentioned above. Currently, the molecular

591 mechanism of ISGs induction by pyrimidine depression is completely unknown. Lead

592 compound 1 could shed light on the antiviral mechanism involving innate immune

593 responses through the deprivation of pyrimidine.

594 In summary, tetrahydrobenzothiazole 1 is a molecular probe that enabled the study of

595 innate cellular immune responses involved with viral infection. More specifically,

596 compound 1 permits mechanistic scrutiny demonstrating that antiviral ISGs, induced by

597 the inhibition of pyrimidine synthesis, leads to broad-spectrum antiviral activity

598 independent of type 1 IFNs.

599 600 Acknowledgements

601 We thank Dr. Igor Lukashevich for LCMV, Dr. Jill Steinbach for HSV-2, and Dr. Robert

602 Tesh at World Reference Center for Emerging Viruses and Arboviruses for VEEV TrD

603 and WNV. We also thank BEI resources for CHIKV, JEV, YFV 17D, EV D71, and HCMV

604 MM. Microarray experiment was performed with assistance of the UofL Genomics

605 Facility, which is supported by NIH, the J. G. Brown Foundation, and user fees. The

606 funders had no role in study design, data collection and interpretation, or the decision to

607 submit the work for publication.

608 609 References

610

611 1. Stedman C. 2014. Sofosbuvir, a NS5B polymerase inhibitor in the treatment of hepatitis 612 C: a review of its clinical potential. Therapeutic advances in gastroenterology 613 doi:10.1177/1756283X13515825:131-140.

614 2. Zeng QL, Zhang JY, Zhang Z, Wang LF, Wang FS. 2013. Sofosbuvir and ABT-450: 615 terminator of hepatitis C virus? World journal of gastroenterology : WJG 616 doi:10.3748/wjg.v19.i21.3199:3199-3206.

617 3. Sidwell RW, Huffman JH, Khare GP, Allen LB, Witkowski JT, Robins RK. 1972. 618 Broad-spectrum antiviral activity of Virazole: 1-beta-D-ribofuranosyl-1,2,4-triazole-3- 619 carboxamide. Science (New York, NY):705-706.

620 4. Kumar R, Tripathi P, Baranwal M, Singh S, Tripathi S, Banerjee G. 2009. 621 Randomized, Controlled Trial of Oral Ribavirin for Japanese Encephalitis in Children in 622 Uttar Pradesh, India. Clinical Infectious Diseases 48:400-406.

623 5. Mertz GJ, Miedzinski L, Goade D, Pavia AT, Hjelle B, Hansbarger CO, Levy H, 624 Koster FT, Baum K, Lindemulder A, Wang W, Riser L, Fernandez H, Whitley RJ, 625 Collaborative Antiviral Study G. 2004. Placebo-controlled, double-blind trial of 626 intravenous ribavirin for the treatment of hantavirus cardiopulmonary syndrome in North 627 America. Clinical infectious diseases : an official publication of the Infectious Diseases 628 Society of America doi:10.1086/425007:1307-1313.

629 6. Briolant S, Garin D, Scaramozzino N, Jouan A, Crance JM. 2004. In vitro inhibition of 630 Chikungunya and Semliki Forest viruses replication by antiviral compounds: synergistic 631 effect of interferon-alpha and ribavirin combination. Antiviral research:111-117.

632 7. Mendenhall M, Russell A, Juelich T, Messina EL, Smee DF, Freiberg AN, Holbrook 633 MR, Furuta Y, de la Torre JC, Nunberg JH, Gowen BB. 2010. T-705 () 634 Inhibition of Arenavirus Replication in Cell Culture. Antimicrob Agents Chemother 635 doi:AAC.01219-10 [pii]

636 10.1128/AAC.01219-10.

637 8. Chung DH, Jonsson CB, Tower NA, Chu YK, Sahin E, Golden JE, Noah JW, 638 Schroeder CE, Sotsky JB, Sosa MI, Cramer DE, McKellip SN, Rasmussen L, White 639 EL, Schmaljohn CS, Julander JG, Smith JM, Filone CM, Connor JH, Sakurai Y, 640 Davey RA. 2014. Discovery of a novel compound with anti-venezuelan equine 641 encephalitis virus activity that targets the nonstructural protein 2. PLOS Pathogens 642 doi:10.1371/journal.ppat.1004213:e1004213.

643 9. Schroeder CE, Yao T, Sotsky J, Smith RA, Roy S, Chu YK, Guo H, Tower NA, Noah 644 JW, McKellip S, Sosa M, Rasmussen L, Smith LH, White EL, Aube J, Jonsson CB, 645 Chung D, Golden JE. 2014. Development of (E)-2-((1,4-Dimethylpiperazin-2- 646 ylidene)amino)-5-nitro-N-phenylbenzamide, ML336: Novel 2-Amidinophenylbenzamides 647 as Potent Inhibitors of Venezuelan Equine Encephalitis Virus. Journal of Medicinal 648 Chemistry doi:10.1021/jm501203v:8608-8621.

649 10. Bernard KA, Klimstra WB, Johnston RE. 2000. Mutations in the E2 glycoprotein of 650 Venezuelan equine encephalitis virus confer heparan interaction, low morbidity, 651 and rapid clearance from blood of mice. Virology 276:93-103.

652 11. Durocher Y, Perret S, Kamen A. 2002. High-level and high-throughput recombinant 653 protein production by transient transfection of suspension-growing human 293-EBNA1 654 cells. Nucleic acids research:E9.

655 12. Towner JS, Paragas J, Dover JE, Gupta M, Goldsmith CS, Huggins JW, Nichol ST. 656 2005. Generation of eGFP expressing recombinant Zaire ebolavirus for analysis of early 657 pathogenesis events and high-throughput antiviral drug screening. Virology 658 doi:10.1016/j.virol.2004.10.048:20-27.

659 13. Leung DW, Borek D, Luthra P, Binning JM, Anantpadma M, Liu G, Harvey IB, Su Z, 660 Endlich-Frazier A, Pan J, Shabman RS, Chiu W, Davey RA, Otwinowski Z, Basler 661 CF, Amarasinghe GK. 2015. An Intrinsically Disordered Peptide from Ebola Virus VP35 662 Controls Viral RNA Synthesis by Modulating Nucleoprotein-RNA Interactions. Cell 663 reports doi:10.1016/j.celrep.2015.03.034:376-389.

664 14. Wei X, Shi X, Koo I, Kim S, Schmidt RH, Arteel GE, Watson WH, McClain C, Zhang 665 X. 2013. MetPP: a computational platform for comprehensive two-dimensional gas 666 chromatography time-of-flight mass spectrometry-based metabolomics. Bioinformatics 667 (Oxford, England) doi:10.1093/bioinformatics/btt275:1786-1792.

668 15. Lucas-Hourani M, Dauzonne D, Jorda P, Cousin G, Lupan A, Helynck O, Caignard 669 G, Janvier G, Andre-Leroux G, Khiar S, Escriou N, Despres P, Jacob Y, Munier- 670 Lehmann H, Tangy F, Vidalain PO. 2013. Inhibition of pyrimidine biosynthesis pathway 671 suppresses viral growth through innate immunity. PLoS pathogens 672 doi:10.1371/journal.ppat.1003678:e1003678.

673 16. Diaz MO, Ziemin S, Le Beau MM, Pitha P, Smith SD, Chilcote RR, Rowley JD. 1988. 674 Homozygous deletion of the alpha- and beta 1-interferon genes in human leukemia and 675 derived cell lines. Proceedings of the National Academy of Sciences of the United States 676 of America:5259-5263.

677 17. Hoffmann HH, Kunz A, Simon VA, Palese P, Shaw ML. 2011. Broad-spectrum 678 antiviral that interferes with de novo pyrimidine biosynthesis. Proceedings of the National 679 Academy of Sciences of the United States of America 680 doi:10.1073/pnas.1101143108:5777-5782.

681 18. Wang QY, Bushell S, Qing M, Xu HY, Bonavia A, Nunes S, Zhou J, Poh MK, Florez 682 de Sessions P, Niyomrattanakit P, Dong H, Hoffmaster K, Goh A, Nilar S, Schul W, 683 Jones S, Kramer L, Compton T, Shi PY. 2011. Inhibition of dengue virus through 684 suppression of host pyrimidine biosynthesis. Journal of virology doi:10.1128/JVI.02510- 685 10:6548-6556. 686 19. Smee DF, Hurst BL, Day CW. 2012. D282, a non-nucleoside inhibitor of influenza virus 687 infection that interferes with de novo pyrimidine biosynthesis. Antiviral chemistry & 688 chemotherapy doi:10.3851/IMP2105:263-272.

689 20. Yablonski MJ, Pasek DA, Han BD, Jones ME, Traut TW. 1996. Intrinsic activity and 690 stability of bifunctional human UMP synthase and its two separate catalytic domains, 691 orotate phosphoribosyltransferase and -5'- decarboxylase. The 692 Journal of biological chemistry:10704-10708.

693 21. Kuo RL, Kao LT, Lin SJ, Wang RY, Shih SR. 2013. MDA5 plays a crucial role in 694 enterovirus 71 RNA-mediated IRF3 activation. PloS one 695 doi:10.1371/journal.pone.0063431:e63431.

696 22. Rasmussen SB, Jensen SB, Nielsen C, Quartin E, Kato H, Chen ZJ, Silverman RH, 697 Akira S, Paludan SR. 2009. Herpes simplex virus infection is sensed by both Toll-like 698 receptors and retinoic acid-inducible gene- like receptors, which synergize to induce type 699 I interferon production. The Journal of general virology doi:10.1099/vir.0.005389-0:74- 700 78.

701 23. Qing M, Zou G, Wang QY, Xu HY, Dong H, Yuan Z, Shi PY. 2010. Characterization of 702 dengue virus resistance to brequinar in cell culture. Antimicrobial agents and 703 chemotherapy doi:10.1128/AAC.00561-10:3686-3695.

704 24. Yeo KL, Chen YL, Xu HY, Dong H, Wang QY, Yokokawa F, Shi PY. 2015. Synergistic 705 suppression of dengue virus replication using a combination of nucleoside analogs and 706 nucleoside synthesis inhibitors. Antimicrobial agents and chemotherapy 707 doi:10.1128/AAC.04779-14:2086-2093.

708 25. Zhu JZ, Zhang YG, Ghosh A, Cuevas RA, Forero A, Dhar J, Ibsen MS, Schmid- 709 Burgk JL, Schmidt T, Ganapathiraju MK, Fujita T, Hartmann R, Barik S, Hornung V, 710 Coyne CB, Sarkar SN. 2014. Antiviral Activity of Human OASL Protein Is Mediated by 711 Enhancing Signaling of the RIG-I RNA Sensor. Immunity 40:936-948.

712 26. Chung D. 2016. The Establishment of an Antiviral State by Pyrimidine Synthesis 713 Inhibitor is Cell Type-Specific. Journal of Antimicrobial Agents 1:1-4.

714 27. Grandin C, Hourani ML, Janin YL, Dauzonne D, Munier-Lehmann H, Paturet A, 715 Taborik F, Vabret A, Contamin H, Tangy F, Vidalain PO. 2016. Respiratory syncytial 716 virus infection in macaques is not suppressed by intranasal sprays of pyrimidine 717 biosynthesis inhibitors. Antiviral research doi:10.1016/j.antiviral.2015.11.006:58-62.

718 28. Bonavia A, Franti M, Pusateri Keaney E, Kuhen K, Seepersaud M, Radetich B, 719 Shao J, Honda A, Dewhurst J, Balabanis K, Monroe J, Wolff K, Osborne C, Lanieri 720 L, Hoffmaster K, Amin J, Markovits J, Broome M, Skuba E, Cornella-Taracido I, 721 Joberty G, Bouwmeester T, Hamann L, Tallarico JA, Tommasi R, Compton T, 722 Bushell SM. 2011. Identification of broad-spectrum antiviral compounds and 723 assessment of the druggability of their target for efficacy against respiratory syncytial 724 virus (RSV). Proceedings of the National Academy of Sciences of the United States of 725 America doi:10.1073/pnas.1017142108:6739-6744. 726 29. Josephson MA, Javaid B, Kadambi PV, Meehan SM, Williams JW. 2006. 727 Leflunomide in solid organ transplantation and polyoma virus infection. Advances in 728 experimental medicine and biology doi:10.1007/0-387-32957-9_18:255-265.

729 30. Dunn MC, Knight DA, Waldman WJ. 2011. Inhibition of respiratory syncytial virus in 730 vitro and in vivo by the immunosuppressive agent leflunomide. Antiviral therapy 731 doi:10.3851/IMP1763:309-317.

732 31. Jung YH, Moon KC, Ha JW, Kim SJ, Ha IS, Cheong HI, Kang HG. 2013. Leflunomide 733 therapy for BK virus allograft nephropathy after pediatric kidney transplantation. Pediatric 734 transplantation doi:10.1111/petr.12029:E50-54.

735

736 737 Figure legend

738 Figure 1. Antiviral activity of compound 1 against VEEV.

739

740 The structures of CID 847035 (A) and compound 1 (B). (C) Viral RNA analysis. Viral

741 RNA was quantified using a quantitative real-time RT-PCR method with the total RNA

742 from the cells. RNA amounts were compared to the DMSO-treated controls. (D) Titer

743 reduction assay results for compound 1. Vero 76 cells grown in 12-well plates were

744 infected with 0.05 MOI of TC-83 and then incubated in the presence of compound 1 at

745 the denoted concentrations. Forty hours later the supernatant was harvested and the

746 titer of the progeny virus was determined. Each point represents the mean from three

747 biological replicates. (E) Time of addition study. Test compound, compound 1, was

748 added to the designated wells by replenishing the culture media with fresh culture

749 media containing 5 µM of the compound at the time points denoted on the x axis. The

750 cells were infected with 3 MOI of virus and the virus titers at 16 hours post-infection

751 from various time of addition points were depicted. Each data point is the mean from

752 two independent replicates with triplication in titration.

753 754 Figure 2. Reversion of antiviral effect of compound 1 by exogenous nucleosides.

755

756 One day-old HEK 293T cells were treated with DMSO, MPA (1 μM) or ML416 (1 μM) in

757 the presence of the denoted supplements for two hours, then cells were infected with

758 V3526-luc (A) or pVSV-luc (B). After eighteen hours of incubation, the luciferase activity

759 from the infected cells was measured. The values represent the means and their

760 standard deviations of 4 replicates samples as a percentage of the values for DMSO

761 control wells. 762 Figure 3. Induction of innate immune genes by CID 70698683.

763

764 * Immunity Volume 40, Issue 6, 19 June 2014, pages 936–948

765 Values in red : fold-change by the treatment of 5 μM of CID 70698683

766 767 Figure 4. Compound 1 induces innate immune response genes independently of virus

768 infection.

769

770 One-day-old HEK 293T cells were treated with 5 µM of compound 1 or DMSO (Control)

771 for 8 hours, then infected with mock or pseudotypeVSV-luc (pVSV-luc) at a MOI of 3.

772 Cells were further incubated for 16 hours in the presence of compound 1 or DMSO then

773 the host gene expression level was measured using quantitative real-time RT-PCR.

774 775 Figure 5. The treatment of compound 1 does not induce the production of type 1 IFNs.

776

777 (A) HEK 293T cells were treated with compound 1 or DMSO for 18 hours and the cell

778 culture media were subjected to IFN α/β ELISA assay to detect interferons. 125 pg/mL

779 of IFN-α and - β were used as controls. (B) HEK-Blue™ IFN-α/β reporter cells

780 incubated with 5 μM of compound 1 or DMSO for 18 hours and then the expressed

781 SEAP, which is controled by IRF9 was measured.

782

783 A C D E 1010 10 1010 109 109 N O 1 108 108 S N S 0.1 H 107

6 7 B 0.01 10 10 5 F 10 6

Relative vRNA copy 0.001 10 S O 104 Progeny virus titer (PFU/mL)

Progeny virus titer (PFU/mL) 3 N N S 0.0001 10 105 0 1.25 5 20 0 5 10 20 -8 -6 -4 -2 0 Ctrl CH3 Compound 1 (µM) Time of addition (hours to infection) A B V3526-luc pVSV-luc DMSO 150 150 A (50 µM) C (50 µM) C (50 µM) U (50 µM) 100 100 DHO (2 mM) Orotate (2 mM)

50 50 Relative luminescence (%) 0 0 DMSO MPA Compound A DMSO MPA Compound A Treatment Treatment CARD CARD CTD CID 70698683 Helicase N RIG-I 4.63 N S S O viral RNA CTD CTD

Helicase NEMO K63 Ub 1.16 1.43 NAP1 IRF3 or IRF7 * TRIM25 TBK1 IKKε homodimers CARD 0.95 Type I IFN transcription OASL Ub Helicase 0.8 Ub Ub Ub Ub 3.76 CARD Ub UbUb Ub Ub Ub IFN-α,β IFNs : 0.9 - 1.1 OASL Ub Ub OASL UbUb IFN-ε : 0.5 Ub

OASL CARD TRADDosome ISGs mean: 1.23 CARD ISGs CARD

* CARD TRADD NEMO GBP2 : 3.5 CARD CARD ISG20 :2.85

CARD 1.26 α β CARD IKK IKK MAVS IFI44 : 2.83 CARD IRF9 : 2.4 1.13 NF-kB Proin amatory IFIT1 : 2.3 cytokines IFI27 : 2.25 IL18 : 0.42 Control Control + pVSV-luc Compound 1 Compound 1 + pVSV-luc

100

10

1 (fold-increase) Gene expression

0.1 MYD88 DDX58 TRADDSTAT1 IFIH1IFIT1 IRF1 IRF3 IRF7 OAS1 OAS2 OASL IFNB1 A B

20000 1.5 IFN-α IFN-β 15000 1.0

10000 620 nm (IFN α / β ) OD 0.5 5000 Luminescence unit

0 0.0 IFN Background DMSO Compound 1 3.5 0.4 DMSO Compound 1 controls IFN-β (U/mL) Table 1. Antiviral activity of compound 1.

Log titer reduction at Virus family Virus EC50 (μM) 5 μM **

VEEV TC-83 0.35 -2.54

VEEV TrD 0.46 -4.77 Togaviridae VEEV V3526-luc 0.17* NT

CHIKV S17 0.29 -4.024

WEEV 0.96 NT

Rhabdovidae pVSV-luc 0.19* NT

Filroviridae Ebola virus-GFP 0.26 NT

Picornaviridae EV-71, MP4 11.1 NT

EMCV MM >25 NT

Influenza virus A Orthomyxoviridae NT -3.03 (H1N1)

Arenaviridae LCMV-ARM NT -3.466

Paramyxoviridae RSV Long NT -2.27

94% inhibition at Retroviridae HIV-1 NT 10 μM JEV NT -1.3

Flaviviridae YFV 17D NT -2.32

WNV NT -2.05

Herpesviridae HSV-2 NT 0.01

For titration reduction assays, the progeny virus titers were compared to the controls

(DMSO treated). For dose-response assays, cells were pretreated for two hours prior to infection and the luciferase activity was measured 16 hours later.

* luciferase-tagged virus assay.

** Negative values mean a decrease in progeny virus titer compared to the mock treated controls. The data represent the means of at least three replicates.

Table 2. Difference in cellular metabolites by the treatment of compound 1.

Groups Name p-value * The dl-Dihydroorotic acid 4.11 x10-4

-3 Metabolites detected Orotic acid 2.26 x10

only in compound 1- l- 1.52 x10-2 treated cells (not -5 detected in mock- 4.11 x10 treated cells) Alloxanoic acid* 4.11 x10-5

2-Ketoisovaleric acid* 4.11 x10-5

Uridine 4.11 x10-5

-4 D-(-)- Erythrofuranose,(isomer)* 4.16 x10

(3R,4R)--2,3,4- 4.11 x10-5 triol*

Metabolites detected Dihydromuconic acid; trans-3- 1.52 x10-2 Hexenedioic acid* only in mock-treated Uridine phosphate* 4.11 x10-4 cells (not detected in compound 1-treated Methanesulfinic acid* 4.11 x10-4 cells) N-Acetyl-L-glutamic acid* 4.11 x10-5

Putrescine; 1,4-Diaminobutane* 4.11 x10-5

Pidolic acid* 3.30 x10-3

2-Hydroxyglutaric acid 2.32 x10-3 compounds that have not been confirmed by using standard compounds.

Table 3. Antiviral activity of compound 1 in various cell lines.

μ EC50 ( M) of

Monensin compound 1

V3526-luc pVSV-Gp V3526-luc pVSV-Gp

0.08 0.19 HEK 293T 0.15 0.17 0.03 0.74

0.17 SY-SH5S 0.02 0.49 3.82 0.48

0.56 0.17 0.20 Vero76 3.87 2.57 0.25 0.68

0.09 0.50 1.12 8.26 BHK 0.24 0.19 0.92 7.16

0.22 0.25 Neuro2A 0.23 >50 >50 0.19 0.29

0.24 NIH 3T3 0.02 >50 >50 0.67

Each number represents EC50 evaluated from a dose-response study with concentrations starting from 25 µM by a five-fold dilution, triplicates for each, in a 96- well format. For assays with HEK 293T, Neuro 2A, and SH-SY5Y cells, 24,000 cells and 2,400 TCID50 units of virus per well was used. For Vero 76 and BHK cells, 12,000 cells and 1200 TCID50 units of virus per well was used. For NIH3T3, 24,000 cells and 20,000 TCID50 units of virus per well was used. EC50 values were calculated using XLfit

(IDBS) formula 205, a 4-parameter Levenburg-Marquardt algorithm with maximum and minimum limits set at 100 and 0, respectively.