AAC Accepted Manuscript Posted Online 26 January 2015 Antimicrob. Agents Chemother. doi:10.1128/AAC.04779-14 Copyright © 2015, American Society for Microbiology. All Rights Reserved.

1 2

Synergistic suppression of dengue virus replication using a combination of nucleoside

3

analogs and nucleoside synthesis inhibitors

4 5

Running Title: Synergy between nucleoside and its synthesis inhibitor

6 7

Kim Long Yeo, Yen-Liang Chen, Hao Ying Xu, Hongping Dong, Qing-Yin Wang,

8

Fumiaki Yokokawa, Pei-Yong Shi*

9 10

Novartis Institute for Tropical Diseases

11

10 Biopolis Road, #05-01 Chromos Building

12

Singapore 138670

13

Singapore

14 15

*Corresponding author:

16

Pei-Yong Shi:

17

Email: [email protected]

18

1

19 20

ABSTRACT Dengue virus (DENV) is the most prevalent mosquito-borne viral pathogen in humans.

21

Currently, there is no clinically approved vaccine or antiviral for DENV. Combination therapy is

22

a common practice in antiviral treatment and a potential approach to search for new treatment for

23

infectious pathogens. In this study, we performed a combination treatment in cell culture using

24

three distinct classes of inhibitors, including ribavirin (a guanosine analog with several antiviral

25

mechanisms), brequinar (a pyrimidine biosynthesis inhibitor), and INX-08189 (a guanosine

26

analog). The compound pairs were evaluated for their antiviral activities using a DENV-2

27

luciferase replicon assay. Our result indicated combination of Ribavirin + INX-08189 exhibited

28

strong antiviral synergy. This result suggests that synergy can be achieved with compound pairs

29

in which one compound suppresses the synthesis of the nucleoside for which the other compound

30

is a corresponding nucleoside analog. In addition, we found that treatment of cells with brequinar

31

alone could activate interferon-stimulated response element (ISRE); furthermore, brequinar and

32

NITD-982 (another pyrimidine biosynthesis inhibitor) can potentiate the interferon-induced

33

ISRE activation. Compared with brequinar, treatment of cells with ribavirin alone could also

34

induce ISRE activation, but to a less extend; however, when cells were co-treated with ribavirin

35

and interferon-β, ribavirin did not augment the interferon-induced ISRE activation.

36 37

2

38 39

INTRODUCTION Over 2.5 billion people worldwide are at risk from dengue virus (DENV) infection, with

40

390 million human infections and 96 million cases with disease manifestations each year (1).

41

DENV is endemic throughout tropical and subtropical climates, and found mostly in urban and

42

semi-urban areas. This positive sense single-stranded RNA virus is mainly transmitted by the

43

Aedes Aegypti mosquito and classified under the genus Flavivirus in the family Flaviviridae.

44

Other notable viruses in this group include yellow fever virus, Japanese encephalitis virus, West

45

Nile virus, and tick-borne encephalitis virus. Currently, neither antiviral nor vaccine is approved

46

for DENV. Care for hospitalized dengue patients is supportive, mainly through optimal

47

replenishment of body fluids. Treatment is intensive for those who succumb to the severe forms

48

of the disease, dengue shock syndrome (DSS) or dengue hemorrhagic fever (DHF).

49

Multiples approaches have been taken to identify inhibitors of DENV (2, 3). Small

50

molecule inhibitors have been reported to target various DENV proteins, including capsid (4, 5),

51

envelope (6), protease (7, 8), non-structural protein (NS) 4B (9, 10), methyltransferase (11, 12),

52

and RNA-dependent RNA polymerase (13-17). Inhibitions of host factors important for viral

53

replication and compounds with immune modulation activities have also been pursued for

54

potential treatment of DENV infections, including iminosugars (18), cholesterol inhibitors (19),

55

chloroquine (20), and prednisolone (21).

56

Ribavirin is a drug with broad-spectrum of antiviral activity. Ribavirin in combination

57

with pegylated interferon (IFN)-α in the past has been the standard treatment regime for hepatitis

58

C virus (HCV), a virus related to DENV from family Flaviviridae. Besides HCV, ribavirin had

59

also shown some success in the treatment of respiratory syncytial virus (22) and Lassa fever

60

virus (23). Ribavirin is a guanosine analog with several antiviral mechanisms, one of which is to

3

61

inhibit de novo biosynthesis of guanine nucleotides through direct binding to cellular inosine

62

monophosphate dehydrogenase (IMPDH) (24). Depletion of intracellular pool of nucleoside

63

triphosphates was proposed to be a major antiviral mechanism for ribavirin to inhibit flavivirus

64

(25). In addition, ribavirin could function as a mutagen to increase error catastrophe (26) and

65

potentiated the antiviral activity of interferon (IFN)-α/β by augmenting the expression of ISGs

66

(IFN-stimulated genes) (27). Similar to ribavirin, brequinar also has a broad antiviral spectrum

67

against both positive and negative-stranded RNA viruses (28, 29). Brequinar inhibits de novo

68

biosynthesis of uracil nucleotides by inhibiting cellular dihydroxyorotate dehydrogenase

69

(DHODH) (30). Depletion of intracellular pyrimidine triphosphates is the main antiviral

70

mechanism for brequinar (28). Brequinar was first identified and developed as an anti-metabolite

71

in cancer and immunosuppression therapy; since tumor cells rely heavily on de novo nucleotide

72

synthesis, lowering the pyrimidine synthesis (by brequinar) could interfere with the rapid

73

proliferation of lymphocytes (31).

74

Combination therapy is commonly practiced in anti-infective treatment to minimize drug

75

resistance. Although there is no clinically approved antivirals for DENV, it is of interest to

76

examine whether compounds that are in clinical use or in preclinical development for other

77

viruses inhibit DENV; and, if so, whether these compounds could have synergistic effect on anti-

78

DENV activity when used in combination. In this study, we selected three clinical and preclinical

79

compounds (Brequinar , Ribavirin, and INX-08189) with known anti-DENV activities and

80

examined their combinatory antiviral activities in a cell culture system. The results showed that

81

combination of the guanosine analog INX-08189 with GTP pool-depleting ribavirin inhibited

82

DENV in a synergistic manner. The observed synergy could potentially be used to reduce the

83

doses and, therefore, increase the safety margins of inhibitors to achieve therapeutic window in

4

84

vivo. In addition, we found that brequinar and another uridine biosynthesis inhibitor can

85

potentiate interferon-induced interferon-stimulated response element (ISRE) activation.

86 87

MATERIALS AND METHODS

88

Cells and culture media

89

Huh-7 cells stably expressing the luciferase-reporting DENV-2 [New Guinea C (NGC)

90

strain] replicon was described previously (32). These cells were maintained at 37oC and 5% CO2

91

in Dulbecco’s modified Eagle medium (DMEM; high glucose; Life Technologies) supplemented

92

with 2 mM L-glutamine (Life Technologies), 0.1 mM non-essential amino acid (Life

93

Technologies), 10 µg/mL puromycin (Clontech), 1% penicillin/streptomycin (Life

94

Technologies), and 10% fetal bovine serum (FBS; Hyclone). The replicon cells seeded for

95

antiviral assay were maintained in phenol red-free DMEM (Life Technologies) supplemented

96

with 1 mM sodium pyruvate (Life Technologies), 2 mM L-glutamine, 0.1 mM Non-essential

97

Amino Acids (NEAA) (Life Technologies), 1% penicillin/streptomycin, and 2% FBS. HEK 293-

98

T cells were maintained at 37oC and 5% CO2 in DMEM (low glucose) supplemented with 0.1

99

mM NEAA, 1% penicillin/streptomycin, and 10% FBS. HEK 293-T cells seeded for assays were

100

maintained with 2% FBS instead.

101

Compounds

102

Ribavirin (CAS number 36791-04-5), brequinar sodium salt hydrate, and recombinant

103

human interferon-β (IFN-β) were purchased from Sigma-Aldrich. INX-08189 and NITD-982

104

(29) were synthesized in-house. All compounds were dissolved in Dimethyl sulfoxide (DMSO).

105

DENV replicon antiviral assay

5

106

The replicon assay was performed as described previously (32). Briefly, replicon cells

107

were treated with 2 or 3-fold serial dilutions of test compounds. After incubation at 37oC for 48

108

h, luciferase substrate (ViviRen, Promega) was added according to manufacturer’s protocol.

109

Luminescence was measured using the Clarity luminescence reader (BioTek) with an integration

110

time of 0.1 second. The concentrations of compounds that decreased the luciferase expression by

111

50% (EC50) and 90% (EC90) were calculated by nonlinear regression analysis (GraphPad Prism).

112

Compound synergy analysis was performed using Chalice Analyzer (Zalicus).

113

Cell viability assay

114

Cell viability was measured using the CellTiter-Glo luminescent cell viability assay

115

(CTG, Promega) according to manufacturer’s protocol. Approximately 1.5×104 replicon cells or

116

2×104 HEK 293-T cells were seeded in a 96-well plate in a total volume of 100 µl. After 16 h of

117

incubation, the cells were treated with test compound. After another 48 h, 25 µL of CTG was

118

added into to each well and the cells incubated at room temperature for 20 min. Luminescence

119

was read with an integration time of 0.1 second using the Clarity luminescence reader (BioTek).

120

Plasmids and transfection of HEK 293-T cells for IFN-stimulated response element (ISRE)

121

induction assay

122

Construct containing a firefly luciferase reporter under the control of an ISRE gene

123

promoter (pISRE-TA-Luc) and construct containing a renilla luciferase reporter under the

124

control of the herpes simplex virus type 2 thymidine kinase gene (HSV-TK) promoter (pGL4.74-

125

hRluc/TK) were purchased from Clontech and Promega, respectively. Batch transfection of HEK

126

293-T cell was performed with jetPRIME (Polyplus). For one 96-well culture plate, 12 µg each

127

of pISRE-TA-Luc and pGL4.74-hRluc/TK were diluted in 600 µl of jetPRIME transfection

128

buffer. Forty-eight µl of jetPRIME was then added, mixed, and incubated for 10 min at room

6

129

temperature. This mixture was added to 2.4 ×106 cells in a final volume of 12 ml DMEM

130

containing 0.1 mM NEAA, 1% penicillin/streptomycin, and 2% FBS. Finally 100 µl of this cell

131

suspension was added to each well of the microplate containing 1 µl of compound. Cells were

132

incubated for 48 h at 37oC in the presence of 5% CO2. Luciferase expression was assayed using

133

the Dual-Glo Stop & Glo assay system (Promega) according to manufacturer’s

134

recommendations. Briefly, media was removed from the wells containing cells and the cells were

135

washed twice with Phosphate buffered saline (PBS) (Life Technologies). Cells were then lysed

136

for 20 min at room temperature on an orbital shaker. Subsequently, 20 µl aliquot of cell lysate

137

from each well was pipetted into wells of a white wall, white bottom plate. Firefly luciferase

138

expression was measured by injecting 100 µl firefly luciferase substrate into each well.

139

Expression was measured 2 seconds later on a Clarity luminescence reader (BioTek) using a 10-

140

second integration time. After the first 20 seconds reading, 100 µl of Stop & Glo reagent was

141

injected into each well. Renilla luciferase expression was measured 2 seconds later with an

142

integration time of 10 seconds.

143

Statistical analysis

144

All the EC50s were calculated using 4 point non-linear regression curve fitting with variable

145

slope and no constrains on both the top and bottom values in the PRISM software. Chalice

146

Analyzer (33) was used to evaluate synergy. The theories of Loewe additivity, Bliss independent

147

and highest single agent (HSA) were described in details in the references (34-36).

148

RESULTS

149

Antiviral activity of individual compounds

150 151

We selected three compounds (ribavirin, brequinar, and INX-08189) to test their combination antiviral activities (left panels, Fig. 1A-C). Each of the selected compounds has

7

152

distinct mode-of-actions (see Introduction). A DENV-2 luciferase replicon assay was used to

153

measure antiviral activity throughout the study. Prior to combination testing, we first examined

154

the anti-DENV activity of individual compounds (middle panels) and cytotoxicity (right panels).

155

Figure 1D summarizes the EC50, EC90, and CC50 values of each compound. The results

156

demonstrate that all selected compounds have anti-DENV activities with good therapeutic

157

window in cell culture.

158 159 160

Antiviral activity of combination treatment We measured antiviral activities of different compound pairs such that the inhibitor of

161

nucleoside pathway is paired with a nucleoside inhibitor, INX-08189. To examine whether

162

combinatory treatments have synergistic or additive effects, we tested each compound at seven

163

concentrations (excluding a control without compound) with dose matrix centered on the EC50

164

value. The left panels in Figure 2 show the percentages of replicon inhibition under each dose

165

matrix (left panel, Fig. 2A). Chalice Analyzer was used to calculate the Loewe excess (right

166

panel, Fig. 2A), Bliss excess (left panels, Fig. 2B), and HSA excess values (right panels, Fig.

167

2B). These parameters are commonly used to indicate the excess percentage inhibition; the

168

excess percentage inhibition is calculated by deducting the expected percentage inhibition values

169

of various combinations, assuming non-synergy pairing in various models, from the

170

experimental percentage inhibition values. These data allowed us to calculate the isobolograms,

171

synergy scores, and best combination indices (CI) for each pair (Fig. 2C). In general, synergy

172

score of >1 and CI of 5 µM and > 800 µM, respectively (Fig. 3D). The results indicate that, in the absence of

205

exogenous IFN, treatment of cells with brequinar or ribavirin alone could induce ISRE

206

activation.

207 208 209

DHODH inhibitors potentiates the IFN-induced ISRE activation Since brequinar or ribavirin alone could induce ISRE activation, we examined whether

210

these compounds could potentiate/enhance the ISRE activation when cells are co-incubated with

211

exogenous IFN. To this end, HEK 293T cells were co-incubated with IFN-β and ribavirin or

212

brequinar. As shown in Figure 4, compared with DMSO control, brequinar significantly

213

potentiated ISRE activation when the cells were co-treated with 5,000 and 10,000 IU/ml IFN-β

214

(p-value < 0.001); no potentiation was observed when the cells were co-treated with lower doses

215

of IFN-β. In contrast, treatment with ribavirin plus IFN-β did not augment ISRE activation (Fig.

216

4). The results indicate that brequinar, a DHODH inhibitor, could enhance the exogenous IFN-

217

induced ISRE activation.

218

Next, we asked whether other pyrimidine biosynthesis inhibitors could also potentiate the

219

IFN-induced ISRE activation. To address this question, we used NITD-982, a compound that

220

was recently reported as a potent inhibitor of DHODH (29). Treatment with NITD-982 at a non-

10

221

cytotoxic concentration significantly augmented the ISRE activation when the cells were co-

222

incubated with 10,000 IU/ml IFN-β (p-value < 0.001; Fig. 4). Taken together, the results

223

demonstrate that DHODH inhibitors (brequinar and NITD-982) can potentiate the IFN-induced

224

ISRE activation.

225 226 227

DISCUSSION In this study, we explored the possibility of a combinatorial therapy selected from three

228

compounds that have anti-DENV activities. Since INX-08189 is a potent nucleoside inhibitor for

229

DENV (Fig. 1C), we chose this compound to perform combination analysis and tested the

230

hypothesis that co-treatment with INX-08189 plus another inhibitor that perturbs the synthesis of

231

natural triphosphate nucleotide(s) could lead to antiviral synergy. Our results showed that

232

compound pair ribavirin + INX-08189 exhibited clear antiviral synergy with strong CI of 0.325

233

(Fig. 2C). The combination has the potential to lower the dose of either compound by 3 times to

234

achieve a similar level of viral inhibition (34); this could translate into a bigger therapeutic

235

window for the treatment of DENV with these two compounds. The synergy is not due to the

236

cytotoxicity of the combination treatment (Fig. 2D). It should be noted that synergy was

237

observed in compound pairs in which one compound suppressed the synthesis of the nucleoside

238

for which the other compound is a corresponding nucleoside analog. This is expected because

239

reduction of the natural nucleoside pool (by the nucleoside synthesis inhibitor) increases the ratio

240

of the nucleoside analog triphosphate versus the natural nucleoside triphosphate, making the

241

nucleoside analog triphosphate more likely to be incorporated into the viral RNA and thus

242

termination of viral RNA synthesis. Specifically, the synergy was found between ribavirin (a

243

guanosine synthesis inhibitor) and INX-08189 (a guanosine analog).

11

244

A similar combination antiviral approach was previously employed for HCV and HIV.

245

For HCV, INX-08189 was tested in combination with ribavirin in the presence or absence of

246

pegylated interferon (PEG-IFN) in HCV patients; unfortunately, adverse effects (heart and renal

247

toxicity) were observed (38). This is not too surprising since INX-08189 had similar

248

toxicological finding when administered as monotherapy (39). For HIV, a CTP synthase

249

inhibitor (3’-deazauridine) strongly potentiated the anti-HIV-1 activity of a 5’-triphoaphate of

250

the cytidine analog (3TC) and a 2,3’-dideoxy cytidine (ddC) in cell culture (40). One explanation

251

for drug synergy is the parallel pathway inhibition model, which suggests that two drugs will be

252

synergistic if they inhibit two proteins on parallel pathways essential for an observed phenotype

253

(41). Our synergy results seem to support the parallel pathway inhibition model. However, we

254

could not exclude other possible mechanisms that could also contribute to the observed synergy.

255

It would be interesting to test whether the antiviral synergy of ribavirin + INX-08189

256

observed in cell culture could be reproduced in the dengue AG129 mouse model. Since

257

nucleoside inhibitor requires host kinases to convert to its active triphosphate form, caution

258

should be taken when considering such in vivo study. Due to the potential species difference in

259

converting INX-08189 to the triphosphate INX-08189 between human and mouse, the relevance

260

of the non-human animal model should be carefully validated experimentally when analyzing

261

nucleoside inhibitors.

262

The potential involvement of IFN pathway in the observed antiviral activity was

263

investigated. For brequinar, besides inhibiting pyrimidine synthesis, our results showed that

264

brequinar alone could activate ISRE in cell culture; furthermore, brequinar could enhance the

265

exogenous IFN-induced ISRE activation. Such enhancement was further validated with another

266

pyrimidine biosynthesis inhibitor (NITD-982). The mechanisms of brequinar/NITD-982-

12

267

mediated ISG activation and IFN-signaling enhancement remain to be determined. Nevertheless,

268

these observations may have clinical implications. When patients receive therapy, DENV has

269

already established its replication, triggering immune response to produce IFN and other

270

cytokines (42, 43). If the patients were treated with brequinar, its ability to augment the IFN-

271

induced ISRE activation should enhance the overall antiviral status of patients.

272

The synergy observed in the ribavirin + INX-08189 treatment is unlikely to go through

273

the ribavirin-mediated ISRE activation. This is because brequinar alone activated ISRE (Fig. 3B)

274

but failed to synergize INX-08189. In addition, the activation level of IRES was modest even at

275

high concentration of ribavirin.

276

There is an urgent unmet medical need to develop a safe and effective antiviral for

277

DENV infection. Although no single compound has been approved for clinical use, the

278

possibility of repurposing clinically tried or approved drugs for DENV is a tractable option.

279

Towards this approach, balapiravir, a cytidine nucleoside analog that was stopped for HCV

280

clinical development, was tested in dengue patients; unfortunately, the compound did not show

281

any efficacy in the dengue clinical trial (14). Similarly, celgosivir, a host alpha-glucosidase

282

inhibitor (initially developed for HCV) also failed to show significant difference compared with

283

placebo (44). An alternative approach to repurpose clinically approved drugs for DENV is to

284

search for combination therapy. Such approach has two conceptual advantages. Firstly, synergy

285

between two drugs would allow achieving efficacy at lower doses, leading to increased

286

therapeutic window of potentially toxic compounds (45). Theoretical and experimental studies

287

have shown that drugs that exhibit synergy for a specific effect are usually not synergistic for

288

side effects (33, 46). Indeed, toxicity experiments suggest that the observed antiviral synergy of

289

ribavirin + INX-08189 combination was not due to cytotoxicity (Fig. 2). However, due to the

13

290

unpredictable nature of toxicity associated with nucleoside analog, caution should be taken when

291

extrapolating the in vitro toxicity results. Secondly, combination treatment would minimize the

292

chance of resistance. In the absence of effective monotherapy for DENV, a combination of two

293

moderately effective drugs may be needed. This was evident in the treatment of HIV where only

294

combination of drugs effectively reduces the viremia to undetectable level. These considerations

295

make synergistic drug pairs ideal candidates for treatment of infectious pathogens. The current

296

study used DENV-2 as a model to achieve proof-of-concept. Future studies are needed to expand

297

the current observation in cell culture to an appropriate in vivo study.

298 299 300

ACKNOWLEDGEMENTS We thank Chek Shik Lim, Christophe Leroy, and Joseph Lehar on data analysis on

301

synergy. We thank colleagues at Novartis Institute for Tropical Diseases (NITD) for helpful

302

discussions and help during the course of this study. All authors are employees of Novartis.

303 304

14

305

REFERENCES

306

1.

307

Brownstein JS, Hoen AG, Sankoh O, Myers MF, George DB, Jaenisch T, Wint GRW,

308

Simmons CP, Scott TW, Farrar JJ, Hay SI. 2013. The global distribution and burden of

309

dengue. Nature 496:504–7.

310

2.

311

Dong H, Liu B, Bodenreider C, Lee G, Ding M, Chan WL, Wang G, Jian YL, Chao AT,

312

Lescar J, Yin Z, Vedananda TR, Keller TH, Shi P-Y. 2011. Small molecule inhibitors that

313

selectively block dengue virus methyltransferase. J. Biol. Chem. 286:6233–40.

314

3.

315

Strategies for development of Dengue virus inhibitors. Antiviral Res. 85:450–62.

316

4.

317

C, Wineinger KA, Page JM, Harver C, Stavale E, Tyavanagimatt S, Stone MA,

318

Bartenschlager R, Scaturro P, Hruby DE, Jordan R. 2013. A novel inhibitor of dengue virus

319

replication that targets the capsid protein. Antimicrob. Agents Chemother. 57:15–25.

320

5.

321

Mohana-Borges R, Castanho M a RB, Almeida FCL, Santos NC, Da Poian AT. 2012. The

322

disordered N-terminal region of dengue virus capsid protein contains a lipid-droplet-binding

323

motif. Biochem. J. 444:405–15.

324

6.

325

Heudi O, Ma NL, Poh MK, Phong WY, Keller TH, Jacoby E, Vasudevan SG. 2009. A

326

small-molecule dengue virus entry inhibitor. Antimicrob. Agents Chemother. 53:1823–31.

Bhatt S, Gething PW, Brady OJ, Messina JP, Farlow AW, Moyes CL, Drake JM,

Lim SP, Sonntag LS, Noble C, Nilar SH, Ng RH, Zou G, Monaghan P, Chung KY,

Noble CG, Chen Y-L, Dong H, Gu F, Lim SP, Schul W, Wang Q-Y, Shi P-Y. 2010.

Byrd CM, Dai D, Grosenbach DW, Berhanu A, Jones KF, Cardwell KB, Schneider

Martins IC, Gomes-Neto F, Faustino AF, Carvalho F a, Carneiro F a, Bozza PT,

Wang Q-Y, Patel SJ, Vangrevelinghe E, Xu HY, Rao R, Jaber D, Schul W, Gu F,

15

327

7.

328

hydroxyquinoline derivatives containing aminobenzothiazole as inhibitors of dengue virus type 2

329

protease in vitro. Antiviral Res. 97:74–80.

330

8.

331

design of therapeutics. Antiviral Res. 96:115–26.

332

9.

333

M, Neyts J, van Kuppeveld FJM, van Rij RP. 2013. Identification of a new dengue virus

334

inhibitor that targets the viral NS4B protein and restricts genomic RNA replication. Antiviral

335

Res. 99:165–71.

336

10.

337

dengue virus by targeting viral NS4B protein. J. Virol. 85:11183–95.

338

11.

339

Dong H, Liu B, Bodenreider C, Lee G, Ding M, Chan WL, Wang G, Jian YL, Chao AT,

340

Lescar J, Yin Z, Vedananda TR, Keller TH, Shi P-Y. 2011. Small molecule inhibitors that

341

selectively block dengue virus methyltransferase. J. Biol. Chem. 286:6233–40.

342

12.

343

Canard B, Qvist J. 2007. Virtual screening and bioassay study of novel inhibitors for dengue

344

virus mRNA cap (nucleoside-2’O)-methyltransferase. Bioorg. Med. Chem. 15:7795–802.

345

13.

346

Kondreddi RR, Goh A, Xu HY, Yip A, Liu B, Weaver M, Dartois V, Keller TH, Shi P-Y.

347

2010. Inhibition of dengue virus by an ester prodrug of an adenosine analog. Antimicrob. Agents

348

Chemother. 54:3255–61.

Lai H, Sridhar Prasad G, Padmanabhan R. 2013. Characterization of 8-

Noble CG, Shi P-Y. 2012. Structural biology of dengue virus enzymes: towards rational

van Cleef KWR, Overheul GJ, Thomassen MC, Kaptein SJF, Davidson AD, Jacobs

Xie X, Wang Q-Y, Xu HY, Qing M, Kramer L, Yuan Z, Shi P-Y. 2011. Inhibition of

Lim SP, Sonntag LS, Noble C, Nilar SH, Ng RH, Zou G, Monaghan P, Chung KY,

Luzhkov VB, Selisko B, Nordqvist A, Peyrane F, Decroly E, Alvarez K, Karlen A,

Chen Y-L, Yin Z, Lakshminarayana SB, Qing M, Schul W, Duraiswamy J,

16

349

14.

350

QTH, Tran HT, Nguyen CVV, Merson L, Hoang LT, Hibberd ML, Aw PPK, Wilm A,

351

Nagarajan N, Nguyen DT, Pham MP, Nguyen TT, Javanbakht H, Klumpp K, Hammond J,

352

Petric R, Wolbers M, Nguyen CT, Simmons CP. 2013. A randomized, double-blind placebo

353

controlled trial of balapiravir, a polymerase inhibitor, in adult dengue patients. J. Infect. Dis.

354

207:1442–50.

355

15.

356

Mueller D, Voshol H, Lim JYH, Nilar S, Keller TH, Shi P-Y. 2010. Inhibition of dengue virus

357

polymerase by blocking of the RNA tunnel. J. Virol. 84:5678–86.

358

16.

359

Conformational flexibility of the Dengue virus RNA-dependent RNA polymerase revealed by a

360

complex with an inhibitor. J. Virol. 87:5291–5.

361

17.

362

Niyomrattanakit P, Lakshminarayana SB, Goh A, Xu HY, Liu W, Liu B, Lim JYH, Ng

363

CY, Qing M, Lim CC, Yip A, Wang G, Chan WL, Tan HP, Lin K, Zhang B, Zou G,

364

Bernard K a, Garrett C, Beltz K, Dong M, Weaver M, He H, Pichota A, Dartois V, Keller

365

TH, Shi P-Y. 2009. An adenosine nucleoside inhibitor of dengue virus. Proc. Natl. Acad. Sci. U.

366

S. A. 106:20435–9.

367

18.

368

Lee HS, Vasudevan SG. 2012. Dose- and schedule-dependent protective efficacy of celgosivir

369

in a lethal mouse model for dengue virus infection informs dosing regimen for a proof of concept

370

clinical trial. Antiviral Res. 96:32–5.

Nguyen NM, Tran CNB, Phung LK, Duong KTH, Huynh HLA, Farrar J, Nguyen

Niyomrattanakit P, Chen Y-L, Dong H, Yin Z, Qing M, Glickman JF, Lin K,

Noble CG, Lim SP, Chen Y-L, Liew CW, Yap L, Lescar J, Shi P-Y. 2013.

Yin Z, Chen Y-L, Schul W, Wang Q-Y, Gu F, Duraiswamy J, Kondreddi RR,

Watanabe S, Rathore APS, Sung C, Lu F, Khoo YM, Connolly J, Low J, Ooi EE,

17

371

19.

372

Wolbers M, Merson L, Dung NTP, Peeling R, Simmons C, Wills B, Farrar J. 2012.

373

Lovastatin for adult patients with dengue: protocol for a randomised controlled trial. Trials

374

13:203.

375

20.

376

2010. A randomized controlled trial of chloroquine for the treatment of dengue in Vietnamese

377

adults. PLoS Negl. Trop. Dis. 4:e785.

378

21.

379

Truong NT, Dung NT, Qui PT, Hien TT, Farrar JJ, Simmons CP, Wolbers M, Wills BA.

380

2012. Effects of short-course oral corticosteroid therapy in early dengue infection in Vietnamese

381

patients: a randomized, placebo-controlled trial. Clin. Infect. Dis. 55:1216–24.

382

22.

383

ribavirin and mycophenolic acid against orthopoxviruses in vitro. Antivir. Chem. Chemother.

384

12:327–35.

385

23.

386

C, Gölnitz U, Fleischer K, Günther S. 2003. Imported Lassa fever in Germany: surveillance

387

and management of contact persons. Clin. Infect. Dis. 36:1254–8.

388

24.

389

replication and reduce GTP levels in Aedes albopictus cells. Virology 110:281–9.

390

25.

391

which ribavirin exerts its antiviral activity in vitro against flaviviruses and paramyxoviruses is

392

mediated by inhibition of IMP dehydrogenase. J. Virol. 79:1943–7.

Whitehorn J, Van Vinh Chau N, Truong NT, Tai LTH, Van Hao N, Hien TT,

Tricou V, Minh NN, Van TP, Lee SJ, Farrar J, Wills B, Tran HT, Simmons CP.

Tam DTH, Ngoc T V, Tien NTH, Kieu NTT, Thuy TTT, Thanh LTC, Tam CT,

Smee DF, Bray M, Huggins JW. 2001. Antiviral activity and mode of action studies of

Haas WH, Breuer T, Pfaff G, Schmitz H, Köhler P, Asper M, Emmerich P, Drosten

Malinoski F, Stollar V. 1981. Inhibitors of IMP dehydrogenase prevent sindbis virus

Leyssen P, Balzarini J, De Clercq E, Neyts J. 2005. The predominant mechanism by

18

393

26.

394

test by using ribavirin. Proc. Natl. Acad. Sci. U. S. A. 98:6895–900.

395

27.

396

interferon action by augmenting interferon-stimulated gene induction in hepatitis C virus cell

397

culture models. Hepatology 53:32–41.

398

28.

399

Characterization of dengue virus resistance to brequinar in cell culture. Antimicrob. Agents

400

Chemother. 54:3686–95.

401

29.

402

Florez de Sessions P, Niyomrattanakit P, Dong H, Hoffmaster K, Goh A, Nilar S, Schul W,

403

Jones S, Kramer L, Compton T, Shi P-Y. 2011. Inhibition of dengue virus through

404

suppression of host pyrimidine biosynthesis. J. Virol. 85:6548–56.

405

30.

406

dihydroorotate dehydrogenase in complex with antiproliferative agents. Structure 8:25–33.

407

31.

408

9:41–54.

409

32.

410

Construction and characterization of a stable subgenomic dengue virus type 2 replicon system

411

for antiviral compound and siRNA testing. Antiviral Res. 76:222–31.

412

33.

413

Short GF, Staunton JE, Jin X, Lee MS, Zimmermann GR, Borisy AA. 2009. Synergistic

414

drug combinations tend to improve therapeutically relevant selectivity. Nat. Biotechnol. 27:659–

415

66.

Crotty S, Cameron CE, Andino R. 2001. RNA virus error catastrophe: direct molecular

Thomas E, Feld JJ, Li Q, Hu Z, Fried MW, Liang TJ. 2011. Ribavirin potentiates

Qing M, Zou G, Wang Q-Y, Xu HY, Dong H, Yuan Z, Shi P-Y. 2010.

Wang Q-Y, Bushell S, Qing M, Xu HY, Bonavia A, Nunes S, Zhou J, Poh MK,

Liu S, Neidhardt EA, Grossman TH, Ocain T, Clardy J. 2000. Structures of human

Batt DG. 1999. Inhibitors of dihydroorotate dehydrogenase. Expert Opin. Ther. Pat.

Ng CY, Gu F, Phong WY, Chen Y-L, Lim SP, Davidson A, Vasudevan SG. 2007.

Lehár J, Krueger AS, Avery W, Heilbut AM, Johansen LM, Price ER, Rickles RJ,

19

416

34.

417

synergism and antagonism in drug combination studies. Pharmacol. Rev. 58:621–81.

418

35.

419

Assessing in vitro combinations of antifungal drugs against yeasts and filamentous fungi:

420

comparison of different drug interaction models. Med. Mycol. 43:133–152.

421

36.

422

53.

423

37.

424

Janssen HLA, van der Laan LJW. 2012. Mycophenolic acid augments interferon-stimulated

425

gene expression and inhibits hepatitis C Virus infection in vitro and in vivo. Hepatology

426

55:1673–83.

427

38.

428

nucleoside and nucleotide polymerase inhibitors and their use in treating hepatitis C virus. Expert

429

Opin. Investig. Drugs 1–13.

430

39.

431

B, Gorovits E, Ganguly B, Hall A, Kolykhalov A, Liu Y, Muhammad J, Raja N, Walters

432

CR, Wang J, Williams K, Patti JM, Henson G, Madela K, Aljarah M, Gilles A, McGuigan

433

C. 2011. INX-08189, a phosphoramidate prodrug of 6-O-methyl-2’-C-methyl guanosine, is a

434

potent inhibitor of hepatitis C virus replication with excellent pharmacokinetic and

435

pharmacodynamic properties. Antimicrob. Agents Chemother. 55:1843–51.

436

40.

437

zalcitabine and lamivudine by a CTP synthase inhibitor, 3-deazauridine. Nucleosides.

438

Nucleotides Nucleic Acids 19:371–7.

Chou T-C. 2006. Theoretical basis, experimental design, and computerized simulation of

Meletiadis J, Verweij PE, Te Dorsthorst DT a., Meis JFGM, Mouton JW. 2005.

Geary N. 2013. Understanding synergy. Am. J. Physiol. Endocrinol. Metab. 304:E237–

Pan Q, de Ruiter PE, Metselaar HJ, Kwekkeboom J, de Jonge J, Tilanus HW,

Gentile I, Coppola N, Buonomo AR, Zappulo E, Borgia G. 2014. Investigational

Vernachio JH, Bleiman B, Bryant KD, Chamberlain S, Hunley D, Hutchins J, Ames

Gao WY, Johns DG, Mitsuya H. 2000. Potentiation of the anti-HIV activity of

20

439

41.

440

of their pairwise interactions. Nat. Genet. 38:489–94.

441

42.

442

Yokohama F, Wang G, Cerny D, Fink K, Blasco F, Shi P-Y. 2014. Activation of peripheral

443

blood mononuclear cells by dengue virus infection depotentiates balapiravir. J. Virol. 88:1740–7.

444

43.

445

Susceptibility and response of human blood monocyte subsets to primary dengue virus infection.

446

PLoS One 7:e36435.

447

44.

448

Chua LT, Hou Y, Chow A, Howe S, Chan WK, Tan KH, Chung JS, Cherng BP, Lye DC,

449

Tambayah P a, Ng LC, Connolly J, Hibberd ML, Leo YS, Cheung YB, Ooi EE, Vasudevan

450

SG. 2014. Efficacy and safety of celgosivir in patients with dengue fever (CELADEN): a phase

451

1b, randomised, double-blind, placebo-controlled, proof-of-concept trial. Lancet Infect. Dis.

452

14:706–715.

453

45.

454

combination therapy in the quest for clinical efficacy. Nat. Chem. Biol. 2:458–66.

455

46.

456

Johansen LM. 2010. Chemical combinations elucidate pathway interactions and regulation

457

relevant to Hepatitis C replication. Mol. Syst. Biol. 6:375.

Yeh P, Tschumi AI, Kishony R. 2006. Functional classification of drugs by properties

Chen Y-L, Abdul Ghafar N, Karuna R, Fu Y, Lim SP, Schul W, Gu F, Herve M,

Wong KL, Chen W, Balakrishnan T, Toh YX, Fink K, Wong S-C. 2012.

Low JG, Sung C, Wijaya L, Wei Y, Rathore APS, Watanabe S, Tan BH, Toh L,

Fitzgerald JB, Schoeberl B, Nielsen UB, Sorger PK. 2006. Systems biology and

Owens CM, Mawhinney C, Grenier JM, Altmeyer R, Lee MS, Borisy AA, Lehár J,

458

21

459

FIGURE LEGENDS

460

Figure 1. Host nucleoside inhibitors and pre-clinical nucleoside analogues exhibit potent anti-

461

DENV replication activity with minimal cyototoxicty in cell culture. (A) The chemical structure

462

of ribavirin, a well-characterized inhibitor of IMPDH, is shown. The EC50 of ribavirin in the

463

Huh-7 NGC replicon is about 8.29 µM and the EC90 is about 61.77 µM. CC50 of ribavirin in

464

Huh-7-NGC replicon is more than 850 µM. (B) The structure of brequinar, a potent DHODH

465

inhibitor, is shown. Its EC50 in Huh-7 NGC replicon is about 51.5 nM and the EC90 is about

466

248.8 nM. The CC50 of brequinar in the Huh-7 NGC replicon is more than 5 µM. (C) The

467

chemical structure of the guanosine analog, INX-08189, is shown. The EC50 of INX-08189 in

468

the Huh-7 NGC replicon is about 14.48 nM and the EC90 is about 131.5 nM. The CC50 of INX-

469

08189 in the Huh-7 NGC replicon is more than 1 µM. (D) The EC50, EC90 and CC50 of

470

respective compounds are summarized, demonstrating that all the selected compounds have anti-

471

DENV activities with good therapeutic window in cell culture. The data is derived the average of

472

three to five independent experiments.

473 474

Figure 2. Chalice analysis of ribavirin and INX-08189 combination treatment on NGC replicon.

475

Luciferase activity was measured 48 h post-treatment with dosing matrix centered on the EC50

476

of each compound in the Huh-7 NGC replicon. (A) Dose matrix response values of the

477

combination showing the inhibition of luciferase activity (1 – treated / untreated) for the serially

478

diluted compounds. Inhibition values were analyzed by the Chalice software to generate

479

combinations’ excess values (the Loewe excess values are shown here). (B) The computed Bliss

480

and highest single agent (HSA) excess of each compound combination. Values are derived from

481

an average of 6 experiments. (C) Isobolograms, synergy score and best combination index (CI) at

22

482

90% inhibition of ribavirin and INX-08189. Values shown are derived from average of eight

483

experiments. (D) Cell viability of drug combination treatment. The concentrations of ribavirin

484

and INX-08189 were identical to those used in the dose matrix for antiviral assay (A). The

485

synergy calculation was derived from average of 8 datasets.

486 487

Figure 3. Dose-dependent induction of ISRE by IFN-β and nucleoside synthesis inhibitors. HEK

488

293T cells batch-transfected with pISRE-TA-Luc and pGL4.74-hRluc/TK, were simultaneously

489

treated with increasing concentrations of IFN-β, brequinar or ribavirin in individual wells. (A)

490

Treatment with IFN-β caused a maximum of 4-fold increase in ISRE activation while treatment

491

with (B) brequinar caused a maximum of 3-fold increase in ISRE activation. (C) Activation of

492

ISRE by ribavirin was modest, showing a maximum of 1.68-fold. (D) The induction of ISRE

493

activity by either host nucleoside inhibitors was not due to cytotoxicity as their CC50 values in

494

the HEK293-T cells were > 5 µM. The average results from two experiments are shown.

495 496

Figure 4. Induction of ISRE by IFN-β is potentiated in the presence of DHODH inhibitors. HEK

497

293-T cells were batch transfected with pISRE-TA-Luc and pGL4.74-hRluc/TK. Transfected

498

cells were simultaneously treated with increasing concentrations of IFN-β and the indicated

499

concentrations of respective compounds. 48 h post-treatment, luciferase activities were assayed

500

using the Dual-Glo Stop & Glo reagent. Co-treatment of HEK293T cells with brequinar and

501

5000 and 10, 000 IU/ml of IFN-β, significantly potentiated ISRE activation when compared to

502

DMSO control. There was no potentiation observed at lower doses of IFN-β. In contrast, co-

503

treatment of cells with ribavirin and IFN-β did not augment ISRE activation. Treatment of cells

23

504

with another pyrimidine biosynthesis inhibitor, NITD-982, with 10, 00 IU/ml of IFN-β, also

505

significantly augmented the ISRE activation.

24

Luminescence (%)

A

Ribavirin (log µM)

Luminescence (%)

B

Brequinar (log µM)

Luminescence (%)

C

Brequinar (µM)

EC50 = 14.2 ± 5.6 nM

INX-08189 (log µM)

INX-08189

D

EC50 = 53.4 ± 2.7 nM

Cell viability (%)

Brequinar

Ribavirin (µM)

Cell viability (%)

Ribavirin

EC50 = 3.18 ± 2.86 µM

Cell viability (%)

Figure 1

INX-08189 (µM)

Compound

EC50 (nM)

CC50 (µM)

Ribavirin

3,178 ± 2860

> 800

Brequinar

53.4 ± 2.7

>5

INX-08189

14.2 ± 5.6

>1

Figure 2 Loewe excess

Dose matrix

11 .14

1.2

Ribavirin (µM)

11 1.2 0

0

.14

Ribavirin (µM)

100

100

A

0

0.014

0.12

0.11

0

1

0.014

B

1

100 11 0

0.14

1.2

Ribavirin (µM)

100 11 1.2

Ribavirin (µM)

0.14 0

0.012

0.11

0 0.0014

1

Cytotoxicity of synergy data

11

59

62

65

70

75

74

69

59

71

73

75

81

88

93

88

71

89

90

93

98 102 110 104 84

99 102 104 105 108 112 107 88 1.2

Ribavirin (µM)

100

D

103 105 107 110 113 114 110 93 100 101 105 104 106 109 107 94 100 101 101 103 106 112 108 97 100 102 101 103 108 110 108 92 0

INX-08189 (0.11 µM)

1

.14

Synergy score: 2.2 ± 0.0475 Best CI: 0.325 ± 0.0143

0.11

0

Isobologram

0.012

INX-08189 (µM)

INX-08189 (µM)

Ribavirin (100 µM)

0.11

Huh-7 NGC replicon HSA excess

Huh-7 NGC replicon Bliss excess

0 0.0014

C

0.12

INX-08189 (µM)

INX-08189 (µM)

0.014

0.12

0.11

INX-08189 (µM)

1

Cell viability (%)

Cell viability (%)

ISRE activity (fold increase) ISRE activity (fold increase)

ISRE activity (fold increase)

Figure 3

A

B IFN-β (IU/mL)

C Brequinar (µM)

D Ribavirin (µM)

Brequinar (µM)

Ribavirin (µM)

Figure 4 1 % DMSO 1 mM Ribavirin 5 µM Brequinar 10 µM NITD-982

15 10

*** ***

*** p value < 0.001 ***

5

IFN- (IU / mL)

0 10 00

50 00

25 00

12 50

0

0

Fold-induction of ISRE

20

Synergistic suppression of dengue virus replication using a combination of nucleoside analogs and nucleoside synthesis inhibitors.

Dengue virus (DENV) is the most prevalent mosquito-borne viral pathogen in humans. Currently, there is no clinically approved vaccine or antiviral for...
1MB Sizes 0 Downloads 7 Views