+ All Categories
Home > Documents > Synthesis and evaluation of a new phosphorylated ribavirin prodrug

Synthesis and evaluation of a new phosphorylated ribavirin prodrug

Date post: 08-Dec-2016
Category:
Upload: mel
View: 215 times
Download: 1 times
Share this document with a friend
9
1 3 Synthesis and evaluation of a new phosphorylated ribavirin prodrug 4 5 6 Steven D. Dong a,Q1 , Chin-Chung Lin a , Mel Schroeder b 7 a Epiphany Biosciences, Inc., San Francisco, CA 94111, United States 8 b Kalexsyn, Inc., 4502 Campus Drive, Kalamazoo, MI 49008, United States 9 10 12 article info 13 Article history: 14 Received 6 September 2012 15 Revised 12 April 2013 16 Accepted 15 April 2013 17 Available online xxxx 18 Keywords: 19 Ribavirin 20 Prodrug 21 Antiviral drug 22 Hepatitis C virus 23 Red blood cell 24 Cellular uptake 25 26 abstract 27 Ribavirin is an important broad-spectrum antiviral drug. However, its utilization can be limited by its 28 potential to cause hemolytic anemia as well as its variability in dosing levels and efficacy outcomes. 29 To overcome these issues, we report on a new alkoxyalkylphosphodiester prodrug of ribavirin (2) that 30 is designed to release the active ribavirin-monophosphate species selectively in nucleated cells while 31 limiting its exposure in anucleated red blood cells (RBCs). Prodrug 2 displays improved in vitro antiviral 32 activity against the hepatitis C virus replicon and influenza virus. Unlike ribavirin, prodrug 2 does not sig- 33 nificantly decrease ATP levels in RBCs. Prodrug 2 demonstrates decreased uptake in RBCs but increased 34 uptake in HepG2 hepatocytes when compared to ribavirin. In vivo, prodrug 2 is orally bioavailable and 35 well-tolerated in rats in which it is processed to ribavirin and accumulates in the liver. These results indi- 36 cate that prodrug 2 has the potential for safer, lower, less frequent, and less variable administration than 37 ribavirin. 38 Ó 2013 Published by Elsevier B.V. 39 40 41 1. Introduction 42 Ribavirin was first disclosed in 1972 but still remains the only 43 approved small molecule antiviral drug with broad-spectrum 44 activity against both RNA and DNA viruses (Sidwell et al., 1972). 45 Clinically, ribavirin is used to treat hepatitis C virus (HCV) in com- 46 bination with peginterferon, respiratory syncytial virus, and Lassa 47 fever virus infections (Crotty et al., 2002). Recent data suggest that 48 ribavirin may continue to play a role in reducing relapse and 49 enhancing a sustained virological response in future therapeutic 50 modalities to treat HCV infection (Feld, 2012; Pockros, 2010; Shiff- 51 man, 2009). 52 Despite its clinical utility, ribavirin’s mechanism of action has 53 remained enigmatic (Parker, 2005). Its antiviral activity has been 54 attributed to both direct and indirect mechanisms that target a 55 variety of viral and host enzymes, which utilize purine nucleoside 56 or nucleotide as a substrate or cofactor. It is well recognized that 57 ribavirin must be phosphorylated to become pharmacologically 58 active. 59 A limitation to the clinical utility of ribavirin is that it can in- 60 duce hemolytic anemia (Russmann et al., 2006). Ribavirin is ac- 61 tively transported into red blood cells (RBCs) by equilibrative 62 nucleoside transporter 1 (ENT1) on the plasma membrane and is 63 subsequently metabolized to various phosphorylated derivatives 64 (Page and Connor, 1990). Since RBCs lack phosphatases, the result- 65 ing phosphorylated ribavirin species become trapped intracellu- 66 larly and accumulate, resulting in very high intra-RBC 67 concentrations that can exceed 1 mM (Inoue et al., 2006). The high 68 intracellular ribavirin concentrations in RBCs competitively de- 69 plete ATP levels, which leads to hemolytic anemia. 70 Additionally, recent studies suggest that ENT1-mediated uptake 71 of ribavirin in hepatocytes may play a pivotal role in its antiviral 72 effectiveness (Bengsch and Thimme, 2010; Ibarra and Pfeiffer, 73 2009; Iikura et al., 2012). The variability in HCV patient response 74 to ribavirin may be due in part to the existence of different ENT1 75 isoforms in human hepatocytes and their varying capacities to 76 transport ribavirin (Fukuchi et al., 2010). Therefore, a ribavirin ana- 77 log that does not solely depend on ENT1 uptake in hepatocytes 78 may have a more uniform effect in patients expressing different 79 isoforms. 80 We sought to develop a new prodrug of ribavirin that would 81 have the potential for improved efficacy, safety, and dosing. Since 82 ribavirin’s mechanism(s) of action requires phosphorylation, our 83 first consideration was the pre-installation of a phosphate group 84 so that the active species could be directly introduced to infected 85 cells and bypass the rate-limiting phosphorylation to ribavirin- 86 monophosphate (1, Fig. 1). Unprotected phosphorylated com- 87 pounds do not readily cross cell membranes, so a suitable prodrug 88 approach was required (He et al., 2007). Hostetler et al. have previ- 89 ously reported an alkoxyalkylphosphonate ester prodrug approach 90 to improve the bioavailability of acyclic nucleoside phosphorous 91 containing drugs such as cidofovir (Hostetler, 2009, 2010; Hostetler 92 et al., 2000, 2001). The long-chain alkoxyalkylphosphonate ester 0166-3542/$ - see front matter Ó 2013 Published by Elsevier B.V. http://dx.doi.org/10.1016/j.antiviral.2013.04.014 Corre Q2 sponding author. Address: Steven Dong, Epiphany Biosciences, 1 California Street, Suite 2800, San Francisco, CA 94111, United States. Tel.: +1 415 765 7193; fax: +1 415 765 7200. Q3 E-mail addresses: [email protected], [email protected] (S.D. Dong). Antiviral Research xxx (2013) xxx–xxx Contents lists available at SciVerse ScienceDirect Antiviral Research journal homepage: www.elsevier.com/locate/antiviral AVR 3191 No. of Pages 9, Model 5G 29 April 2013 Please cite this article in press as: Dong, S.D., et al. Synthesis and evaluation of a new phosphorylated ribavirin prodrug. Antiviral Res. (2013), http:// dx.doi.org/10.1016/j.antiviral.2013.04.014
Transcript
Page 1: Synthesis and evaluation of a new phosphorylated ribavirin prodrug

1

3

4

5

6 Q1

78

910

1 2

1314151617

1819202122232425

2 6

40

41

42

43

44

45

46

47

48

49

50

51

52

53

54

55

56

57

58

59

60

61

62

63

Q2

Q3

Antiviral Research xxx (2013) xxx–xxx

AVR 3191 No. of Pages 9, Model 5G

29 April 2013

Contents lists available at SciVerse ScienceDirect

Antiviral Research

journal homepage: www.elsevier .com/locate /ant iv i ra l

Synthesis and evaluation of a new phosphorylated ribavirin prodrug

0166-3542/$ - see front matter � 2013 Published by Elsevier B.V.http://dx.doi.org/10.1016/j.antiviral.2013.04.014

⇑ Corresponding author. Address: Steven Dong, Epiphany Biosciences, 1 CaliforniaStreet, Suite 2800, San Francisco, CA 94111, United States. Tel.: +1 415 765 7193;fax: +1 415 765 7200.

E-mail addresses: [email protected], [email protected] (S.D. Dong).

Please cite this article in press as: Dong, S.D., et al. Synthesis and evaluation of a new phosphorylated ribavirin prodrug. Antiviral Res. (2013),dx.doi.org/10.1016/j.antiviral.2013.04.014

Steven D. Dong a,⇑, Chin-Chung Lin a, Mel Schroeder b

a Epiphany Biosciences, Inc., San Francisco, CA 94111, United Statesb Kalexsyn, Inc., 4502 Campus Drive, Kalamazoo, MI 49008, United States

272829303132333435363738

a r t i c l e i n f o

Article history:Received 6 September 2012Revised 12 April 2013Accepted 15 April 2013Available online xxxx

Keywords:RibavirinProdrugAntiviral drugHepatitis C virusRed blood cellCellular uptake

a b s t r a c t

Ribavirin is an important broad-spectrum antiviral drug. However, its utilization can be limited by itspotential to cause hemolytic anemia as well as its variability in dosing levels and efficacy outcomes.To overcome these issues, we report on a new alkoxyalkylphosphodiester prodrug of ribavirin (2) thatis designed to release the active ribavirin-monophosphate species selectively in nucleated cells whilelimiting its exposure in anucleated red blood cells (RBCs). Prodrug 2 displays improved in vitro antiviralactivity against the hepatitis C virus replicon and influenza virus. Unlike ribavirin, prodrug 2 does not sig-nificantly decrease ATP levels in RBCs. Prodrug 2 demonstrates decreased uptake in RBCs but increaseduptake in HepG2 hepatocytes when compared to ribavirin. In vivo, prodrug 2 is orally bioavailable andwell-tolerated in rats in which it is processed to ribavirin and accumulates in the liver. These results indi-cate that prodrug 2 has the potential for safer, lower, less frequent, and less variable administration thanribavirin.

� 2013 Published by Elsevier B.V.

39

64

65

66

67

68

69

70

71

72

73

74

75

76

77

78

79

80

81

82

83

84

85

86

87

1. Introduction

Ribavirin was first disclosed in 1972 but still remains the onlyapproved small molecule antiviral drug with broad-spectrumactivity against both RNA and DNA viruses (Sidwell et al., 1972).Clinically, ribavirin is used to treat hepatitis C virus (HCV) in com-bination with peginterferon, respiratory syncytial virus, and Lassafever virus infections (Crotty et al., 2002). Recent data suggest thatribavirin may continue to play a role in reducing relapse andenhancing a sustained virological response in future therapeuticmodalities to treat HCV infection (Feld, 2012; Pockros, 2010; Shiff-man, 2009).

Despite its clinical utility, ribavirin’s mechanism of action hasremained enigmatic (Parker, 2005). Its antiviral activity has beenattributed to both direct and indirect mechanisms that target avariety of viral and host enzymes, which utilize purine nucleosideor nucleotide as a substrate or cofactor. It is well recognized thatribavirin must be phosphorylated to become pharmacologicallyactive.

A limitation to the clinical utility of ribavirin is that it can in-duce hemolytic anemia (Russmann et al., 2006). Ribavirin is ac-tively transported into red blood cells (RBCs) by equilibrativenucleoside transporter 1 (ENT1) on the plasma membrane and issubsequently metabolized to various phosphorylated derivatives

88

89

90

91

92

(Page and Connor, 1990). Since RBCs lack phosphatases, the result-ing phosphorylated ribavirin species become trapped intracellu-larly and accumulate, resulting in very high intra-RBCconcentrations that can exceed 1 mM (Inoue et al., 2006). The highintracellular ribavirin concentrations in RBCs competitively de-plete ATP levels, which leads to hemolytic anemia.

Additionally, recent studies suggest that ENT1-mediated uptakeof ribavirin in hepatocytes may play a pivotal role in its antiviraleffectiveness (Bengsch and Thimme, 2010; Ibarra and Pfeiffer,2009; Iikura et al., 2012). The variability in HCV patient responseto ribavirin may be due in part to the existence of different ENT1isoforms in human hepatocytes and their varying capacities totransport ribavirin (Fukuchi et al., 2010). Therefore, a ribavirin ana-log that does not solely depend on ENT1 uptake in hepatocytesmay have a more uniform effect in patients expressing differentisoforms.

We sought to develop a new prodrug of ribavirin that wouldhave the potential for improved efficacy, safety, and dosing. Sinceribavirin’s mechanism(s) of action requires phosphorylation, ourfirst consideration was the pre-installation of a phosphate groupso that the active species could be directly introduced to infectedcells and bypass the rate-limiting phosphorylation to ribavirin-monophosphate (1, Fig. 1). Unprotected phosphorylated com-pounds do not readily cross cell membranes, so a suitable prodrugapproach was required (He et al., 2007). Hostetler et al. have previ-ously reported an alkoxyalkylphosphonate ester prodrug approachto improve the bioavailability of acyclic nucleoside phosphorouscontaining drugs such as cidofovir (Hostetler, 2009, 2010; Hostetleret al., 2000, 2001). The long-chain alkoxyalkylphosphonate ester

http://

Page 2: Synthesis and evaluation of a new phosphorylated ribavirin prodrug

93

94

95

96

97

98

99

100

101

102

103

104

105

106

107

108

109

110

111

112

113

114

115

116

117

118

119

120

121

122

123

124

125

126

127

128

129

130

131

132

133

134

135

136

137

138

139

140

141

142

143

144

145

146

147

148

149

150

151

152

153

O

P

OH

OH NOO

OH

NNH2

N

O

OH

1

No Conversion

O

P

OH

O NOO

OH

NNH2

N

O

OH

R

Hepatocyte

Red Blood Cell

Nucleate

Anucleate 2R = -CH2CH2O(CH2)17CH3

PLC

Fig. 1. Proposed mechanism of intracellular cleavage of octadecyloxyethyl ribavirin phosphodiester prodrug 2 by phospholipase C (PLC) yields ribavirin mono-phosphate (1)in nucleated cells such as hepatocytes containing PLC but not in anucleated cells such as red blood cells which do not have PLC.

2 S.D. Dong et al. / Antiviral Research xxx (2013) xxx–xxx

AVR 3191 No. of Pages 9, Model 5G

29 April 2013

prodrug moiety mimics lysophosphoplipids which are readily takenup in the gastrointestinal tract and into cells. After being taken up inthe cell, the alkoxyalkylphosphonate ester prodrug moiety is thencleaved by phospholipase C (PLC) to release the unprotected drugas the free phosphonic acid.

We hypothesized that a similar prodrug approach would beapplicable to the ribavirin-monophosphate nucleotide 1 and couldprovide several advantages, most notably a differential prodrugactivation profile in nucleated cells versus anucleated cells(Fig. 1). 1H spin-echo NMR experiments in human RBC lysates dem-onstrated that PLC is not present in anucleated RBCs (Selle et al.,1992). Consequently, PLC-mediated cleavage of an alkoxyalkyl pro-drug in RBCs is not possible. However, in nucleated cells such ashepatocytes where PLC is present, an alkoxyalkyl group can becleaved to yield 1. Direct introduction of the active species into tar-get nucleated cells without concurrent processing in anucleatedRBCs could result in an overall improved therapeutic index. Herein,we report a new ribavirin prodrug (2) and its preliminary in vitroand in vivo characterization.

154

155

156

157

158

159

160

161

162

163

164

165

166

167

168

169

170

171

172

173

174

175

176

177

178

179

180

2. Materials and methods

2.1. Compounds and radiochemicals

1-{5-O-[(2-chlorophenoxy)(octadecyloxy)phosphoryl]-2,3-O-iso-propylidene-beta-D-ribofuranosyl}-1H-1,2,4-triazole-3-carboxamide(4). Triazole (0.146 g, 2.12 mmol), triethylamine (0.215 g,2.12 mmol), and dry THF (2.1 mL) were added to 2-chlorophenylphosphorodichloridate (0.259 g, 1.06 mmol) in dry THF (1.3 mL).The reaction was stirred at room temperature for 1 h, filtered,and washed with THF (2.1 mL). 20,30-Isopropylidene ribavirin (3,0.226 g, 0.795 mmol, Toronto Research Chemicals; Toronto, ON,Canada) and 1-methylimidazole (0.084 mL, 1.06 mmol) in THF(1.2 mL) were added to the filtrate and stirred at room temperaturefor 1 h. 2-(octadecyloxy)ethanol (0.250 g, 0.795 mmol) was added,stirred at room temperature overnight, and concentrated in vacuo.Purification by flash chromatography (CH2Cl2 + 0 to 4% MeOH) andconcentration in vacuo provided 4 as a colorless oil (0.439 g, 71%).See Supplementary Information for spectral characterization.

1-(5-O-{hydroxy[2-(octadecyloxy)ethoxy]phosphoryl}-2,3-O-iso-propylidene-beta-D-ribofuranosyl)-1H-1,2,4-triazole-3-carboxamide(5). 1,1,3,3-tetramethylguanidine (0.378 g, 3.28 mmol) and syn-2-pyridinealdoxime (0.401 g, 3.28 mmol) in THF (4.2 mL) were addedto a solution of 4 (0.448 g, 0.581 mmol) in THF (12.2 mL) and stir-red at room temperature overnight. The reaction was concentratedin vacuo, and the residue was purified by flash chromatography(CH2Cl2 + 0 to 30% MeOH). Following concentration, the resulting

Please cite this article in press as: Dong, S.D., et al. Synthesis and evaluationdx.doi.org/10.1016/j.antiviral.2013.04.014

material was partitioned between THF: EtOAc (1:1) and cold H2O(pH 1–2), extracted thrice with THF: EtOAc, dried with Na2SO4 con-centrated in vacuo, and azeotroped from MeOH to yield 5 as awhite solid (0.312 g, 81%).

1-(5-O-{hydroxy[2-(octadecyloxy)ethoxy]phosphoryl}-beta-D-ribofuranosyl)-1H-1,2,4-triazole-3-carboxamide (2). Acetonide 5(0.304 g, 0.460 mmol) was dissolved in TFA:H2O (9:1, 4 mL) andstirred at room temperature for 45 min. The reaction was concen-trated, azeotroped from toluene, suspended in MeOH, and concen-trated thrice to produce prodrug 2 as a white solid (0.264 g, 93%).

5-[14C]-ribavirin with a specific activity of 55.8 mCi/mmol waspurchased from Moravek Biochemicals (Brea, CA). 5-[14C]-2 witha specific activity of 56.3 mCi/mmol was prepared from 5-[14C]-ribavirin according to the previous methods.

2.2. In vitro assays

2.2.1. Antiviral assaysHCV antiviral activity was assessed according to a previously re-

ported method in the stably HCV RNA-replicating cell line AVA5(genotype 1b, subgenomic replicon) and APC103 (genotype 1a,subgenomic replicon) (Okuse et al., 2005; Blight et al., 2000). Com-pounds were added to dividing cultures daily for three days withcultures generally at 30–50% confluence at assay initiation andreaching confluence during the last day of treatment. IntracellularHCV RNA levels and cytotoxicity were assessed 72 h after treat-ment. Intracellular HCV RNA levels were measured using a conven-tional blot hybridization method in which HCV RNA wasnormalized to b-actin mRNA for each individual culture. Cytotoxic-ity was measured using a previously reported neutral red dye up-take assay (Korba and Gerin, 1992).

Anti-influenza A activity was determined against influenza A/Hong Kong/8/68 (ATCC; Manassas, VA) in Madin-Darby canine kid-ney (MDCK) cells (ATCC) using a previously reported colorimetricreadout utilizing the CellTiter 96� AQueous One Solution Cell Pro-liferation Assay (Promega; Madison, WI) to determine virus-in-duced cytopathogenic effects (Fletcher et al., 2000). The MDCKcells were plated at 10,000 cells/well in 96 well plates with 2% ser-um as stationary monolayers for both antiviral and cytotoxicity as-says. Cytotoxicity was measured using the previously reportedCytoTox-ONE™ Homogeneous Membrane Integrity Assay(Promega).

2.2.2. Nucleotide pool assayHuh-7.5 cells (Dr. Ralf Bartenschlager; University of Heidelberg,

Germany) were incubated for 24 h with either ribavirin, prodrug 2,or dimethyl sulfoxide (DMSO) (vehicle control), and the resulting

of a new phosphorylated ribavirin prodrug. Antiviral Res. (2013), http://

Page 3: Synthesis and evaluation of a new phosphorylated ribavirin prodrug

181

182

183

184

185

186

187

188

189

190

191

192

193

194

195

196

197

198

199

200

201

202

203

204

205

206

207

208

209

210

211

212

213

214

215

216

217

218

219

220

221

222

223

224

225

226

227

228

229

230

231

232

233

234

235

236

237

238

239

240

241

242

243

244

245

246

247

248

249

250

251

252

253

254

255

256

257

258

259

260

261

262

263

264

265

266

267

268

269

270

271

272

273

274

275

276

277

278

279

280

281

282

283

284

285

286

287

288

289

290

291

292

293

294

295

296

297

298

299

S.D. Dong et al. / Antiviral Research xxx (2013) xxx–xxx 3

AVR 3191 No. of Pages 9, Model 5G

29 April 2013

nucleotide pools were extracted and analyzed by HPLC accordingto methods previously described (NovoCIB S.A.S.; Lyon, France)(Randall et al., 2003; Di Pierro et al., 1995).

2.2.3. ATP level determinationHuman RBCs were incubated with either 1 mM 2, ribavirin, or

DMSO (vehicle control) for 12 h employing previously disclosedconditions (De Franceschi et al., 2000). Just prior to ATP level deter-mination, a vehicle control sample was spiked with 1 mM ribavirinto normalize the observed luminescence for any interference fromribavirin itself. ATP levels were quantified using the CellTiter-Glo�

Luminescent Cell Viability Assay (Promega).

2.2.4. Uptake assays in RBCs and HepG2 cellsUptake of [14C]-2 or 5-[14C]-ribavirin into RBCs was determined

using a previously reported approach (Lin et al., 2003). Wholeblood was collected from human donors, the hematocrit (H) deter-mined, and the RBCs separated from plasma by centrifugation.Plasma was spiked with the radiolabeled test article to achieve atarget concentration of 100 lg (0.1 lCi)/mL in whole blood andgently mixed with blood cell suspension. Samples were incubatedin duplicate for 0, 0.5, 1, 2, 4 and 24 h. RBCs were separated fromplasma by centrifugation. Following RBC decolorization withH2O2, the concentration in the reconstituted blood (Cb) and plasma(Cp) was determined by liquid scintillation counting (LSC, PerkinEl-mer). The RBC concentration (Crbc) was calculated as follows:Crbc = [Cb � Cp(1 � H)]/H, and the ratios of Crbc/(Crbc + Cp) and Cp/(Crbc + Cp) were calculated.

To determine uptake in a cultured hepatocyte line, HepG2 cells(50,000 cells/well; ATCC) were incubated with either 100 lg(0.1 lCi)/mL [14C]-2 or 5-[14C]-ribavirin at 5% CO2 and 37 �C for24 h (Fig. 6). At various timepoints (0, 1, 2, 4, and 24 h), the super-natant was removed, cells were washed with cold PBS, and a totalcell lysate was prepared. Radioactivity in the supernatant (CSuperna-

tant) and the cell lysate (CHep) was counted by LSC, and the ratio ofCHep/(CHep + CSupernatant) was calculated.

2.3. In vivo studies

2.3.1. Animal studiesThe 28-day dosing study was conducted at Bridge Laboratories

(Beijing, China), and the tissue distribution study was conducted atXenometrics (Stilwell, KS). These test facilities were accredited bythe Association for Assessment and Accreditation of LaboratoryAnimal Care International, and all protocols were approved bythe facilities’s respective Institutional Animal Care and Use Com-mittees. All procedures involving animals were conducted huma-nely and were performed by or under the direction of trainedand experienced personnel. These studies did not unnecessarilyduplicate previous studies.

Sprague Dawley rats (Vital River Laboratory Animal Technol-ogy; Beijing, China) received either 2 (304.8 mg/kg/day) or ribavi-rin (120 mg/kg/day) daily by oral gavage for 28 consecutive days.Dosing formulations were prepared by dissolving an appropriateamount of test article into DMSO (10% of the final volume), adding5% glucose injection (90% of the final volume), and homogeniza-tion. Blood samples from three non-fasted animals/group at eachtime point were collected on Days 1, 14, and 28 prior to doseand at 1, 3, and 6 h post-dose. Parameters evaluated included bodyweights and food consumption, and macroscopic pathology. Plas-ma was analyzed by LC/MS/MS (Supplementary Information).

For the tissue distribution studies, [14C]-2 was formulated in40% (w/v%) hydroxypropyl-b-cyclodextrin in pH 7.4 buffer andadministered to Sprague Dawley rats at 10 mg/kg by oral gavagewith a total dosed radioactivity of approximately 20 lCi/kg.5-[14C]-Ribavirin was formulated in sterile water and was

Please cite this article in press as: Dong, S.D., et al. Synthesis and evaluationdx.doi.org/10.1016/j.antiviral.2013.04.014

administered at 10 mg/kg with a total dosed radioactivity ofapproximately 225 lCi/kg. Blood samples were collected at 1 and3 h into blood tubes containing K3EDTA. An aliquot of whole bloodwas kept, and the remaining blood was centrifuged to separate theplasma and RBCs. Following the terminal sacrifice at 1 and 3 h, theentire liver and lungs were collected. Samples were homogenizedand analyzed by LSC (Supplementary Information).

3. Results

3.1. Synthesis

A very efficient synthesis of 2 was developed from ribavirin(Fig. 2). The 20,30-protected ribavirin acetonide 4 was coupled with2-chlorophenyl phosphorchloridate via the phosphotriester ap-proach. In situ ester formation with hexadecyloxypropanol and10-methylimidazole produced phosphotriester 5. Sequential depro-tections provided the target prodrug 2 in 62.5% overall yield from 4at >95% purity by HPLC.

3.2. In vitro studies

3.2.1. Antiviral activity against HCV and influenza A virusProdrug 2 was screened for in vitro activity against HCV and

influenza virus. Prodrug 2 was approximately 10-fold more activethan ribavirin against the HCV 1b replicon and 6-fold more activeagainst the HCV 1a replicon. Prodrug 2 displayed an approximately13–25-fold improved selectivity index compared to ribavirin inthese assays (Table 1). Prodrug 2 also demonstrated an approxi-mately 74-fold enhanced activity against influenza A virus com-pared to ribavirin (Table 1). The improvement in in vitro activitywas consistent with the prodrug 2 being converted directly tothe pharmacologically species 1, thereby bypassing the rate-limit-ing phosphorylation step required by ribavirin.

3.2.2. Cellular GTP depletionInosine monophosphate (IMP) dehydrogenase controls the

guanine nucleotide pool by catalyzing the conversion of IMP toxanthosine monophosphate, the pivotal step in guanine nucleotidebiosynthesis. Inhibition of IMP dehydrogenase has been proposedas one of the mechanisms by which phosphorylated ribavirin 1 dis-rupts viral replication through interruption of host nucleotide bio-synthesis and/or viral genome transcription/translation (Dixit andPerelson, 2006). We examined prodrug 2’s effect on cellular GTPand nucleotide pool levels in a whole-cell assay (Fig. 3). Prodrug2 depleted cellular guanine nucleotide pools in a dose-dependentmanner. At a lower concentration of 30 lm, prodrug 2 reducedGTP levels similar to the more concentrated ribavirin control(100 lm). These results are consistent with 2 being released di-rectly as the pharmacologically active species 1.

3.2.3. ATP levels in RBCsThe effect of prodrug 2 on ATP levels in RBCs was evaluated

in vitro (Fig. 4). A previous study demonstrated that RBC ATP levelswere markedly reduced (>50%) after incubation with ribavirin(1 mM) for 12 h (De Franceschi et al., 2000). In our study, washedRBCs were incubated with either 2 or ribavirin. RBCs treated with 2displayed an ATP level nearly identical to the normalized controlwith the luminescence dropping only 3%. In contrast, the ribavi-rin-treated RBCs showed an approximately 73% reduction in ATPlevels compared to the normalized control, a result consistent withprevious reports (De Franceschi et al., 2000). These results indicatethat 2 does not significantly reduce ATP levels and therefore is notreadily metabolized to ribavirin and/or 1 in RBCs.

of a new phosphorylated ribavirin prodrug. Antiviral Res. (2013), http://

Page 4: Synthesis and evaluation of a new phosphorylated ribavirin prodrug

300

301

302

303

304

305

306

307

308

309

310

311

312

313

314

315

316

317

318

319

320

321

322

323

324

325

326

327

328

329

330

331

332

333

334

335

336

3

NO

OH

O

N

NH2N

O

O

Cl O P

O

ClCl

1.

1,2,4-triazole, THF, NEt3

2. 1-methylimidazole3. CH3(CH2)17)CH2CH2OH

O

ClO

P

O

NO

O

O

N

NH2N

O

O

R

4R = -CH2CH2O(CH2)17CH3

OOH

P

ON

OO

OH

N

NH2N

O

OH

R

2R = -CH2CH2O(CH2)17CH3

OOH

P

O

NO

O

O

N

NH2N

O

O

R

5R = -CH2CH2O(CH2)17CH3

syn-2-Pyridinealdoxime

TMG, THF

TFA, H2O

Fig. 2. Preparation of prodrug 2. See text for details.

Table 1In vitro antiviral activity of prodrug 2 versus ribavirin.

Virus Compound EC50 (lM) (SD) CC50 (lM) (SD) Selectivity index

HCV replicon (genotype 1b) Ribavirin (1) 31 ± 3.5 76 ± 2.1 2.5Prodrug 2 2.9 ± 0.5 182 ± 4.1 63

HCV replicon (genotype 1a) Ribavirin (1) 38 ± 1.2 78 ± 1.1 2.1Prodrug 2 6.3 ± 0.9 185 ± 3.2 29

Influenza A (HK strain H3N2) Ribavirin (1) 37 >410 >11Prodrug 2 0.5 66 132

HCV and influenza assays were performed in triplicate. Cytotoxicity assays were performed in triplicate for HCV and in duplicate for influenza. Values presented (± standarddeviations [S.D.]) were calculated by linear regression analysis using data combined from all treated cultures. S.D. was calculated using the standard error of regressiongenerated from the linear regression analyses (QuattroProTM). EC50, drug concentration at which a 2-fold depression of HCV RNA (relative to the average levels in untreatedcultures) or the minimum inhibitory drug concentration which reduces the CPE by 50%; CC50, drug concentration at which a 2-fold depression of neutral red dye uptake(relative to the average levels in untreated cultures) was observed in AVA5 or APC103 cells or minimum toxic drug concentration which causes the reduction of viable MDCKcells by 50%.

4 S.D. Dong et al. / Antiviral Research xxx (2013) xxx–xxx

AVR 3191 No. of Pages 9, Model 5G

29 April 2013

3.2.4. In vitro uptake studies in RBCs and HepG2 cellsThe uptake of 2 and ribavirin in RBCs was determined using

radiolabeled analogs (Fig. 5) (Yeh et al., 2005; Lin et al., 2003). 5-[14C]-ribavirin was rapidly transported from human plasma intoRBCs with nearly 75% of the radioactivity partitioning into RBCsin just two hours. In the [14C]-2-treated plasma, only approxi-mately 7% of the radioactivity was transported over 4 h into RBCs,indicating that 2 is not readily taken up from plasma. It is possiblethat the lipophilic nature of the prodrug may cause it to be plasmaprotein bound which may contribute to the limited observed up-take into RBCs.

The uptake of 2 and ribavirin in the HepG2 human liver carci-noma cell line was then evaluated (Fig. 6). HepG2 cells were incu-bated with either [14C]-2 or 5-[14C]-ribavirin for 24 h. The observeduptake levels of ribavirin in HepG2 cells were consistent with pre-viously reported in vitro results using [3H-]-ribavirin (Lau andHong, 2004; Ibarra and Pfeiffer, 2009). The uptake level of 2 wasmore than 10-fold higher than ribavirin, suggesting that theprodrug is not solely dependent on ENT1 active transport. Basedon the total radioactivity recovered, a significant amount of

Please cite this article in press as: Dong, S.D., et al. Synthesis and evaluationdx.doi.org/10.1016/j.antiviral.2013.04.014

non-specific binding of 2 to the polystyrene plate (approximately28%) was observed. Therefore, the observed uptake may only rep-resent the lower limit of the actual uptake. The higher uptake of 2in HepG2 cells is consistent with its improved antiviral efficacy aswell as 2’s improved effect on nucleotide pools.

3.3. In vivo studies

3.3.1. Plasma stabilityPrior to in vivo evaluations, the stability of 2 was evaluated in

rat plasma at 37 �C over 6 h (Supplementary Information). Therewas no trend of decreasing concentrations of 2 nor was there anydetectable ribavirin over the course of the study, indicating that2 was stable in rat plasma (data not shown).

3.3.2. 28-Day dosing in ratsTo evaluate prodrug 2 in vivo, male Sprague Dawley rats were

dosed orally either with 2 (304.8 mg/kg) or a molar equivalent dos-age of ribavirin (120 mg/kg/day) once-daily for 28 consecutivedays (Lin et al., 2003; Dadgostari et al., 2004). At these high doses,

of a new phosphorylated ribavirin prodrug. Antiviral Res. (2013), http://

Page 5: Synthesis and evaluation of a new phosphorylated ribavirin prodrug

337

338

339

340

341

342

343

344

345

346

347

348

349

350

351

352

353

354

355

356

357

358

359

360

361

362

0.0

0.5

1.0

1.5

DMSO Prodrug(Ctrl) 2 (1mM) Ribavirin (1mM)

Mea

n A

TP C

once

ntra

tion

(uM

)

Fig. 4. The effect of prodrug 2, ribavirin, or DMSO on ATP concentrations in RBCs. Washed RBCs were incubated with either DMSO vehicle control, prodrug 2 (1 mM), orribavirin (1 mM) for 12 h and the resulting ATP levels were determined by CellTiter-Glo�. The experiment was performed in duplicate, and the DMSO control was normalizedfor ribavirin interference by adding ribavirin to the sample just prior to luminescence measurement. RBCs treated with two displayed an ATP level nearly identical to thenormalized control. Ribavirin-treated RBCs showed an approximately 73% reduction in ATP levels (mean values are shown; error bars represent S.D.).

0%

25%

50%

75%

100%

125%

150%

GTP ATP CTP UTP

% o

f Nat

ural

NTP

DM

SO C

ontro

l

Ribavirin (100µM)

2 (3µM)

2 (10µM)

2 (30µM)

Fig. 3. The effect of prodrug 2 or ribavirin on cellular GTP and nucleotide pool levels in a whole-cell assay. Huh-7.5 cells were incubated with DMSO (vehicle control),ribavirin (positive control), or prodrug 2 for 24 h and then extracted to collect nucleotide triphosphate (NTP) pools. In a single experiment, HPLC was used to identify andquantify various NTP levels based on characteristic UV absorption spectra and retention times. Percent changes in the NTP peak areas of the treated samples relative to theNTP peak areas found in the DMSO control are shown for ribavirin (100 lM, white bar) and prodrug 2 at three different concentrations (3 lM, solid black bar; 10 lM, grey bar;30 lM, cross-hatched bar). Two depleted GTP pools in a dose-dependent fashion but with modest effect on other NTP pools.

S.D. Dong et al. / Antiviral Research xxx (2013) xxx–xxx 5

AVR 3191 No. of Pages 9, Model 5G

29 April 2013

both prodrug 2 and ribavirin were generally well-tolerated andwithout mortality. Over the course of the study, lower bodyweights, lower body weight gains, and reduced food consumptionwere observed for both dosing groups. Plasma samples were col-lected at pre-dose and 1, 3, and 6 h post-dose on days 1, 14, and28 after dosing. Plasma levels of 2 and ribavirin were determinedsimultaneously using a single LCMS/MS method (SupplementaryInformation). Following oral dosing of 2, the prodrug was orally ab-sorbed and increasing plasma levels were observed over time,reaching a maximum at 6 h (Fig. 7a). Modest levels of ribavirin de-rived from 2 were also observed, confirming that 2 converted toribavirin in vivo. Over the course of the 28-day study, Areas Underthe Curve (AUC(0–24 h)) for both plasma ribavirin and 2 were deter-

Please cite this article in press as: Dong, S.D., et al. Synthesis and evaluationdx.doi.org/10.1016/j.antiviral.2013.04.014

mined at days 1, 14, and 28 (Fig. 7b). In the control group receivingribavirin, the plasma ribavirin AUCs(0–24 h) were: 18,787; 16,267;and 19,958 ng�h/mL for days 1, 14, and 28, respectively, indicatingthat steady-state plasma levels of ribavirin were obtained follow-ing the first dose (Fig. 7b). In the group receiving prodrug 2, theAUCs(0–24 h) for 2 were similar for days 1, 14, and 28 (31,678;30,323; 42,637 ng�h/mL), indicating that the 2 steady-state plasmaAUC(0–24 h) was reached immediately after the first dosage of 2(Fig. 7b). For the group treated with 2, the plasma ribavirinAUCs(0–24 h) were: 5946; 33,140; and 48,942 ng�h/mL for days 1,14, and 28 (Fig. 7b). Plasma ribavirin levels in the group receivingprodrug 2 increased and accumulated as a function of the admin-istration duration. Overall, 2 yielded a 104% higher plasma ribavi-

of a new phosphorylated ribavirin prodrug. Antiviral Res. (2013), http://

Page 6: Synthesis and evaluation of a new phosphorylated ribavirin prodrug

363

364

365

366

367

368

0%

20%

40%

60%

80%

100%

0 1 2 3 4Time (h)

% T

otal

Rad

ioac

tivity

Ribavirin (RBCs)

Prodrug 2 (RBCs)

Ribavirin (Plasma)

Prodrug 2 (Plasma)

Fig. 5. The uptake of [14C]-2 or 5-[14C]-ribavirin into RBCs. Uptake was determined by spiking plasma with the radiolabeled test article and then combining with the bloodcell suspension to achieve a target concentration of 100 lg (0.1 lCi)/mL in the reconstituted whole blood. Assays were performed in duplicate with three samples collectedper timepoint (0, 0.5, 1, 2, and 4 h) following RBC separation from plasma by centrifugation. The percent total radioactivity in the RBCs (solid line) and plasma (dashed line)was determined by LSC (mean values are shown; error bars represent S.D.).

0%

2%

4%

6%

8%

10%

12%

0 6 12 18 24Time (h)

% R

adio

activ

ity in

lysa

te

Ribavirin

Prodrug 2

Fig. 6. The uptake of [14C]-2 or 5-[14C]-ribavirin into liver cells. HepG2 cells were incubated with either 100 lg (0.1 lCi)/mL [14C]-2 or 5-[14C]-ribavirin for 24 h. Assays wereperformed in duplicate with three samples per timepoint (0, 0.5, 1, 2, 4, and 24 h) followed by removal of supernatant, cold PBS wash of cells, and a total cell lysatepreparation. The percent lysate radioactivity was determined by liquid scintillation counting (mean values are shown; error bars represent S.D.).

6 S.D. Dong et al. / Antiviral Research xxx (2013) xxx–xxx

AVR 3191 No. of Pages 9, Model 5G

29 April 2013

rin AUC(0–24 h) on day 14 and 147% higher plasma ribavirin AUC(0–

24 h) on day 28 than that administrated with equivalent ribavirincontrol.

Please cite this article in press as: Dong, S.D., et al. Synthesis and evaluationdx.doi.org/10.1016/j.antiviral.2013.04.014

3.3.3. Tissue distributionThe tissue distribution of 2 and ribavirin was studied in male

Sprague Dawley rats after a single oral dose of either [14C]-2 or

of a new phosphorylated ribavirin prodrug. Antiviral Res. (2013), http://

Page 7: Synthesis and evaluation of a new phosphorylated ribavirin prodrug

369

370

371

372

373

374

375

376

377

378

379

380

381

382

383

384

385

386

387

388

389

390

391

392

0

500

1000

1500

2000

2500

3000

0 2 4 6Time (h)

Plas

ma

Leve

ls (n

g/m

L)

Prodrug 2

Ribavirin (derived from 2)

Ribavirin (ctrl)

µ

0

10

20

30

40

50

60

Day 1 Day 14 Day 28

AUC (

0-24

hr) (

µg/m

L)

Ribavirin (ctrl)Ribavirin (derived from 2)Prodrug 2

(a) (b)

Fig. 7. The in vivo pharmacokinetic behavior of prodrug 2 and ribavirin. Sprague Dawley rats received equivalent molar dosages of either two (304.8 mg/kg) or ribavirin(120 mg/kg) by daily oral gavage for 28 consecutive days. The study was performed a single time with blood samples collected from three animals per timepoint per testarticle (prior to dose and at 1, 3, and 6 h post-dose) on days 1, 14, and 28. Plasma was analyzed by LC/MS/MS. (a) Day 1 plasma levels of prodrug 2 (solid line) and ribavirinderived from 2 (dashed line) in rats (n = 6) or plasma levels from the ribavirin control (alternating dashed line) in rats (n = 6) (mean values are shown; error bars representS.D.). (b) Plasma AUCs(0–24 h) for ribavirin control (solid white bars, n = 22); for ribavirin derived from two (cross-hatched bars, n = 22); for two (dashed line bars) on days 1, 14,and 28 (mean values are shown; error bars represent S.D.).

0

5

10

15

20

25

30

35

Plasma(1h)

Plasma(3h)

RBCs(1h)

RBCs(3h)

Liver(1h)

Liver(3h)

Lung(1h)

Lung(3h)

Matrix (sampling time pt)

Conc

entra

tion

( µg/

g)

[14C]-2[14C]-Ribavirin

Fig. 8. The tissue distribution of total radioactivity derived from [14C]-prodrug 2 or [14C]-ribavirin. Sprague Dawley rats received a single dose (10 mg/kg) of either [14C]-prodrug 2 (white bar, n = 4) or [14C]-ribavirin (cross-hatched bar, n = 4) by oral gavage. The study was performed a single time with two animals per timepoint per test article(at 1 and 3 h post-dose). Following terminal sacrifice at 1 or 3 h, blood, liver, and lung samples were collected, and the plasma and RBCs separated by centrifugation. Duplicatealiquots from each homogenized sample where quantified by LSC to determine radioactivity concentrations (lg equivalent/g) in plasma, red blood cells, liver and lung at 1and 3 h (mean values are shown; error bars represent S.D.).

S.D. Dong et al. / Antiviral Research xxx (2013) xxx–xxx 7

AVR 3191 No. of Pages 9, Model 5G

29 April 2013

5-[14C]-ribavirin (10 mg/kg). All animals exhibited normal behav-ior following dose administration and exhibited no overt signs oftoxicity throughout the study. Total radioactivity concentrationsin plasma, RBCs, liver and lung at 1 and 3 h were determined byLSC (Fig. 8), but specific species such as 2, 1, ribavirin, and othermetabolites were not explicitly identified in the study. For both 2and ribavirin, liver exhibited the highest radioactivity concentra-tions followed by lung. After dosing of prodrug [14C]-2, radioactiv-ity concentrations approximately doubled from 1 to 3 h in alltissues, with liver radioactivity increasing from 11.5 lg equiva-lent/g at 1 h to 28.5 lg equivalent/g at 3 h. On the other hand, after[14C]-ribavirin administration, radioactivity concentrations

Please cite this article in press as: Dong, S.D., et al. Synthesis and evaluationdx.doi.org/10.1016/j.antiviral.2013.04.014

decreased from 1 to 3 h in all tissues except for lung which re-mained steady. In particular, the liver radioactivity decreased from29.4 lg equivalent/g at 1 h to 18.2 lg equivalent/g at 3 h. Thesedata are consistent with the pharmacokinetic study results(Fig. 7a) wherein peak concentrations for prodrug 2 were observedat approximately 4–6 h, but earlier for ribavirin (3 h). Assumingthat liver radioactivity at 3 and 24 h were similar, the estimated li-ver AUC(0–24 h)’s for prodrug 2 and ribavirin were calculated to be644 and 445 lg equivalent/g�h, respectively, suggesting that 2could achieve substantially higher liver radioactivity levels thanribavirin after a single oral dose. The potential for increased liverexposure suggests that prodrug 2 could be administered at a much

of a new phosphorylated ribavirin prodrug. Antiviral Res. (2013), http://

Page 8: Synthesis and evaluation of a new phosphorylated ribavirin prodrug

393

394

395

396

397

398

399

400

401

402

403

404

405

406

407

408

409

410

411

412

413

414

415

416

417

418

419

420

421

422

423

424

425

426

427

428

429

430

431

432

433

434

435

436

437

438

439

440

441

442

443

444

445

446

447

448

449

450

451

452

453454455456457458459460461462463464465466467468469470471472473474475476477478479480481482483Q4484485486487488489490491492493494495496497498499500501502503504505506507508509510511512513514515516517518519520521522523524525526527528529530531532533534

8 S.D. Dong et al. / Antiviral Research xxx (2013) xxx–xxx

AVR 3191 No. of Pages 9, Model 5G

29 April 2013

lower dose than ribavirin yet still achieve liver levels necessary foractivity while significantly reducing the exposure to RBCs.

4. Discussion

Compound 2 represents a novel approach toward a ribavirinprodrug with potentially superior antiviral activity and better tol-erability characteristics. The in vitro data suggest that the phospho-diester prodrugging approach can successfully deliver andsubsequently release active ribavirin mono-phosphate 1 in nucle-ated cells such as hepatocytes. Consequently, 2 is more active thanribavirin against HCV and influenza virus as well as more effectivein depleting guanine nucleotide pools. In anucleated RBCs, prodrug2 has minimal impact on ATP levels and limited uptake. Con-versely, prodrug 2 has much higher uptake levels in nucleatedHepG2 hepatocytes consistent with its improved activity againstthe HCV replicon. The improved uptake observed of 2 in HepG2cells suggests a possible approach to overcoming the variabilityof ribavirin hepatocytic uptake in the broader patient populationwith chronic HCV infection by avoiding sole reliance on the vari-able uptake capacities of different ENT1 isoforms.

In vivo studies indicate that the prodrug 2 is orally bioavailableand well tolerated. Unlike ribavirin which achieves steady-stateequilibrium plasma levels quickly, uptake of prodrug 2 was slowerin rats but continued accumulating compared to the ribavirin con-trol (Fig. 7b). When dosed at a molar equivalent level to ribavirin,because of its different accumulation profile, prodrug 2 producedplasma AUC(0–24 h) levels of ribavirin over the span of the 28-daystudy that exceeded the levels derived from the ribavirin control.At the same time, tissue distribution studies (Fig. 8) show that pro-drug 2 radioactivity levels in the liver significantly surpass thosegenerated from the ribavirin control when both compounds wereadministered at equal doses. When administered at molar equiva-lent dosages over long periods, prodrug 2 may produce plasmaribavirin levels similar to ribavirin, which could limit some of thesafety potential advantages of the prodrug at this dose. However,because of prodrug 2’s greater antiviral activity and better liverdistribution, these data suggest 2 could potentially be adminis-tered at a fraction of the conventional ribavirin dosage therebyminimizing RBC exposure yet still achieving liver levels of the ac-tive phosphorylated species similar to ribavirin. Additionally, thehigher uptake and accumulation of prodrug 2 suggest that it couldalso be dosed less frequently than ribavirin, which must be admin-istered from 500 to 600 mg twice a day, every day, during treat-ment of hepatitis C.

As new therapeutic modalities for HCV infection arise, there islikely to be continued interest in complementary ribavirin analogsthat have less variability in both efficacy and dosing and can bemore readily combined with other antivirals (Paeshuyse et al.,2011; Vermehren and Sarrazin, 2011). Future work will examine2’s efficacy compared to a ribavirin control in relevant in vivo mod-els such as the murine influenza model, as well as optimizing dos-ing regimens to define further the potentially larger therapeuticwindow between activity and toxicity (Matsuoka et al., 2009).

Acknowledgments

We gratefully acknowledge Drs. Vaishali Chaudhuri and How-ard J. Worman for critically reviewing this manuscript.

Appendix A. Supplementary data

Supplementary data associated with this article can be found, inthe online version, at http://dx.doi.org/10.1016/j.antiviral.2013.04.014.

Please cite this article in press as: Dong, S.D., et al. Synthesis and evaluationdx.doi.org/10.1016/j.antiviral.2013.04.014

References

Bengsch, B., Thimme, R., 2010. Ribavirin ante portas: uptake transporters intohepatocytes dissected. J. Hepatol. 52, 469–471.

Blight, K.J., Kolykhalov, A.A., Rice, C.M., 2000. Efficient initiation of HCV RNAreplication in cell culture. Science 290, 1972–1974.

Crotty, S., Cameron, C., Andino, R., 2002. Ribavirin’s antiviral mechanism of action:lethal mutagenesis? J. Mol. Med 80, 86–95.

Dadgostari, S., Xu, C., Yeh, L.-T., Lin, C.C., Vitarella, D., 2004. Viramidinedemonstrates better safety than ribavirin in monkeys but not rats. Drug.Chem. Toxicol. 27, 191–211.

De Franceschi, L., Fattovich, G., Turrini, F., Ayi, K., Brugnara, C., Manzato, F., Noventa,F., Stanzial, A.M., Solero, P., Corrocher, R., 2000. Hemolytic anemia induced byribavirin therapy in patients with chronic hepatitis C virus infection: role ofmembrane oxidative damage. Hepatology 31, 997–1004.

Di Pierro, D., Tavazzi, B., Perno, C.F., Bartolini, M., Balestra, E., Caliò, R., Giardina, B.,Lazzarino, G., 1995. An ion-pairing high-performance liquid chromatographicmethod for the direct simultaneous determination of nucleotides,deoxynucleotides, nicotinic coenzymes, oxypurines, nucleosides, and bases inperchloric acid cell extracts. Anal. Biochem. 231, 407–412.

Dixit, N.M., Perelson, A.S., 2006. The metabolism, pharmacokinetics andmechanisms of antiviral activity of ribavirin against hepatitis C virus. Cell.Mol. Life Sci. 63, 832–842.

Feld, J.J., 2012. Is there a role for ribavirin in the era of hepatitis C virus direct-actingantivirals? Gastroenterology 142, 1356–1359.

Fletcher, T., Ptak, R., Bartram, S., Halliday, S., Buckheit, R., Moravec, R., Riss, T., 2000.A HTS system for screening antiviral compounds using the cell Titer� 96aqueous one solution assay. Promega Notes 75, 13–16.

Fukuchi, Y., Furihata, T., Hashizume, M., Iikura, M., Chiba, K., 2010. Characterizationof ribavirin uptake systems in human hepatocytes. J. Hepatol. 52, 486–492.

He, G.-X., Krise, J.P., Oliyai, R., 2007. Prodrugs of phosphonates, phosphinates, andphosphates. In: Stella, V.J., Borchardt, R.T., Hageman, M.J., Oliyai, R., Maag, H.,Tilley, J.W. (Eds.), Prodrugs. Springer, New York, NY, pp. 923–964.

Hostetler, K.Y., Beadle, J.R., Hornbuckle, W.E., Bellezza, C.A., Tochkov, I.A., Cote, P.J.,Gerin, J.L., Korba, B.E., Tennant, B.C., 2000. Antiviral activities of oral 1-O-hexadecylpropanediol-3-phosphoacyclovir and acyclovir in woodchucks withchronic woodchuck hepatitis virus infection. Antimicrob. Agents Chemother.44, 1964–1969.

Hostetler, K.Y., Rybak, R.J., Beadle, J.R., Gardner, M.F., Aldern, K.A., Wright, K.N., Kern,E.R., 2001. In vitro and in vivo activity of 1-O-hexadecylpropanediol-3-phospho-ganciclovir and 1-O-hexadecylpropanediol-3-phospho-penciclovir incytomegalovirus and herpes simplex virus infections. Antivir. Chem.Chemother. 12, 61–70.

Hostetler, Karl Y., 2009. Alkoxyalkyl prodrugs of acyclic nucleoside phosphonatesenhance oral antiviral activity and reduce toxicity: current state of the art.Antiviral Res. 82, A84–98.

Hostetler, Karl Y., 2010. Synthesis and early development of hexadecyloxypropyl-cidofovir: an oral antipoxvirus nucleoside phosphonate. Viruses 2, 2213–2225.

Ibarra, K.D., Pfeiffer, J.K., 2009. Reduced ribavirin antiviral efficacy via nucleosidetransporter-mediated drug resistance. J. Virol. 83, 4538–4547.

Iikura, M., Furihata, T., Mizuguchi, M., Nagai, M., Ikeda, M., Kato, N., Tsubota, A.,Chiba, K., 2012. ENT1, a ribavirin transporter, plays a pivotal role in antiviralefficacy of ribavirin in a hepatitis C virus replication cell system. Antimicrob.Agents Chemother. 56, 1407–1413.

Inoue, Y., Homma, M., Matsuzaki, Y., Shibata, M., Matsumura, T., Ito, T., Kohda, Y.,2006. Erythrocyte ribavirin concentration for assessing hemoglobin reductionin interferon and ribavirin combination therapy. Hepatol. Res. 34, 23–27.

Korba, B.E., Gerin, J.L., 1992. Use of a standardized cell culture assay to assessactivities of nucleoside analogs against hepatitis B virus replication. AntiviralRes. 19, 55–70.

Lau, J., Hong, Z., 2004. Methods of drug delivery to hepatocytes and treatment offlaviviridae infections [WWW Document]. <http://patentscope.wipo.int/search/en/WO2002015904> (accessed 5.23.12).

Lin, C., Yeh, L.-T., Vitarella, D., Hong, Z., 2003a. Viramidine, a prodrug of ribavirin,shows better liver-targeting properties and safety profiles than ribavirin inanimals. Antivir. Chem. Chemother. 14, 145–152.

Lin, C.-C., Luu, K., Lourenco, D., Yeh, L.-T., 2003b. Pharmacokinetics and metabolismof [14C]viramidine in rats and cynomolgus monkeys. Antimicrob. AgentsChemother. 47, 2458–2463.

Matsuoka, Y., Lamirande, E.W., Subbarao, K., 2009. The mouse model for influenza.In: Coico, R., Kowalik, T., Quarles, J., Stevenson, B., Taylor, R. (Eds.), CurrentProtocols in Microbiology. John Wiley & Sons, Inc., Hoboken, NJ, USA.

Okuse, C., Rinaudo, J.A., Farrar, K., Wells, F., Korba, Brent.E., 2005. Enhancement ofantiviral activity against hepatitis C virus in vitro by interferon combinationtherapy. Antiviral Res. 65, 23–34.

Paeshuyse, J., Dallmeier, K., Neyts, J., 2011. Ribavirin for the treatment of chronichepatitis C virus infection: a review of the proposed mechanisms of action. Curr.Opin. Virol. 1, 590–598.

Page, T., Connor, J.D., 1990. The metabolism of ribavirin in erythrocytes andnucleated cells. Int. J. Biochem. 22, 379–383.

Parker, W.B., 2005. Metabolism and antiviral activity of ribavirin. Virus Res. 107,165–171.

Pockros, P.J., 2010. New direct-acting antivirals in the development for hepatitis Cvirus infection. Therap. Adv. Gastroenterol. 3, 191–202.

of a new phosphorylated ribavirin prodrug. Antiviral Res. (2013), http://

Page 9: Synthesis and evaluation of a new phosphorylated ribavirin prodrug

535536537538539540541542543544

545546547548549550551552553

554

S.D. Dong et al. / Antiviral Research xxx (2013) xxx–xxx 9

AVR 3191 No. of Pages 9, Model 5G

29 April 2013

Randall, G., Grakoui, A., Rice, Charles M., 2003. Clearance of replicating hepatitis Cvirus replicon RNAs in cell culture by small interfering RNAs. PNAS 100, 235–240.

Russmann, S., Grattagliano, I., Portincasa, P., Palmieri, V.O., Palasciano, G., 2006.Ribavirin-induced anemia: mechanisms, risk factors and related targets forfuture research. Curr. Med. Chem. 13, 3351–3357.

Selle, H., Chapman, B.E., Kuchel, P.W., 1992. Release of choline by phospholipase Dand a related phosphoric diester hydrolase in human erythrocytes. 1H spin-echon.m.r. studies. Biochem. J. 284 (Pt. 1), 61–65.

Shiffman, M.L., 2009. What future for ribavirin? Liver Int. 29 (Suppl. 1), 68–73.

Please cite this article in press as: Dong, S.D., et al. Synthesis and evaluationdx.doi.org/10.1016/j.antiviral.2013.04.014

Sidwell, R.W., Huffman, J.H., Khare, G.P., Allen, L.B., Witkowski, J.T., Robins, R.K.,1972. Broad-spectrum antiviral activity of Virazole: 1-beta-D-ribofuranosyl-1,2,4-triazole-3-carboxamide. Science 177, 705–706.

Vermehren, J., Sarrazin, C., 2011. New HCV therapies on the horizon. Clin. Microbiol.Infect. 17, 122–134.

Yeh, L.-T., Nguyen, M., Lourenco, D., Lin, C.-C., 2005. A sensitive and specific methodfor the determination of total ribavirin in monkey liver by high-performanceliquid chromatography with tandem mass spectrometry. J. Pharm. Biomed.Anal. 38, 34–40.

of a new phosphorylated ribavirin prodrug. Antiviral Res. (2013), http://


Recommended