57
3C protease of enterovirus 68: Structure-based design of Michael acceptor inhibitors and their broad-spectrum antiviral effects against picornaviruses 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 Jinzhi Tan a,b, , Shyla George a,b , Yuri Kusov a,b , Markus Perbandt a,c , Stefan Anemüller a,b , Jeroen R. Mesters a,b , Helene Norder d , Bruno Coutard e , Céline Lacroix f , Pieter Leyssen f , Johan Neyts f , Rolf Hilgenfeld a,b,c,g, # Institute of Biochemistry, Center for Structural and Cell Biology in Medicine, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany a ; German Centre for Infection Research (DZIF), University of Lübeck b ; Laboratory for Structural Biology of Infection and Inflammation, Universities of Lübeck and Hamburg, Building 22a, c/o DESY, Notkestr. 85, 22603 Hamburg, Germany c ; Department of Clinical Microbiology, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden d ; Laboratoire Architecture et Fonction des Macromolécules Biologiques, UMR 6098, Centre National de la Recherche Scientifique and Universités d'Aix-Marseille I et II, Case 925, 163 avenue de Luminy, 13288 Marseille Cedex 9, France e ; Rega Institute for Medical Research, Katholieke Universiteit Leuven, Minderbroedersstraat 10, 3000 Leuven, Belgium f ; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Rd., Shanghai 201203, China g # Corresponding author. Mailing address for Rolf Hilgenfeld: Institute of Biochemistry, Center for Structural and Cell Biology in Medicine, University of Copyright © 2013, American Society for Microbiology. All Rights Reserved. J. Virol. doi:10.1128/JVI.01123-12 JVI Accepts, published online ahead of print on 6 February 2013

JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

3C protease of enterovirus 68: Structure-based design of Michael acceptor

inhibitors and their broad-spectrum antiviral effects against picornaviruses

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

Jinzhi Tana,b,∗, Shyla Georgea,b, Yuri Kusova,b, Markus Perbandta,c, Stefan

Anemüllera,b, Jeroen R. Mestersa,b, Helene Norderd, Bruno Coutarde, Céline

Lacroixf, Pieter Leyssenf, Johan Neytsf, Rolf Hilgenfelda,b,c,g,#

Institute of Biochemistry, Center for Structural and Cell Biology in Medicine,

University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germanya; German

Centre for Infection Research (DZIF), University of Lübeckb; Laboratory for

Structural Biology of Infection and Inflammation, Universities of Lübeck and

Hamburg, Building 22a, c/o DESY, Notkestr. 85, 22603 Hamburg, Germanyc;

Department of Clinical Microbiology, Sahlgrenska Academy, Gothenburg

University, Gothenburg, Swedend; Laboratoire Architecture et Fonction des

Macromolécules Biologiques, UMR 6098, Centre National de la Recherche

Scientifique and Universités d'Aix-Marseille I et II, Case 925, 163 avenue de

Luminy, 13288 Marseille Cedex 9, Francee; Rega Institute for Medical Research,

Katholieke Universiteit Leuven, Minderbroedersstraat 10, 3000 Leuven, Belgiumf;

Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu

Chong Zhi Rd., Shanghai 201203, Chinag

# Corresponding author. Mailing address for Rolf Hilgenfeld: Institute of

Biochemistry, Center for Structural and Cell Biology in Medicine, University of

Copyright © 2013, American Society for Microbiology. All Rights Reserved.J. Virol. doi:10.1128/JVI.01123-12 JVI Accepts, published online ahead of print on 6 February 2013

Page 2: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany. Phone: +49-451-500-

4060, fax: +49-451-500-4068. Email:

24

[email protected] 25

26

27

28

29

30

31

32

33

34

35

36

37

∗ Present address: Shanghai Institute of Materia Medica, Chinese Academy of

Sciences, 555 Zu Chong Zhi Rd., Shanghai 201203, China

Running title: Enterovirus 3Cpro: Broad-spectrum inhibitor design

Word counts:

Title: 18 (150 characters including spaces)

Abstract: 247

Main text: 6661

2

Page 3: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

Abstract 38

39

40

41

42

43

44

45

46

47

48

49

50

51

52

53

54

55

56

57

58

59

60

We have determined the cleavage specificity and the crystal structure of the 3C

protease of enterovirus 68 (EV68 3Cpro). The protease exhibits a typical

chymotrypsin fold with a Cys...His...Glu catalytic triad; its three-dimensional

structure is closely related to that of the 3Cpro of rhinovirus 2 as well as to that of

poliovirus. The phylogenetic position of the EV68 3Cpro between the

corresponding enzymes of rhinoviruses on the one hand and enteroviruses on

the other prompted us to use the crystal structure for the design of irreversible

inhibitors, with the goal of discovering broad-spectrum antiviral compounds. We

synthesized a series of peptidic α,β-unsaturated ethyl esters of increasing length

and for each inhibitor candidate, we determined a crystal structure of its complex

with the EV68 3Cpro, which served as the basis for the next design round. To

exhibit inhibitory activity, compounds must span at least P3 to P1'; the most

potent inhibitors comprise P4 to P1'. Inhibitory activities were found against the

purified 3C protease of EV68 as well as with replicons for poliovirus and EV71

(EC50 = 0.5 μM for the best compound). Antiviral activities were determined using

cell cultures infected with EV71, poliovirus, echovirus 11, and various rhinovirus

serotypes. The most potent inhibitor, SG85, exhibited activity with EC50 values of

≈180 nM against EV71 and ≈60 nM against human rhinovirus 14 in a live virus-

cell-based assay. Even the shorter SG75, spanning only P3 to P1', displayed

significant activity (EC50 = 2 to 5 μM) against various rhinoviruses.

3

Page 4: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

Introduction 61

62

63

64

65

66

67

68

69

70

71

72

73

74

75

76

77

78

79

80

81

82

83

Enteroviruses comprise several pathogens that are implicated in a large variety

of clinical manifestations ranging from mild illnesses to more serious or even life-

threatening diseases, such as meningitis, encephalitis, myocarditis, pancreatitis,

acute paralysis, or neonatal sepsis (30, 32). In recent years, China and several

countries in South East Asia have been hit by outbreaks of Hand-, Foot- and

Mouth Disease (HFMD) caused by enterovirus (EV) 71 or Coxsackievirus A16

(more than 488,000 cases in the 2008 epidemic in China alone (42)).

To date, no approved specific antiviral therapy for diseases caused by

enteroviruses is available. There is an urgent need for safe and broad-spectrum

drugs against the existing pathogenic EVs. The same holds true for other

members of the picornavirus family, in particular rhinoviruses, as it is now clear

that some of the latter cause exacerbations of asthma and COPD (30). Also,

drugs against poliovirus are needed to aid in the completion of polio eradication

(9).

The enteroviral genome consists of a single-stranded, positive-sense RNA of

approximately 7,500 bases in length. The coding region of the viral genome is

divided into three primary precursor molecules (P1, P2, P3) containing the four

structural (derived from P1) and 7 nonstructural viral proteins (derived from P2

and P3). The genome gives rise to the viral polyprotein, which is processed co-

and post-translationally through a series of primary and secondary proteolytic

4

Page 5: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

cleavages by the virus-encoded proteases 2Apro and 3Cpro/3CDpro. 2Apro cleaves

the bond between the P1 and P2 segments of the viral polyprotein, whereas the

3Cpro and its precursor, 3CDpro, are responsible for generating the majority of

precursor and mature proteins (see, e.g., (30)).

84

85

86

87

88

89

90

91

92

93

94

95

96

97

98

99

100

101

102

103

104

105

106

Here, we describe the substrate cleavage specificity and the crystal structure (at

2.4 Å resolution) of the 3C protease of enterovirus 68 (EV68, also called EV-D68

to indicate its membership in the human enterovirus D family). This is the first

crystal structure of a protein from a group-D enterovirus. Like the other members

of the enterovirus family, the EV68 3Cpro is a cysteine protease containing a

Cys...His...Glu catalytic triad and exhibiting a two-domain fold similar to that of

the serine proteases of the chymotrypsin family. A structural comparison of the

3C proteases of known crystal structure revealed an intermediate position of the

EV68 3Cpro between the corresponding enzymes from human rhinovirus 2 (HRV2)

and those from enteroviruses, and prompted us to use the structure for the

design of broad-spectrum antivirals directed against picornaviruses in general,

even though EV68 itself does not play any important role as a pathogen.

Because of their importance in the viral replication cycle and their unique

specificity for glutamine in the P1 position of the substrate (which is not found in

any known host-cell protease), 3C proteases are attractive targets for antiviral

drug discovery (16, 35). However, only one such drug candidate entered clinical

trials so far. Rupintrivir (AG7088) was developed as an inhibitor of the 3Cpro of

human rhinoviruses, but development was stopped after phase II because of

5

Page 6: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

107

108

109

110

111

112

113

114

115

116

117

118

119

120

121

122

123

124

125

126

127

128

129

limited efficacy in the treatment of the common cold (15). Carrying an α,β-

unsaturated alkyl ester moiety, rupintrivir acts as a Michael acceptor for the

nucleophilic Cys residue in the catalytic center of the 3Cpro (25). A series of

studies found α,β-unsaturated alkyl esters to be the most active rhinovirus 3C

protease inhibitors among various electrophilic compounds (11). We and others

have occasionally described α,β-unsaturated esters as inhibitors of coronavirus

or enterovirus proteases (1, 18, 20, 21, 43), but the only systematic study

involving X-ray crystallography and structure-based drug design as well as in-

vivo evaluation deals with the coronavirus main protease as a target (43).

We synthesized a series of peptides of increasing lengths that carry an α,β-

unsaturated ester as a Michael acceptor for the nucleophilic cysteine residue of

the enzyme. A crystal structure of each compound in complex with the EV68

3Cpro revealed details of the interactions occurring in the specificity subsites of

the enzyme and suggested modifications to be applied in the next round of the

design process. We found that the potency of the compounds increased with

length. SG85, which occupied 3Cpro subsites S4 to S1', was the most potent

among these inhibitors, with kobs/[I] = 202200 M-1s-1. Most members of this series

of compounds were not toxic to Vero A, Hela Rh, BGM, RD, and Huh-T7 cells.

They proved also active on the replication of subgenomic replicons and in cells

infected with other picornaviruses such as enterovirus 71 (also called EV-A71 to

indicate its classification as a human enterovirus A (HEV-A) species), poliovirus,

echovirus 11, and rhinoviruses. Thus, our structure-based drug design approach

6

Page 7: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

130

131

132

133

134

135

136

137

138

139

140

141

142

143

144

145

146

147

148

149

150

151

152

underlines the suitability of the picornaviral 3C protease as a target for broad-

spectrum antivirals.

MATERIALS AND METHODS

Synthesis of α,β-unsaturated ester compounds (see Fig. 1A, B, and

Supplemental Material): SG74 was synthesized using a published procedure

(28). To a solution of SG74 (for SG75 and SG81) or SG81 (for SG82, SG83,

SG84, SG85, and SG98) (1 mmol, 1 equiv.) in 1 ml of CH2Cl2, 0.5 ml of TFA was

added at 0°C. The dark brown solution was stirred at room temperature for 2

hours and then concentrated in vacuo. 0.5 ml of diisopropylethylamine was

added to neutralize the residual in situ and the resulting solution was used for the

next step without further purification. Protected amino acid (1 mmol, 1 equiv.),

EDC·HCl (1.1 mmol, 1.1 equiv.), and HOBt (1.1 mmol, 1.1 equiv.) were added to

a solution of the above amine in 5 ml of DMF at 0°C. The resulting solution was

stirred at room temperature for 18 h. Then the reaction mixture was diluted with

ethyl acetate and the organic phase was washed with 10% citric acid, saturated

aqueous NaHCO3 and brine, dried over MgSO4, filtered and concentrated in

vacuo. The resulting white solid was further purified through silica gel

chromatography.

Recombinant protein production. cDNA corresponding to the 3Cpro domain of

EV68 (strain 3799) was amplified prior to cloning into the pOPINE plasmid in

order to produce the EV68 3Cpro in fusion with a lysine residue and a (His)6-tag at

7

Page 8: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

153

154

155

156

157

158

159

160

161

162

163

164

165

166

167

168

169

170

171

172

173

174

175

176

the C-terminus (4). The coding region was verified by DNA sequencing and the

construct was transformed into Escherichia coli Tuner (DE3) plac I cells for

expression.

The protein was purified using immobilized-metal-affinity chromatography

(Histrap FF column, GE Healthcare), and eluted with 25 mM Tris-HCl, pH 8.0,

300 mM NaCl, and 250 mM imidazole. Further purification was achieved by size-

exclusion chromatography using 25 mM Tris-HCl, pH 8.0, 200 mM NaCl, 5 mM

DTT, and a HiLoad 16/60 Superdex 75 prep grade column. Protein concentration

was determined using the Bio-Rad Protein Assay, and the sample was stored at

-70°C.

Crystallization and data collection. The protein was concentrated to 10 mg/ml

for crystallization. Initial crystallization screening was performed using the sitting-

drop vapor-diffusion method in 96-well Intelli-Plates (Dunn Laboratories). Several

commercial kits (Sigma, Jena Bioscience, and Hampton Research) were used for

screening. Using a Phoenix robotic system (Art Robbins), drops were made of

260 nl protein and 260 nl precipitant solution. The optimized crystallization

condition was identified as 0.07 M sodium acetate (pH 4.6), 5 – 15% PEG 4000,

and 10 – 30% glycerol.

The peptidic Michael acceptor compounds (SG74, SG75, SG81, SG82, SG83,

SG84, SG85, and SG98) were dissolved in DMSO. Aliquots of EV68 3C protease

in 25 mM Tris-HCl, pH 8.0, 200 mM NaCl, 5 mM DTT were incubated with

8

Page 9: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

177

178

179

180

181

182

183

184

185

186

187

188

189

190

191

192

193

194

195

196

197

198

199

200

inhibitor solution at a molar ratio of 1:5 for 30 min at room temperature, and then

subjected to co-crystallization using the sitting-drop vapor diffusion method under

the same crystallization conditions as used for the free enzyme.

No cryoprotectant was added for data collection, since the reservoir solutions

contained sufficient amounts of glycerol. All diffraction data were collected at 100

K from a single crystal for each inhibitor complex at beamline BL14.1, BESSY

(Berlin, Germany), using an MX225 CCD detector (Rayonics). Data were

processed with MOSFLM (22), and reduced and scaled using the SCALA (13)

program from the CCP4 suite (8).

Structure determination. The crystal structure of the EV68 3C protease was

determined by molecular replacement (33) using MOLREP (37) and the atomic

coordinates of Human Rhinovirus-2 3Cpro (25) as the search model. The initial

model was built using Coot (12), and the program Refmac5 (8, 29) was used for

model refinement. The complex structures were determined by molecular

replacement using the structure of the free enzyme and refined as mentioned

above, to resolutions between 1.80 and 2.65 Å. Refinement statistics are

summarized in Table 1.

The stereochemistry of the structures was assessed using PROCHECK (19). The

quality of the structure-factor data and their agreement with the atomic model

was evaluated with the program SFCHECK (38). The atomic coordinates and

structure factors for all structures have been deposited with the RCSB Protein

9

Page 10: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

201

202

203

204

205

206

207

208

209

210

211

212

213

214

215

216

217

218

219

220

221

222

Data Bank; the PDB accession codes are listed in Table 1. All figures were

generated using PyMOL (Schroedinger).

Determination of substrate specificity and enzyme kinetics. Eight

dodecapeptide substrates (see Fig. 2) for the HPLC-based cleavage assay were

purchased from GL Biochem (Shanghai) Ltd. These substrates represent the

eight putative cleavage sites of the 3Cpro in the EV68 polyprotein. At their N- and

C-termini, the peptides carried tryptophan residues for easy detection by UV

spectroscopy, as described earlier for peptide substrates of the SARS-

coronavirus main protease (36). To determine the substrate specificity of the

protease, 50 μM of each peptide (dissolved in water) was incubated with 1 μM

protease in buffer A containing 20 mM Tris (pH 7.3), 100 mM NaCl, 1 mM EDTA

at room temperature for 24 hours. Reactions were terminated by adding 0.1%

trifluoroacetic acid, and the samples were analyzed using a reverse-phase HPLC

C12 column (Jupiter 4u Proteo 90Å; Phenomenex) with detection at 280 nm. The

peak areas of the products were integrated to calculate the reaction rate of each

substrate under the catalysis of the enzyme. The optimum substrate with the

fastest reaction rate was selected for determination of the enzyme kinetics using

fluorescence spectrophotometry. The FRET peptide used as the substrate,

Dabcyl-KEALFQ↓GPPQFE-Edans amide (95% purity, Biosyntan), contained the

2C/3A cleavage site (indicated by the arrow).

10

Page 11: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

223

224

225

226

227

228

229

230

231

232

233

234

235

236

237

238

239

240

241

242

243

244

245

Kinetic measurements were performed in the same buffer as for the HPLC test.

The enhanced fluorescence due to the cleavage of this substrate as catalyzed by

the enzyme was monitored at 490 nm with excitation at 340 nm, using a Cary

Eclipse fluorescence spectrophotometer. The experiments were performed in a

fluorescence cuvette (volume: 1 ml) with a light-path of 1 cm for excitation and a

light-path of about 2 mm for emission, at a 90-degree angle. Slit widths were 5

nm in both cases. The photomultiplier voltage was 600 V, and the assay volume

was 400 µl. The enzyme concentration for measuring Km and kcat was 0.2 μM,

and the concentrations of the FRET peptide were varied from 2 to 20 μM. The

initial rate, within 10% of the substrate consumption, was used to calculate the

kinetic parameters using Michaelis-Menten equation fitting with the Origin

program (OriginLab). Owing to solubility limitations, the FRET substrate could not

be used at concentrations higher than 20 μM; the Km value was therefore

estimated by non-linear regression analysis according to ref. 41. By using only

relatively low substrate concentrations, we also avoided potential inner-filter

effects caused by the substrate.

Enzyme inhibition assay. The FRET-based assay was also used for the

determination of the inhibition constants of the α,β-unsaturated esters. At the

outset, we experimentally characterized the inhibitors with respect to possible

inner-filter effects. The absorption spectra of all of them were measured at a

concentration of 100 µM in DMSO in the range from 260 to 600 nm. Only

compound SG84 displayed a significant absorption above 260 nm, and up to 310

11

Page 12: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

246

247

248

249

250

251

252

253

254

255

256

257

258

259

260

261

262

263

264

nm. However, even from the latter compound, no absorption was registered

around 340 nm, the excitation wavelength used in the fluorimetric assay. Also, at

the emission wavelength used for the fluorimetric assay (i.e., 490 nm), none of

the compounds showed significant absorption. Finally, fluorescence emission

spectra were also recorded for all inhibitors, using the same excitation

wavelength (340 nm) as for the fluorimetric assay. None of the compounds

displayed a significant fluorescence signal around 490 nm, the emission

wavelength used in the enzymatic test system. Thus, potential inner-filter effects

did not have to be taken into account in our experiments.

The principle of the assays and more details have been described elsewhere (39,

41). Briefly, time-dependent progress curves were fitted to a first-order

exponential (equation 1) to obtain an observed first-order inhibition rate constant

(kobs) (26). F is the product fluorescence (measured in arbitrary units), v0 is the

initial velocity, t is time, and D is a displacement term accounting for the non-zero

emission at the start of data collection. Since in the case of fast inactivation, the

measurement of Ki and k3 tends to be difficult, kobs/[I] was used as an

approximation of the pseudo-second-order rate constant, in order to evaluate all

inhibitors for easy comparison.

( ) DkvF tk

obs

obs +−⋅⎟⎟⎠

⎞⎜⎜⎝

⎛= ⋅−exp10

(1) 265

266

267

Reactions were performed with 0.2 μM protease and 20 μM FRET peptide in

buffer A (20 mM Tris (pH 7.3), 100 mM NaCl, 1 mM EDTA) by addition of the

12

Page 13: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

268

269

270

271

272

273

274

275

276

277

278

279

280

281

282

283

284

285

286

287

288

289

290

enzyme to the reaction buffer containing substrate and inhibitor. Three to five

different inhibitor concentrations were tested, varied over a range from 0.1 μM to

100 μM. Data from the continuous assays were analyzed with the non-linear

regression analysis program Origin (OriginLab) to obtain kobs for enzyme

inactivation at each inhibitor concentration. The slope of a graph of kobs vs [I] was

calculated using Origin and is reported as kobs/[I] in Table 2.

Cells and viruses. A derivative of human hepatocellular carcinoma cells,

constitutively expressing a T7 RNA polymerase (Huh-T7) (34), was grown in

Dulbecco’s modified minimal essential medium (DMEM) supplemented with 2

mM glutamine, 100 units/ml penicillin, 100 μg/ml streptomycin sulphate, 400

μg/ml geneticin (G-418 sulphate), and fetal calf serum (10% in growth medium

and 2% in maintenance medium). Other cells used were Vero A cells, a

derivative of Vero cells (African green monkey kidney cells), BGM (buffalo green

monkey kidney cells), and RD (rhabdomyosarcoma) cells, which were grown in

MEM Rega3 (Invitrogen) supplemented with 10% FCS for regular subculturing

and 2% FCS for antiviral assays (Integro), 5 ml of 200 mM L-glutamine, and 5 ml

of 7.5% sodium bicarbonate. Hela Rh cells, a HeLa cell clone selected for

susceptibility to HRV replication, were grown in MEM (41965-039, 500 ml per

bottle, Invitrogen) supplemented with 10% FCS for regular subculturing and 2%

FCS for antiviral assays (Integro), 5 ml of 200 mM L-glutamine, and 10 ml of 1 M

HEPES (15630-056, Invitrogen).

13

Page 14: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

291

292

293

294

295

296

297

298

299

300

301

302

303

304

305

306

307

308

309

310

311

312

313

Echovirus 11 (ECHO11; strain Gregory) and rhinoviruses (HRV types 2, 14, 29,

70, and 95) were a kind gift of K. Andries (Johnson & Johnson). Enterovirus 71

(EV71; strain BrCr) was provided by F. van Kuppeveld (Nijmegen, The

Netherlands). Poliovirus 1 (Sabin) was obtained from the NIBSC, Hertfordshire

(UK) (J. Martin).

Viral replicons and transfection. Subgenomic replicon DNAs of PV (pRLuc31,

(2)) and HAV (p18f-Luc, (14)) encoded a full-length copy of poliovirus (strain

Mahoney) and Hepatitis A Virus (18f strain), respectively, in which the P1 capsid-

coding sequence was replaced by the firefly luciferase gene. The whole

sequence was cloned into the pGEM1 or pGEM2 vector (Promega) behind a T7

RNA polymerase promoter. The EV71 replicon DNA (pACYC-RLuc), kindly

provided by Bo Zhang, contained the EV71 sequence placed into the pACYC/177

vector (New England Biolabs), under control of the T7 promoter. In this construct,

the P1 region was replaced by the Renilla luciferase gene. Replication-deficient

replicons containing a deletion or mutation within the 3D polymerase gene were

also used as a control. To prepare replicon RNA transcripts, replicon DNAs of PV

(pRluc31), HAV (p18f-Luc), and EV71 (pACYC-EV71-RLuc) were completely

linearized by digestion with MluI, BshTI (MBI, Fermentas), and HindIII (New

England Biolabs), respectively. Copy RNA transcripts were synthesized in vitro

using linearized DNA templates, T7 RNA polymerase, and the T7 RiboMax™

Large-Scale RNA Production System (Promega) according to the manufacturer’s

recommendations. Huh-T7 cell monolayers grown in 12-well plates to a

14

Page 15: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

314

315

316

317

318

319

320

321

322

323

324

325

326

327

328

329

330

331

332

333

334

335

336

confluency of 80 – 90% were washed with 1 ml OptiMEM (Invitrogen) and

transfected with 0.5 μg of the replication-competent PV, EV71, and HAV replicon

DNA pRLuc31, pACYC-EV71-RLuc, and p18f-Luc, as well as 4.5 μl

Lipofectamin2000 (Invitrogen) in 300 μl OptiMEM (final volume). To transfect

replicon RNAs, RLuc31, EV71-RLuc, and 18f-Luc, the DMRIE-C reagent (3 μl/μg

of purified RNA) was used as recommended (Invitrogen). After 4 to 5 hours of

incubation at 37°C, the transfection mixtures were replaced with growth medium

containing different concentrations of the candidate inhibitor to be tested (40 μM

in preliminary screening experiments or increasing concentrations, from 0 μM to

40 μM, when studying the concentration-dependence). After 24 hours of

incubation, the cells were washed with 1 ml phosphate-buffered saline (PBS) and

lysed in 0.15 ml Passive lysis buffer (Promega) at room temperature (RT) for 10

min. After freezing (-80°C) and thawing (RT), the cell debris was removed by

centrifugation (13,000 rpm, 1 min) and the supernatant (10 μl or 20 μl) was

assayed for firefly or Renilla luciferase activity (Promega) using an Anthos Lucy-3

luminescence plate reader (Anthos Labtec instruments). All experiments were

carried out in triplicate or quadruplicate and the results are presented as mean

values with standard deviations (SD).

Cell toxicity. The CellTiter 96Aqueous One Solution Cell Proliferation Assay

(MTS test, Promega) was used to determine the cytotoxic effect of inhibitors. In

brief, the confluent monolayer of cells was incubated overnight under the

pressure of different concentrations of the tested inhibitor in a total volume of 100

15

Page 16: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

337

338

339

340

341

342

343

344

345

346

347

348

349

350

351

352

353

354

355

356

357

358

359

μl/well. 100% of cell lysis was achieved by incubation with 0.1% Triton-X100

(positive control). After incubation, 50 μl of medium was discarded and 10 μl of

freshly prepared single-assay reagent was added to the cells. The color reaction

was measured at 490 nm using the Anthos Lucy-3 luminescence plate reader

(see above) after 1 or 2 hrs of cell incubation at 37°C. In addition, to detect subtle

differences in cytotoxicity of the tested compounds towards Huh-T7 cells,

analysis of cell death was performed using the ToxiLight Non-destructive

Cytotoxicity BioAssay kit as recommended by the manufacturer (Lonza

Rockland).

Antiviral assays in virus-infected cells. Compound dilutions (dilution factor of

5, 4-point dose-response curve for initial screening; dilution factor of 2, 8-point

dose-response curve for further refinement) were prepared in the respective

assay medium (see above) and added to empty wells of a 96-well assay plate by

a liquid-handling robot (EVO200, Tecan). Subsequently, 50 µl of a 4x virus

dilution in assay medium (supplemented with 15 ml of 1.0 M MgCl2 (Sigma) in

case of HRV) was added, immediately followed by addition of 50 µl of the

respective cell suspension in assay medium, yielding a total assay volume of 200

µl. The assay plates were returned to the incubator (35°C for HRV, 37°C for the

other viruses) for 3 – 4 days, the time at which a maximal cytopathic effect is

observed.

16

Page 17: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

360

361

362

363

364

365

366

367

368

369

370

371

372

373

374

375

376

377

378

379

380

381

382

For the evaluation of the antiviral effect, the assay medium was aspirated by a

plate washer (Hydroflex, Tecan), replaced with 75 μl of a 5% MTS (Promega)

solution in phenolsulfonphthalein-free medium (EVO200, Tecan) and incubated

for 1.5 hours (37°C, 5% CO2, 95-99% relative humidity). Absorbance (OD cell

control condition varying between an OD of 0.6-0.8) was measured at a

wavelength of 498 nm (Safire², Tecan) and OD values were converted to

percentage of untreated controls and compiled into antiviral dose-response

curves.

Analysis of the raw data and calculation, if possible, of the EC50 and CC50 values

were performed employing a custom-made data processing software package

(Accelrys). The EC50 (value derived from the dose-response curve) represents

the concentration at which 50% inhibition of viral replication would be observed.

The CC50 (value derived from the dose-response curve) represents the

concentration at which the metabolic activity of the cells is reduced to 50% of the

metabolic activity of untreated cells. All assay wells yielding a percentage of

inhibition larger than 50 were inspected microscopically and scored for inhibition

of virus-induced cytopathic effects as well as evaluated for minor adverse effects

on the host cells.

RESULTS

17

Page 18: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

Substrate specificity and kinetics of EV68 3Cpro 383

384

385

386

387

388

389

390

391

392

393

394

395

396

397

398

399

400

401

402

403

404

405

The substrate peptides used in this study corresponded to the presumable

processing sites of EV68 3Cpro within the viral polyprotein but carried N-terminal

and C-terminal tryptophan residues for easy UV-spectroscopic detection of

cleavage products at 280 nm. The individual peptides used were

WHAITQ↓GVPTYW (VP2↓VP3), WPDIAQ↓LDHLDW (VP3↓VP1),

WDAMEQ↓GITDYW (2A↓2B), WYVPRQ↓SESWLW (2B↓2C),

WEALFQ↓GPPQFW (2C↓3A), WFAGIQ↓GPYTGW (3A↓3B),

WTAKVQ↓GPGFDW (3B↓3C), and WFTDTQ↓GEIVSW (3C↓3D). All these

peptides, with the exception of VP3↓VP1, contain a Gln followed by a small

amino-acid residue (Gly or Ser) framing the cleavage site. Overall, Gly in the P1’

site leads to higher cleavage activities than Ser or Leu (Fig. 2). We observed the

highest cleavage rates for 3B↓3C and 2C↓3A; the latter was chosen for kinetic

studies. Consequently, the peptide substrate used for the FRET assay was

Dabcyl-KEALFQ↓GPPQFE-Edans amide. As Lee et al. (20) reported that

Coxsackievirus B3 (CVB3) 3Cpro exhibits higher activity with the SARS-CoV Mpro

substrate (SAVLQ↓SGFRK) than with the best CVB3 3Cpro substrate peptide, we

also tested this substrate against EV68 3Cpro, but could not detect any activity.

The kcat and Km values of the protease using the substrate corresponding to the

2C↓3A cleavage site were determined as 0.54 s-1 and 22.4 μM, respectively (not

shown).

Crystal structures of EV68 3Cpro and its complexes

18

Page 19: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

EV68 3Cpro was cloned into the pOPINE vector, which codes for an extra Lys

residue and a (His)6-tag at the C-terminus. We decided to have these extra

residues at the C-terminus of the enzyme, as in case of the picornaviral 3C

proteases, the C-terminus is oriented away from the globular protein and

additional residues are unlikely to influence the structure or catalytic activity. This

is supported by Kuo et al. (18), who showed that C-terminally tagged EV71 3Cpro

has the same catalytic activity as the tag-free enzyme. The expression yield was

about 5.0 mg of soluble protein per liter of medium. The protein was concentrated

to 10.0 mg/ml for crystallization. Crystals grew from 0.07 M sodium acetate (pH

4.6), 5 – 15% PEG 4000, and 10 – 30% glycerol. For both the free enzyme and

its inhibitor complexes, these crystals belonged to space group P3121, with one

3Cpro monomer per asymmetric unit (Table 1). In solution, EV68 3Cpro is a

monomer as well, as indicated by gel filtration and Dynamic Light-Scattering (not

shown). The crystal structure of the free enzyme was determined by molecular

replacement using the structure of rhinovirus 3C protease (25) as a search

model, and refined to 2.4 Å resolution. All amino-acid residues were defined by

electron density, including four histidines of the C-terminal His6-tag. Alternative

conformations were detected in the electron density for the side-chains of active-

site His40 and for Cys60. The final R-factor for this crystal structure was 20.7%

(Rfree = 23.8%). The complex structures were determined based on the crystal

structure of the free enzyme, at resolutions between 1.80 and 2.65 Å. The

inhibitors could all be easily modeled according to the Fo-Fc difference density.

More details can be found in Table 1.

406

407

408

409

410

411

412

413

414

415

416

417

418

419

420

421

422

423

424

425

426

427

428

19

Page 20: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

429

430

431

432

433

434

435

436

437

438

439

440

441

442

443

444

445

446

447

448

449

450

451

Structure of EV68 3Cpro

EV68 3Cpro adopts a chymotrypsin-like fold consisting of two β-barrel domains.

These two domains comprise residues 15 – 77 and 99 – 173, respectively, and

pack against each other to form a shallow substrate-binding cleft (Fig. 3A) that

harbors the catalytic triad of Cys147, His40, and Glu71. There are two

conformations for the side-chain of His40. One conformation, with side-chain

torsion angles χ1 = 69.0o ((+)-synclinal = +sc) and χ2 = -98.7o (anticlinal = ac),

corresponds to the catalytically competent form, in which the sulfur atom of

Cys147 is in plane with the imidazole of His40 and the carboxylate group of

Glu71. In this conformation, the distance between Cys147 Sγ and His40 Nε2 is

3.3 Å and the one between His40 Nδ1 and the carboxylate of Glu71 is 2.7 Å. In

the other conformation, with side-chain torsion angles χ1 = 179.7o (antiperiplanar

= ap) and χ2 = 86.6o (+sc), the imidazole of His40 is exposed to the solvent, with

a distance of 6.8 Å between Cys147 Sγ and His40 Nε2. A likely reason for the

occurrence of this second conformation is the low pH (4.6) of crystallization, at

which part of the His40 side-chains may be protonated, unable to interact with

the sulfhydryl of Cys147. Unfortunately, crystallization trials at higher pH

invariably led to precipitation of the protein and equilibrating low-pH crystals in

buffer of higher pH resulted in destruction of the crystals.

Substrate hydrolysis by cysteine and serine proteases occurs through a covalent

tetrahedral intermediate resulting from attack of the active-site nucleophile onto

20

Page 21: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

the carbonyl carbon of the scissile bond. The developing oxyanion is stabilized by

hydrogen bonds donated by amide groups of the enzyme. In EV68 3Cpro, the

oxyanion hole is formed by the main-chain amides of Gly145 and the active-site

Cys147. In the structure of the free enzyme, the oxyanion hole is occupied by a

water molecule that interacts with the first of these amides.

452

453

454

455

456

457

458

459

460

461

462

463

464

465

466

467

468

469

470

471

472

473

474

Comparison of EV68 3Cpro and related picornaviral proteases

A sequence alignment (Fig. 3B) shows that the identity between the EV68 3Cpro

and the corresponding enzymes of HRV2, EV71, CVB3, EV93, and PV is 49%,

53%, 67%, 67%, and 66%, respectively, whereas the 3C proteases of HAV and

FMDV are more distantly related (sequence identities of 25 - 26%). In terms of

three-dimensional structure, the closest relatives of the EV68 3Cpro are the

enzymes of HRV2 ((25); r.m.s.d. 0.58 Å for 170 superimposed Cα atoms) and PV

(27), a member of subgroup C of human enteroviruses (HEV-C; r.m.s.d. 0.64 Å

for 173 Cα atoms). In contrast to the amino-acid sequence comparisons (see

above), the superimpositions of the three-dimensional 3Cpro structures reflect the

classification into different subgroups of enteroviruses. Thus, the enzymes of

HEV-B members CVB3 (20) (Tan et al., in preparation) and EV93 (10) exhibit

r.m.s. distances from EV68 3Cpro of 0.78 Å (for 173 Cα atoms) and 0.81 Å (for

159 Cα atoms), respectively. The 3Cpro of EV71 (23), a member of the HEV-A

subgroup, exhibits an r.m.s.d. of 0.74 Å for 174 Cα atoms. The relatively large

distance between the enteroviruses on the one hand and Hepatitis A Virus (HAV)

as well as Foot-and-Mouth Disease Virus (FMDV) on the other is reflected in high

21

Page 22: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

r.m.s. distances between the EV68 3Cpro and the corresponding proteases of

these viruses (3,5), with r.m.s.d. values of 2.04 Å for 166 Cα atoms, and 1.93 Å

for 170 Cα atoms, respectively. The intermediate position of the EV68 3Cpro

between the proteases of human rhinoviruses and those of other enteroviruses

suggested that compounds optimized for inhibition of the EV68 enzyme might

exhibit activities against a wide spectrum of picornaviruses.

475

476

477

478

479

480

481

482

483

484

485

486

487

488

489

490

491

492

493

494

495

496

Peptidic α,β-unsaturated esters: design, crystal structures of complexes

with the target enzyme, and inhibitory activities

General strategy

It was reported that several α,β-unsaturated esters acted as Michael acceptors

with excellent inhibitory activity against HRV 3C protease. The compounds

exhibited encouraging antiviral activity, stability in the presence of non-enzymatic

thiols, low cellular toxicity, and were relatively easy to synthesize (25). Based on

the substrate sequence (2C/3A site) and the three-dimensional structure of the

EV68 3Cpro (see above), we have designed and synthesized a series of α,β-

unsaturated ethyl ester compounds of increasing length directed at the enzyme

Fig. 1A and B). Immediately after synthesis, new inhibitor candidates were

cocrystallized with the target protease and had the X-ray structures of their

complexes determined, in order to visualize details of ligand binding as a basis

for the subsequent round of inhibitor design.

22

Page 23: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

497

498

499

500

501

502

503

504

505

506

507

508

509

510

511

512

513

514

515

516

517

518

519

Using the above-mentioned FRET-peptide substrate for the kinetic assay, the

inhibitory activities of these compounds have been determined. Being Michael

acceptors, these inhibitors initially form a reversible encounter complex with the

3Cpro, and then undergo a chemical step (nucleophilic attack by Cys147 leading

to irreversible covalent-bond formation). The observed second-order rate

constant for inactivation (kobs/[I]) depends on both the equilibrium binding

constant, Ki = k2/k1, and the chemical rate for covalent bond formation, k3. For fast

irreversible protease inhibitors, Ki tends to be very difficult to obtain; therefore,

usage of kobs/[I] is more suitable (25, 26). In this case, a higher value of kobs/[I]

indicates that the inhibitor is more active.

P1 position

The P1 residue was not altered in our series of inhibitor candidates. A five-

membered lactam was chosen as a surrogate for the glutamine side-chain amide

in this position, as the relatively rigid lactam side chain will likely lead to a

reduced loss of conformational entropy upon binding to the S1 pocket, compared

to the more flexible Gln, and therefore might lead to tighter binding of the inhibitor

to the 3C protease. This can cause an increase of kobs/[I] by a factor of 10,

compared to the Gln-containing compound (25). In fact, in all of our inhibitor

complex structures, the lactam occupies the S1 subsite (Fig. 4), which involves

residues Thr142, Arg143mc (“mc” stands for main chain), Ala144, Cys147,

His161, and Gly163. At the bottom of the site, the Nε2 atom of His161 donates a

hydrogen bond to the oxygen atom of the lactam; the length of this hydrogen

23

Page 24: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

520

521

522

523

524

525

526

527

528

529

530

531

532

533

534

535

536

537

538

539

540

541

542

bond among all of the complexes is 2.8 Å, indicating that this is a very stable and

conserved interaction.

P2 position

The first compound to be synthesized, SG74 (Boc-GlnLactam-CH=CH-COOEt),

just spans positions P2 – P1'. Its P2-Boc group partly fills the S2 pocket. This

compound does not show any inhibition of the EV68 3Cpro (Table 2). However, it

still binds to the enzyme and features clear electron density in the crystal

structure (Fig. 4A), an interesting demonstration of the sensitivity of X-ray

crystallography.

On the basis of the structure with SG74, we decided that the S2 subsite should

be occupied by a larger hydrophobic residue, and kept Phe invariant in the P2

position in all subsequent inhibitor candidates. The S2 subsite is formed by the

catalytic His40…Glu71 pair on one side, and Leu127 and Gly128 on the other,

and closed off by Thr130 and Asn69 "at the top" (referring to the orientation

shown in Fig. 4H). The pocket is hydrophobic in nature, explaining the preference

of the EV68 3C protease for hydrophobic residues such as Phe, Leu, or Val in

position P2 of the substrates. Surprisingly, the side-chain of His40 has moved out

of the catalytic site in all of the complexes (Fig. 4). As mentioned previously, we

detected two alternative conformations for this side-chain in the free enzyme, one

(+sc, ap) in hydrogen-bonding contact to the catalytic Cys147, and the other (ap,

+sc) exposed to the solvent. However, with χ1 = -80.3 (-sc), χ2 = 59.0 (+sc), the

24

Page 25: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

543

544

545

546

547

548

549

550

551

552

553

554

555

556

557

558

559

560

561

562

563

564

565

conformation of His40 in the complexes corresponds to a third, totally different

orientation. This conformation is stabilized by an ion-pair interaction (but not a

hydrogen bond) between His40 and the carboxylate group of Glu71. However,

while Glu71 is held roughly in the same position as in the free enzyme through a

hydrogen-bond from Asn69, the mutual orientation of Glu71 and His40 is different

from the arrangement in the catalytically active triad and the distance between

the Sγ atom of Cys147 and the Nε2 atom of His40 is about 10.8 Å in all of the

complex structures (see Fig. 4). In other complex crystal structures of

picornavirus 3C proteases, such as that of rhinovirus (25), no such

conformational change was reported. In the EV68 3Cpro complexes with P2-Phe,

the unusual conformation of His40 appears to be stabilized by a C-H...π

hydrogen bond (7, 40) between the Cβ of His40 and the P2 phenyl ring of the

inhibitors. This interaction may also contribute to an unusual orientation of the

P2-Phe residue, which does not really fill the S2 pocket (see Fig. 4H), but moves

towards S1', in concert with the unusual orientation of His40. In order to fill the S2

pocket, the χ1 torsion angle of the phenylalanine side chain should be in the -sc

range, but in fact, it is ap in the structures of the complexes described here.

P3 position

The first inhibitors to contain P2 = Phe, SG75 (Cbz-Phe-GlnLactam-CH=CH-

COOEt) and SG81 (Boc-Phe-GlnLactam-CH=CH-COOEt), span positions P3 to

P1'. Carrying a Cbz cap in the P3 position, SG75 exhibits significant inhibition

(kobs/[I] = 3246 M-1s-1), whereas SG81, which has a Boc cap in the same position,

25

Page 26: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

is not an inhibitor (kobs/[I] = 42 M-1s-1) (Table 2). From the crystal structures of the

SG75 and SG81 complexes (Fig. 4B and 4C), it can be seen that both the Cbz

and Boc protecting groups follow the binding path of the main chain, rather than

binding in the orientation usually taken by a P3 side-chain, and the bulky Cbz

moiety even partially extends into the S4 subsite. In addition, the carbonyl

oxygen of the protecting group of SG75 accepts a 3.3-Å hydrogen bond (3.4 Å in

case of SG81) from the NH of Gly164. When we increased the lengths of the

compounds by one more amino-acid residue, i.e. having the protecting group in

P4 and an amino acid in P3, orientation of the side-chain of the latter towards the

solvent was observed. The EV68 3Cpro (like its homologues from other viruses)

has no S3 pocket, and hence, a large number of side-chains in P3 are tolerated

by the enzyme. However, there was an interesting correlation between the length

of an aliphatic side-chain in P3 and cell toxicity. Carrying a tert-butyl ester of

glutamate in P3, SG82 (Cbz-GluOtBu-Phe-GlnLactam-CH=CH-COOEt; kobs/[I] =

43225 M-1s-1; Table 2 and Fig. 4D) was slightly toxic to Huh-T7 cells and also

exhibited some adverse effects in BGM and Hela Rh cells (see Table 3).

However, when we shortened the P3 side-chain by one methylene group to give

the tert-butyl ester of aspartate in SG83 (Cbz-AspOtBu-Phe-GlnLactam-CH=CH-

COOEt, kobs/[I] = 174500 s-1M-1; Table 2 and Fig. 4E), the compound was non-

toxic (CC50 = 295 µM). Finally, the P3 side-chain is a tert-butyl ether of serine in

our most potent compound so far (SG85, Cbz-Ser(OtBu)-Phe-GlnLactam-

CH=CH-COOEt, kobs/[I] = 202200 s-1M-1; Table 2 and Fig. 4G). This compound is

also non-toxic (CC50 = 265 μM) for cells (see Supplemental Fig. S1).

566

567

568

569

570

571

572

573

574

575

576

577

578

579

580

581

582

583

584

585

586

587

588

26

Page 27: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

589

590

591

592

593

594

595

596

597

598

599

600

601

602

603

604

605

606

607

608

609

610

611

P4 position

The S4 pocket can only tolerate small side chains, with alanine being the most

favored one by the 3Cpro of EV68. Since our inhibitors have much larger groups

at this position, such as Cbz in SG82, SG83, and SG85, Fmoc in SG84, and Boc

in SG98, these groups cannot penetrate into the S4 pocket; rather they follow the

path of the substrate main-chain and protrude partly into S5. There is no clear

electron density for Fmoc in SG84 (Fig. 4F), and the compound, which was an

intermediate in the synthesis of other inhibitors, is both toxic and has low in-vitro

activity (kobs/[I] = 9225 M-1s-1) when compared to SG83 or SG85. In contrast, the

Cbz and Boc caps of SG82, SG83, SG85, and SG98 can be easily modeled into

the Fo-Fc density. The P4-Cbz group makes strong hydrophobic interactions with

residues Leu125, Asn165, Phe170, and Tyr122 of the S4 site (Fig. 4D,E,G). A

comparison of SG85 (Cbz-Ser(OtBu)-Phe-GlnLactam-CH=CH-COOEt) with

SG98 (Boc-Thr(OtBu)-Phe-GlnLactam-CH=CH-COOEt; kobs/[I] = 11045 M-1s-1)

suggests that Cbz in P4 leads to stronger hydrophobic interactions (Fig. 4G,H)

and explains the much higher inhibitory activity of SG85.

Inhibitory effect on subgenomic replicons

Using enterovirus (PV and EV71) and hepatovirus (HAV) replicons, initial

evaluation of the inhibitory activity of our compounds was performed at 40 μM

concentration (Fig. 5). When examined microscopically, SG84 exhibited strong

27

Page 28: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

612

613

614

615

616

617

618

619

620

621

622

623

624

625

626

627

628

629

630

631

632

633

634

cellular toxicity and was therefore not studied further. However, at a

concentration of 100 µM, none of our compounds reduced by more than 10%

(Table 2 and Supplemental Fig. S1) the metabolic activity of Huh-T7 cells and

their viability according to cell toxicity assays (see Materials & Methods). SG81

did not result in inhibition of replicon replication and was therefore excluded as

well. SG75, SG83, SG85, and SG98 efficiently inhibited the replication of the PV

and EV71 replicons, but none inhibited the replication of the HAV replicon (Fig.

5). The HAV 3C protease is an outlier among the picornaviral 3C proteases, with

significant deviations in the specificity-determining subsites (overall r.m.s.d. from

EV68 3Cpro: 2.04 Å), probably explaining this lack of activity. The HAV replicon

was consequently excluded from further investigation. From the dose-response

curves presented in Fig. 6, SG85 and SG83 appear to be the most potent

inhibitors of PV replicon replication (EC50 = 0.5 µM and 1.0 µM, respectively).

SG98 and SG75 were markedly less effective (4.0 μM and 7.5 μM, respectively).

Comparable results were obtained with the EV71 replicon (Fig. 7), where SG85

and SG83 proved more potent than SG98 and SG75. Direct comparison of the

results of DNA and RNA replicon transfection in Huh-T7 cells excluded the

involvement of the cellular T7 RNA polymerase in inhibition, implying that mainly

replicon genome translation and replication steps were targeted by the inhibitors

(see Supplemental Material). Moreover, based on the observed predominant

inhibition of the replication-competent (up to 95%), as compared to the

replication-deficient (20 to 50%) replicon (Supplemental text and Fig. S2), the

picornavirus 3C protease, the proteolytic activity of which is a prerequisite for the

28

Page 29: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

635

636

637

638

639

640

641

642

643

644

645

646

647

648

649

650

651

652

653

654

655

656

657

formation of a functional replication complex (30, 32), was confirmed as the likely

in-vivo target of our compounds.

Antiviral activity of peptidic α,β-unsaturated esters against picornaviruses

The effect of most compounds was also evaluated on the replication of various

picornaviruses in live virus-cell-based assays employing ECHO virus 11, EV71,

multiple HRV types, and PV1. Both SG74 and SG81 proved to be inactive

against these viruses at the highest concentration tested or to produce high-µM

EC50 values. Interestingly and despite of the high EC50 values, both compounds

were shown to perfectly inhibit virus-induced cytopathic effects for at least one

concentration in the HRV29 assay (Table 3) without any adverse effects on the

host cell. Compound SG75, with Cbz in the P3 position (as opposed to SG81,

which has Boc in this position), exhibited potent and selective antiviral activity

against all viruses included in the test panel, except for PV1, for which it showed

a high EC50 value. Most potent and same order-of-magnitude activity (low-µM

EC50 values) was observed against the different rhinovirus types (order of

susceptibility HRV70 > 14 > 2 > 29 > 85). Upon comparison, SG82, SG83, and

SG85 showed a similar antiviral potency for each individual virus. Throughout the

entire data set and similar to SG75, SG82, SG83, and SG85 were shown to

completely protect host cells from virus-induced cytopathic effects except in the

PV1 assay and to some extent in the HRV2 assay. Compared to SG75, activity

was significantly more pronounced as evident from the lower EC50 values. For

most rhinoviruses, on average, even high nM values were reached. Activity

29

Page 30: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

658

659

660

661

662

663

664

665

666

667

668

669

670

671

672

673

674

675

676

677

678

679

680

against ECHO11 and PV1 was markedly less pronounced. Most of the results of

the cell-based assays show the same trends as those from the enzymatic assay

and the replicons, except for SG82 that proved somewhat less active at the

enzymatic level as compared to SG83 and SG85.

DISCUSSION

It has been observed that enterovirus 68 shares properties with the rhinoviruses

and the enteroviruses (6, 17, 31). This is in line with our X-ray crystallographic

analysis of the EV68 3C protease. The three-dimensional structure of the

enzyme is closely related to those of its homologues from rhinovirus 2 and

poliovirus, whereas the structures of the 3Cpro of enterovirus 71, enterovirus 93,

and Coxsackievirus B3 are somewhat more distantly related and those of

Hepatitis A Virus and Foot-and-Mouth Disease Virus are markedly different.

We have shown that the EV68 3C protease is a target for anti-EV68 drug

discovery and, beyond that, is very useful for the design of broad-spectrum anti-

picornavirus inhibitors. A series of peptidic α,β-unsaturated ethyl esters of

growing lengths has been designed on the basis of the crystal structure of the

EV68 3Cpro and synthesized. In agreement with earlier reports on short inhibitors

of the EV71 3Cpro (18), SG74 (Boc-GlnLactam-CH=CH-COOEt), a compound

containing only residues P2 to P1', had no inhibitory activity against the enzyme

of EV68. In spite of that, we could identify clear electron density for the

compound in its complex with the enzyme. SG81, with Boc as the P3 cap, was

30

Page 31: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

681

682

683

684

685

686

687

688

689

690

691

692

693

694

695

696

697

698

699

700

701

702

703

an intermediate compound and did not show good inhibition. Again, however, it

did bind to the substrate-binding pocket in the crystal structure. With introduction

of the Cbz group at the P3 position (as in SG75), the inhibitory activity increased.

This can be explained by the hydrophobic contacts that the Cbz group makes by

occupying the S3 binding site for the substrate's main chain and, beyond that, by

partly extending into S4.

A comparison of SG82, SG83, SG84, SG85, and SG98, which comprise residues

P4 to P1’, reveals variable inhibitory potency and cytotoxicity. SG82 has a larger

group (GluOtBu) in P3 compared to SG83 and SG85; it showed lower inhibition

and some cytotoxicity in Huh-T7 cells. Upon shortening the side chain by one

methylene group to yield AspOtBu as in SG83, the cytotoxicity disappeared

completely. In contrast, SG84 has a fluorescein group in P4 and was cytotoxic.

After replacing this group by Boc, as in SG98, cytotoxicity was no longer

observed. However, with the Boc group in P4, the inhibitory activity of SG98 was

by far not as good as that of SG85, which has SerOtBu in P3 and Cbz in P4. So

far, SG85 proved to be the best inhibitor against the EV68 3C protease. We have

tested all these compounds as inhibitors of picornaviral replicons and also

against infection by EV71, poliovirus, ECHO virus 11, and various rhinovirus

types at the cellular level. Again, SG85 showed the best inhibitory activity with

(sub)micromolar EC50 values in the PV and EV71 replicons. SG83 and SG85

proved to be the most potent inhibitors of the series in terms of inhibition of

rhinovirus replication and, together with SG82, are about equipotent as inhibitors

31

Page 32: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

704

705

706

707

708

709

710

711

712

713

714

715

716

717

718

719

720

721

722

723

724

725

of EV71 replication. Our next aim is to optimize our lead compound, SG85,

further, to yield antivirals with improved stability, bioavailability, and activity

against a broad spectrum of picornaviruses.

Acknowledgements

We are grateful to Raul Andino and to Bo Zhang for providing PV and EV71

replicon DNAs, resp., to Jiajie Zhang for discussion, and to Doris Mutschall,

Walter Verheyen, Maria Michailow, Ekaterina Shimanovskaya, and Violaien

Lantez for expert technical assistance. We also would like to acknowledge Stijn

Delmotte, Mieke Flament, Tom Bellon, Annelies De Ceulaer, and Kim Donckers

for their help in collecting the live virus-cell-based data. The work presented here

was supported by the European Commission, initially through its VIZIER project

(contract no. LSHG-CT-2004-511960) and subsequently by its SILVER project

(contract no HEALTH-F3-2010-260644). We also acknowledge support from the

Schleswig-Holstein Innovation Fund. RH thanks the DFG Cluster of Excellence

"Inflammation at Interfaces" (EXC 306) and the Fonds der Chemischen Industrie

for continuous support. He is also supported by a Chinese Academy of Sciences

Visiting professorship for Senior International Scientists, grant no. 2010T1S6.

Céline Lacroix is supported by a Ph.D. grant of the Agency for Innovation by

Science and Technology (IWT). We acknowledge access to beamline BL14.1

operated by the Helmholtz-Zentrum Berlin (HZB) at the BESSY II electron

storage ring (Berlin-Adlershof, Germany).

32

Page 33: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

726

727

728

729

730

731

732

733

734

735

736

737

738

739

740

741

742

743

744

745

746

747

748

References

1. Anand K, Ziebuhr J, Wadhwani P, Mesters JR, Hilgenfeld R. 2003.

Coronavirus main proteinase (3CLpro) structure: basis for design of anti-

SARS drugs. Science 300:1763-1767.

2. Andino R, Rieckhof GE, Achacoso PL, Baltimore D. 1993. Poliovirus

RNA synthesis utilizes an RNP complex formed around the 5`- end of viral

RNA. EMBO J. 12:3587-3598.

3. Bergmann EM, Mosimann SC, Chernaia MM, Malcolm BA, James MN.

1997. The refined crystal structure of the 3C gene product from hepatitis A

virus: specific proteinase activity and RNA recognition. J. Virol. 71:2436-

2448.

4. Berrow NS, Alderton D, Sainsbury S, Nettleship J, Assenberg R,

Rahman N, Stuart DI, Owens RJ. 2007. A versatile ligation-independent

cloning method for high-throughput expression screening applications.

Nucleic Acids Res. 35:e45

5. Birtley JR, Knox SR, Jaulent AM, Brick P, Leatherbarrow RJ, Curry

S. 2005. Crystal structure of Foot- and Mouth-disease virus 3C protease.

J. Biol. Chem. 280:11520-11527.

33

Page 34: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

749

750

751

752

753

754

755

756

757

758

759

760

761

762

763

764

765

766

767

768

769

770

771

6. Blomqvist S, Savolainen C, Råman L, Roivainen M, Hovi T. 2002.

Human rhinovirus 87 and enterovirus 68 represent a unique serotype

with rhinovirus and enterovirus properties. J. Clin. Microbiol. 40:4218-

4223.

7. Brandl M, Weiss MS, Sühnel J, Hilgenfeld R. 2001. C-H...π

interactions in proteins. J. Mol. Biol. 307:357-377.

8. Collaborative Computational Project No. 4. 1994. The CCP4 suite:

programs for protein crystallography. Acta Crystallogr., Sect. D: Biol.

Crystallogr. 50:760-763.

9. Collett MS, Neyts J, Modlin JF. 2008. A case for developing antiviral

drugs against polio. Antiviral Res. 79:179-187.

10. Costenaro L, Kaczmarska Z, Arnan C, Janowski R, Coutard B, Sola

M, Gorbalenya AE, Norder H, Canard B, Coll M. 2011. Structural basis

of antiviral inhibition of the main protease, 3C, from enterovirus 93. J.

Virol. 85:10764-10773.

11. Dragovich PS, Prins TJ, Zhou R, Webber SE, Marakovits JT,

Fuhrman SA, Patick AK, Matthews DA, Lee CA, Ford CE, Burke BJ,

34

Page 35: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

Rejto PA, Hendrickson TF, Tuntland T, Brown EL, Meador JW, Ferre

RA, Harr JEV, Kosa MB, Worland ST. 1999. Structure-based design,

synthesis, and biological evaluation of irreversible human rhinovirus 3C

protease inhibitors. 4. Incorporation of P1 lactam moieties as L-glutamine

replacements. J. Med. Chem. 42:1213-1224.

772

773

774

775

776

777

778

779

780

781

782

783

784

785

786

787

788

789

790

791

792

793

794

12. Emsley P, Cowtan K. 2004. Coot: Model-building tools for molecular

graphics. Acta Crystallogr. D Biol. Crystallogr. D60:2126-2132.

13. Evans P. 2006. Scaling and assessment of data quality. Acta Crystallogr.

D Biol. Crystallogr. 62:72-82.

14. Gauss-Müller V, Kusov Y. 2002. Replication of a hepatitis A virus

replicon detected by genetic recombination in vivo. J. Gen. Virol.

83:2183-2192.

15. Hayden FG, Turner RB, Gwaltney JM, Chi-Burris K, Gersten M, Hsyu

P, Patick AK, Smith III GJ, Zalman LS. 2003. Phase II, randomized,

double-blind, placebo-controlled studies of rupintrivir nasal spray 2-

percent suspension for prevention and treatment of experimentally

induced rhinovirus colds in healthy volunteers. Antimicrob. Agents

Chemother. 47:3907–3916.

35

Page 36: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

16. Hilgenfeld R. 2010. Structure-based antivirals for emerging and

neglected RNA viruses: an emerging field for medicinal chemistry in

academia. Future Med. Chem. 2:1061-1067.

795

796

797

798

799

800

801

802

803

804

805

806

807

808

809

810

811

812

813

814

815

816

817

17. Ishiko H, Miura R, Shimada Y, Hayashi A, Nakajima H, Yamazaki S,

Takeda N. 2002. Human rhinovirus 87 identified as human enterovirus

68 by VP4-based molecular diagosis. Intervirology 45:136-141.

18. Kuo CJ, Shie JJ, Fang JM, Yen GR, Hsu JTA, Liu HG, Tseng SN,

Chang SC, Lee CY, Shih SR, Liang PH. 2008. Design, synthesis, and

evaluation of 3C protease inhibitors as anti-enterovirus 71 agents. Bioorg.

Med. Chem.16:7388-7398.

19. Laskowski RA, MacArthur MW, Moss DS, Thornton JM. 1993.

PROCHECK: a program to check the stereochemical quality of protein

structures. J. Appl. Crystallogr. 26:283-291.

20. Lee CC, Kuo CJ, Ko TP, Hsu MF, Tsui YC, Chang SC, Yang S, Chen

SJ, Chen HC, Hsu MC, Shih SR, Liang PH, Wang AH. 2009. Structural

basis of inhibition specificities of 3C and 3C-like proteases by zinc-

coordinating and peptidomimetic compounds. J. Biol. Chem. 284:7646-

7655.

36

Page 37: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

21. Lee ES, Lee WG, Yun SH, Rho SH, Im I, Yang ST, Sellamuthu S, Lee

YJ, Kwon SJ, Park OK, Jeon ES, Park WJ, Kim YC. 2007.

Development of potent inhibitors of the coxsackievirus 3C protease.

Biochem. Biophys. Res. Commun. 358:7-11.

818

819

820

821

822

823

824

825

826

827

828

829

830

831

832

833

834

835

836

837

838

839

22. Leslie AGW, Powell HR. 2007. Evolving Methods for Macromolecular

Crystallography, eds Read RJ, Sussmann JL (Springer, Berlin), vol 245,

pp 41–51.

23. Lu GW, Qi JX, Chen ZJ, Xu X, Gao F, Lin DZ, Qian WK, Liu H, Jiang

HL, Yan JH, Gao GF. 2011. Enterovirus 71 and Coxsackievirus A16 3C

proteases: Binding to rupintrivir and their substrates and anti-Hand, Foot,

and Mouth Disease Virus drug design. J. Virol. 85:10319-10331.

24. Matthews BW. 1968. Solvent content of protein crystals. J. Mol. Biol.

33:491-497.

25. Matthews DA, Dragovich PS, Webber SE, Fuhrman SA, Patick AK,

Zalman LS, Hendrickson TF, Love RA, Prins TJ, Marakovits JT,

Zhou R, Tikhe J, Ford CE, Meador JW, Ferre RA, Brown EL, Binford

SL, Brothers MA, DeLisle DM, Worland ST. 1999. Structure-assisted

design of mechanism-based irreversible inhibitors of human rhinovirus

37

Page 38: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

840

841

842

843

844

845

846

847

848

849

850

851

852

853

854

855

856

857

858

859

860

861

862

3C protease with potent antiviral activity against multiple rhinovirus

serotypes. Proc. Natl. Acad. Sci. USA 96:11000-11007.

26. Meara JP, Rich DH. 1995. Measurement of individual rate constants of

irreversible inhibition of a cysteine proteinase by an epoxysuccinyl

inhibitor. Bioorg. Med. Chem. Lett. 5:2277-2282.

27. Mosimann SC, Cherney MM, Sia S, Plotch S, James MN. 1997.

Refined X-ray crystallographic structure of the poliovirus 3C gene

product. J. Mol. Biol. 273:1032-1047.

28. Mou K, Xu B, Ma C, Yang X, Zou X, Lü Y, Xu P. 2008. Novel CADD-

based peptidyl vinyl ester derivatives as potential proteasome inhibitors.

Bioorg. Med. Chem. Lett. 18:2198–2202.

29. Murshudov GN, Vagin AA, Dodson EJ. 1997. Refinement of

macromolecular structures by the maximum-likelihood method. Acta

Crystallogr. D Biol. Crystallogr. 53:240-255.

30. Norder H, De Palma AM, Selisko B, Costenaro L, Papageorgiou N,

Arnand C, Coutard B, Lantez V, De Lamballerie X, Baronti C, Solà M,

Tan J, Neyts J, Canard B, Coll M, Gorbalenya AE, Hilgenfeld R. 2011.

Picornavirus non-structural proteins as targets for new anti-virals with

38

Page 39: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

broad activity. Antiviral Res. 89:204-218. 863

864

865

866

867

868

869

870

871

872

873

874

875

876

877

878

879

880

881

882

883

31. Oberste MS, Maher K, Schnurr D, Flemister MR, Lovchik JC, Peters

H, Sessions W, Kirk C, Chatterjee N, Fuller S, Hanauer JM,

Pallansch MA. 2004. Enterovirus 68 is associated with respiratory illness

and shares biological features with both the enteroviruses and the

rhinoviruses. J. Gen. Virol. 85:2577-2584.

32. Pallansch M, Roos R. 2007. Enteroviruses: Polioviruses,

coxsackieviruses, echoviruses, and newer enteroviruses. In: Knipe, D.

M., and P. M. Howley, eds., Fields Virology, 5th Edition. Lippincott

Williams & Wilkins. pp. 840-893.

33. Rossmann MG. 1990. The molecular replacement method. Acta

Crystallogr. A Foundations Crystallogr. 46:73-82.

34. Schultz DE, Honda M, Whetter LE, McKnight KL, Lemon SM. 1996.

Mutations within the 5` nontranslated RNA of cell culture-adapted

hepatitis A virus which enhance cap-independent translation in cultured

African green monkey cells. J. Virol. 70:1041-1049.

39

Page 40: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

35. Steuber H, Hilgenfeld R. 2010. Recent advances in targeting viral

proteases for the discovery of novel antivirals. Curr. Top. Med. Chem.

23:323-345.

884

885

886

887

888

889

890

891

892

893

894

895

896

897

898

899

900

901

902

903

904

905

906

36. Tan J, Verschueren KH, Anand K, Shen J, Yang M, Xu Y, Rao Z,

Bigalke J, Heisen B, Mesters JR, Chen K, Shen X, Jiang H,

Hilgenfeld R. 2005. pH-dependent conformational flexibility of the

SARS-CoV main proteinase (Mpro) dimer: molecular dynamics

simulations and multiple X-ray structure analyses. J. Mol. Biol. 354:25-

40.

37. Vagin A, Teplyakov A. 1997. MOLREP: an automated program for

molecular replacement. J. Appl. Cryst. 30:1022–1025.

38. Vaguine AA, Richelle J, Wodak SJ. 1999. SFCHECK: a unified set of

procedures for evaluating the quality of macromolecular structure-factor

data and their agreement with the atomic model. Acta Crystallogr. D Biol.

Crystallogr. 55:191-205.

39. Verschueren KHG, Pumpor K, Anemüller S, Chen S, Mesters JR,

Hilgenfeld R. 2008. A structural view of the inactivation of the SARS-

coronavirus main proteinase by benzotriazole esters. Chem. Biol.

15:597-606.

40

Page 41: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

907

908

909

910

911

912

913

914

915

916

917

918

919

920

921

922

923

40. Weiss MS, Brandl M, Sühnel J, Pal D, Hilgenfeld R. 2001. More

hydrogen bonds for the (structural) biologist. Trends Biochem. Sci.

26:521-523.

41. Wilkinson GN. 1961. Statistical estimations in enzyme kinetics,

Biochem. J. 80:324-332.

42. Yang F, Ren L, Xiong Z, Li J, Xiao Y, Zhao R, He Y, Bu G, Zhou S,

Wang J, Qi J. 2009. Enterovirus 71 outbreak in the People’s Republic of

China in 2008. J. Clin. Microbiol. 47:2351-2352.

43. Yang H, Xie W, Xue X, Yang K, Ma J, Liang W, Zhao Q, Zhou Z, Pei

D, Ziebuhr J, Hilgenfeld R, Yuen KY, Wong L, Gao G, Chen S, Chen

Z, Ma D, Bartlam M, Rao Z. 2005. Design of wide-spectrum inhibitors

targeting coronavirus main proteases. PLoS Biol. 3:e324.

41

Page 42: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

924

925

926

927

928

929

930

931

932

933

934

935

936

937

938

939

940

941

942

943

944

945

946

Figure legends

Fig. 1. (A) SG74 was synthesized using a published procedure (28). Reagents

and conditions: (a) (i) Me3SiCl, MeOH, 0-25°C, 18 h; (ii) Boc2O, Et3N, THF, 0-

25°C, 12 h, 96%; (b) LiN(SiMe3)2, THF, -78°C, 1 h; then BrCH2CN, 4 h; (c)

NaBH4, CoCl2.6H2O, MeOH, 0-25°C, 12-48 h, 73%; (d) NaBH4, LiCl, EtOH, THF

(1:1), 0-25°C, 12 h, 50%; (e) (i) SO3.Py, CH2Cl2, 0-25°C, 2 h; (ii) PPh3CHCO2Et,

25°C, 2 h, 96%; (f) (i) TFA:CH2Cl2 (1:2), 2 h; (ii) EDCI, HOBt, Z-Phe-OH, DMF, 0-

25°C, 18 h, 62%; (g) (i) TFA:CH2Cl2 (1:2), 2 h; (ii) EDCI, HOBt, Boc-Phe-OH,

DMF, 0-25°C, 18 h, 74%. (B) Reagents and conditions: (a) (i) TFA:CH2Cl2 (1:2),

2 h; (ii) EDCI, HOBt, protected amino acid, DMF, 0-25°C, 18 h, 46-70%.

Fig. 2. Substrate specificity of EV68 3Cpro. Among the peptides corresponding to

its polypeptide cleavage sites, the EV68 3Cpro displayed high activities towards

EALFQ↓GPPQF (2C↓3A) and TAKVQ↓GPGFD (3B↓3C); the former was

selected for further characterization of the enzyme's activity (not shown).

Fig. 3. (A) The three-dimensional structure of EV68 3Cpro. The catalytic triad

consisting of Cys147, His40, and Glu71 is shown in stick mode. Hydrogen bonds

between these residues are indicated by dashed lines, with the corresponding

distances given in Å. (B) 2Fo-Fc electron density (contoured at 1σ above the

mean) for the catalytic center. Note the two alternative conformations for His40;

42

Page 43: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

947

948

949

950

951

952

953

954

955

956

957

958

959

960

961

962

963

964

965

966

967

968

only one of these interacts with Cys147. (C) Structure-based sequence alignment

of 3C proteases of various picornaviruses. The secondary structure (as found in

the crystal structure of the EV68 enzyme) is shown at the top, invariant residues

are highlighted with red background, and conserved residues are shown in red.

Fig. 4. Binding-mode analysis by X-ray crystallography of α,β-unsaturated ethyl

ester inhibitors. (A) to (H) show the binding modes of SG74, SG75, SG81, SG82,

SG83, SG84, SG85, and SG98, respectively. The catalytic triad is shown in

yellow, while other residues involved in the binding site are shown in pink.

Inhibitors are shown in green along with an omit 2Fo-Fc map (blue) contoured at

1.0 σ above the mean. A stereo view of this figure can be found in the on-line

Supplemental Material.

Fig. 5. Initial evaluation of selected Michael acceptor compounds (40 µM) for

inhibition of PV, EV71, and HAV replicons in Huh-T7 cells. “-” – no inhibitor

added to the cell-growth medium after PV, EV71, or HAV replicon transfection. *

– compound SG82 (yellow bar) was not tested in the EV71 replicon. RLU –

relative light units of firefly (PV and HAV replicons) or Renilla (EV71 replicon)

luciferase. The results are means of quadruplicate +/- SD.

Fig. 6. The 50% effective concentration (EC50) of SG75 (filled circles), SG83

(empty circles), SG85 (filled triangles), and SG98 (empty triangles) on the

43

Page 44: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

replication of the PV replicon in Huh-T7 cells. The values represent the means of

quadruplicate wells +/- SD.

969

970

971

972

973

974

975

976

977

978

979

980

981

Fig. 7. Concentration-dependent inhibition of EV71 replication-competent

replicon DNA by SG75 (filled circles), SG83 (empty circles), SG85 (filled

triangles), and SG98 (empty triangles).

44

Page 45: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

Table 1. Data collection and refinement statistics for crystals of EV68 3Cpro and

its complexes

982

983

EV68 3Cpro EV68 3Cpro/SG74

EV68 3Cpro/SG75

EV68 3Cpro/SG81

EV68 3Cpro/SG82

Data collection statistics

Space group P3121 P3121 P3121 P3121 P3121 Unit cell parameters (Å)

56.34, 56.34, 171.00

55.99, 55.99, 170.55

56.10, 56.10, 170.41

56.32, 56.32, 170.34

56.49, 56.49, 170.36

Estimated solvent contenta (%)

67.05 66.55 66.65 66.90 67.10

Wavelength (Å) 0.9184 0.9184 0.9184 0.9184 0.9184 Resolution range (Å) 42.75-2.40

(2.53-2.40) 34.11-2.05 (2.16-2.05)

36.92-2.20 (2.32-2.20)

48.77-2.65 (2.79-2.65)

34.07-2.00 (2.11-2.00)

Number of reflections measured

100289 (14756)

183050 (26814)

118096 (17334)

172402 (25467)

200417 (29073)

Unique reflections 13045 (1878) 20341 (2929) 16608 (2389) 9730 (1394) 22202 (3176) Completeness (%) 100 (100) 100 (100) 100 (100) 100 (100) 100 (100) Rmerge (%)b,c 0.099 (0.569) 0.092 (0.480) 0.089 (0.455) 0.137 (0.480) 0.086 (0.454) Redundancy 7.7 (7.9) 9.0 (9.2) 7.1 (7.3) 17.7 (18.3) 9.0 (9.2) I/σ(I) 14.1 (3.9) 15.7 (4.6) 16.0 (5.4) 18.7 (6.8) 17.2 (5.4) Mosaicity (o) 0.48 0.47 0.45 0.33 0.34 Refinement statistics Rcryst/Rfree (%) 20.7 (23.8) 18.9 (21.8) 19.3 (22.8) 19.1 (25.9) 19.3 (22.2) r.m.s deviation from ideal geometry

Bonds (Å) 0.019 0.019 0.018 0.019 0.019 Angles (°) 1.953 1.600 1.624 1.936 1.662 Ramanchandran plot Favored (%) 95.2 96.2 95.2 93.0 97.3 Allowed (%) 4.8 3.2 3.8 7.0 2.7 Outlier (%) 0.0 0.5 1.0 0.0 0.0 PDB code 3zv8 3zv9 3zva 3zvb 3zvc

984

45

Page 46: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

985

986

987

988

Table 1 (continued). Data collection and refinement statistics for crystals of EV68

3Cpro and its complexes

EV68

3Cpro/SG83 EV68 3Cpro/SG84

EV68 3Cpro/SG85

EV68 3Cpro/SG98

Data collection statistics

Space group P3121 P3121 P3121 P3121 Unit cell parameters (Å)

56.31, 56.31, 170.35

56.30, 56.30, 170.35

55.87, 55.87, 170.15

56.20, 56.20, 170.23

Estimated solvent contenta (%)

66.89 66.88 66.33 66.74

Wavelength (Å) 0.9184 0.9184 0.9184 0.9184 Resolution range (Å) 36.99-2.25

(2.37-2.25) 34.07-1.80 (1.90-1.80)

36.81-2.50 (2.64-2.50)

32.04-2.10 (2.21-2.10)

Number of reflections measured

140050 (20152)

255740 (36767)

95365 (13852)

131896 (19127)

Unique reflections 15635 (2205) 29927 (4268) 11333 (1606) 19052 (2724) Completeness (%) 100 (100) 100 (100) 100 (100) 100 (100) Rmerge(%)b,c 0.110 (0.468) 0.064 (0.445) 0.128 (0.554) 0.094 (0.445) Redundancy 9.0 (9.1) 8.5 (8.6) 8.4 (8.6) 6.9 (7.0) I/σ(I) 15.4 (5.4) 17.3 (4.2) 11.9 (4.0) 13.2 (4.6) Mosaicity (o) 0.39 0.46 1.31 0.29 Refinement statistics Rcryst/Rfree (%) 19.3 (24.7) 20.4 (23.1) 19.5 (24.5) 19.9 (23.8) r.m.s deviation from ideal geometry

Bonds (Å) 0.019 0.016 0.020 0.019 Angles (°) 1.660 1.622 1.969 1.680 Ramanchandran plot Core (%) 96.2 96.8 94.6 95.2 Allowed (%) 3.2 3.2 5.4 4.8 General (%) 0.5 0.0 0.0 0.0 PDB code 3zvd 3zve 3zvf 3zvg

989 990 991

992

Values in parentheses are for the highest resolution shell.

a Solvent content estimated according to Matthews (24).

b ( ) ( )| | ( )∑ ∑∑ ∑ −= iihkliihklmerge hklIhklIhklIR , where ( )hklI is the intensity of

reflection hkl and

993

( )hklI is the average intensity over all equivalent reflections. 994

46

Page 47: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

c ( ) ( )| | ( )∑∑ −= hklFhklFhklFR ohklcohklcryst . freeR was calculated for a test set of

reflections (5%) omitted from the refinement.

995

996

997

998

999

47

Page 48: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

Table 2. Enzyme inhibition and inhibition of replicons by α,β-unsaturated esters 1000 1001

EV68 3Cpro

inhibition inhibition of replicons

(EC50, μM)

Toxicity (CC50, μM)

Name Molecular structure kobs/[I] (M-1s-1) PV EV71 HAV Huh-T7

SG74 O N

H

OO

O

NHO

ND > 40 ND* ND* >40

SG75 NH

OO

O

NHO

HNO

O

3246±107 7.5±1.5 7.0±1.1 30±12 320±30

SG81 NH

OO

O

NHO

HNO

O

42±3 > 40 > 40 ≥40 ND*

SG82 NH

OO

O

NHO

HN

O

O O

O

NH

O

43225±1850 ND* 1.2±0.2 >20 >100

SG83 NH

NHO

O

O

OHN

ONH

O

O

O

O

174500±9245 1.0±0.3 1.3±0.5 12±2.5 295±15

SG84 NH

OO

O

NHO

OHN

O

O

NH

O

9225±423 ND* 2.5±0.4 ND* >100

SG85 NH

OO

O

NHO

OO H

N

ONH

O

202200±8103 0.5±0.1 1.0±0.2 15±5 265±15

48

Page 49: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

SG98 NH

OO

O

NHO

HN

ONH

O

OO

11045±661 4.0±1.4 2.5±0.5 >40 250±10

1002 1003

* ND – not determined

49

Page 50: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position

50

1004 1005 1006 1007

Table 3. Antiviral activity of SG compounds on the replication of a panel of entero- and rhinoviruses in a live virus-cell-based assay

Code Echo11 (Gregory; BGM)

EV71 (BRCR; RD)

HRV02 (Hela Rh)

HRV14 (Hela Rh)

HRV29 (Hela Rh)

HRV70 (Hela Rh)

HRV85 (Hela Rh)

PV1 (Sabin; BGM)

SG74 97 ± 18 >307 57 ± 28 >307 55 ± 21 >307 242 >307

SG75 63 ± 10 9.3 ± 1.0 2.0 ± 0.5 1.5 ± 1.0 2.2 ± 1.2 1.5 ± 0.7 5 ± 2 132 ± 1

SG81 98 ± 28 >211 59 ± 33 53 ± 28 31 ± 16 >211 >211 >211

SG82 19 ± 5 0.26 ± 0.26 1.4 ± 1.4 0.11 ± 0.07 0.19 ± 0.14 0.4 ± 0.1 0.4 ± 0.1 41 ± 18

SG83 29 ± 3 0.15 ± 0.03 1.9 ± 1.9 0.05 ± 0.02 0.27 ± 0.18 0.8 ± 0.3 0.6 ± 0.1 43 ± 20

SG85 26 ± 2 0.18 ± 0.19 1.7 ± 2.3 0.06 ± 0.01 0.05 ± 0.03 0.5 ± 0.2 0.14 ± 0.04 47 ± 23

Enviroxime 0.8 ± 0.6 0.8 ± 0.6 0.04 ± 0.02 0.3 ± 0.1 0.15 ± 0.8 0.4 ± 0.3 0.2 ± 0.1 0.5 ± 0.2

1008 1009

1010

1011

1012

1013

1014

1015

1016

1017

1018

Data represent the 50% effective concentration (EC50) expressed in µM and are

averages ± SD for at least 2 independent experiments. Note that relatively high

standard deviations are common in such experiments and may arise because of

minor instabilities of compounds when stored in DMSO, varying dilution factors,

or slight differences between assays. Data shown in grey shade indicate that,

following microscopic inspection, no compound-induced adverse effects were

observed on the host cell and the compound, for at least one concentration,

completely prevented the virus from inducing any cytopathic effects. For

comparison, data for enviroxime, an inhibitor of the 2C NTPase of picornaviruses,

are also shown.

Page 51: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position
Page 52: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position
Page 53: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position
Page 54: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position
Page 55: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position
Page 56: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position
Page 57: JVI Accepts, published online ahead of print on 6 February ...Because of their importance in the viral replication cycle and their unique specificity for glutamine in the P1 position