50
1 A Novel Inhibitor of Dengue Virus Replication that Targets the Capsid Protein 1 2 Chelsea M. Byrd 1, *, Dongcheng Dai 1 , Douglas W. Grosenbach 1 , Aklile Berhanu 1 , Kevin 3 F. Jones 1 , Kara B. Cardwell 1 , Christine Schneider 1 , Kristin A. Wineinger 1 , Jessica M. 4 Page 1 , Chris Harver 1 , Eric Stavale 1 , Shanthakumar Tyavanagimatt 1 , Melialani A. 5 Stone 1 , Ralf Bartenschlager 2 , Pietro Scaturro 2 , Dennis E. Hruby 1 , and Robert Jordan 1+ 6 7 1 SIGA Technologies, Inc., Corvallis, OR 97333 8 + Current address: Gilead Sciences, Inc., Foster City, CA 94404 9 2 Department of Infectious Diseases, Molecular Virology, University of Heidelberg, 10 Heidelberg, Germany 11 12 * Corresponding author: Mailing address: 4575 SW Research Way, Corvallis, OR 13 97333. Phone: 541-753-2000. Fax: 541-753-9999. E-mail: [email protected] 14 15 Running title: A dengue virus inhibitor 16 Keywords: 17 Flavivirus 18 Flavivirus Capsid protein 19 Capsid 20 Antiviral 21 Dengue 22 23 Copyright © 2012, American Society for Microbiology. All Rights Reserved. Antimicrob. Agents Chemother. doi:10.1128/AAC.01429-12 AAC Accepts, published online ahead of print on 15 October 2012 on May 7, 2018 by guest http://aac.asm.org/ Downloaded from

A Novel Inhibitor of Dengue Virus Replication that Targ ...aac.asm.org/content/early/2012/10/09/AAC.01429-12.full.pdf · While there are several promising vaccine cand idates in clinical

  • Upload
    tranbao

  • View
    215

  • Download
    1

Embed Size (px)

Citation preview

1

A Novel Inhibitor of Dengue Virus Replication that Targets the Capsid Protein 1

2

Chelsea M. Byrd1,*, Dongcheng Dai1, Douglas W. Grosenbach1, Aklile Berhanu1, Kevin 3

F. Jones1, Kara B. Cardwell1, Christine Schneider1, Kristin A. Wineinger1, Jessica M. 4

Page1, Chris Harver1, Eric Stavale1, Shanthakumar Tyavanagimatt1, Melialani A. 5

Stone1, Ralf Bartenschlager2, Pietro Scaturro2, Dennis E. Hruby1, and Robert Jordan1+ 6

7

1SIGA Technologies, Inc., Corvallis, OR 97333 8

+Current address: Gilead Sciences, Inc., Foster City, CA 94404 9

2 Department of Infectious Diseases, Molecular Virology, University of Heidelberg, 10

Heidelberg, Germany 11

12

* Corresponding author: Mailing address: 4575 SW Research Way, Corvallis, OR 13

97333. Phone: 541-753-2000. Fax: 541-753-9999. E-mail: [email protected] 14

15

Running title: A dengue virus inhibitor 16

Keywords: 17

Flavivirus 18

Flavivirus Capsid protein 19

Capsid 20

Antiviral 21

Dengue 22

23

Copyright © 2012, American Society for Microbiology. All Rights Reserved.Antimicrob. Agents Chemother. doi:10.1128/AAC.01429-12 AAC Accepts, published online ahead of print on 15 October 2012

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

2

ABSTRACT 24

Dengue viruses (DENV) infect 50-100 million people worldwide per year, of 25

which 500,000 develop severe life-threatening disease. This mosquito-borne illness is 26

endemic in most tropical and sub-tropical countries, and has spread significantly over 27

the last decade. While there are several promising vaccine candidates in clinical trials, 28

there are currently no approved vaccines or therapeutics available for treatment of 29

dengue infection. Here we describe a novel small-molecule compound, ST-148, that is a 30

potent inhibitor of all four serotypes of DENV in vitro. ST-148 significantly reduced 31

viremia and viral load in vital organs, and tended to lower cytokine levels in the plasma 32

in a non-lethal model of DENV infection in AG129 mice. Compound resistance mapped 33

to the DENV capsid (C) gene, and a direct interaction of ST-148 with C protein is 34

suggested by alterations of the intrinsic fluorescence of the protein in the presence of 35

compound. Thus, ST-148 appears to interact with the DENV C protein and inhibits 36

distinct step(s) of the viral replication cycle. 37

38

39

40

41

42

43

44

45

46

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

3

INTRODUCTION 47

Dengue fever (DF) is an acute, febrile illness caused by infection with dengue 48

virus (DENV). DENV circulates in populations as one of four distinct serotypes and is 49

transmitted to humans via the bite of the Aedes mosquito. While DF is a debilitating 50

illness, most cases resolve without sequelae (9). In a small percentage of cases, 51

individuals develop a severe capillary leakage syndrome referred to as dengue 52

hemorrhagic fever (DHF), which can lead to a more severe disease called dengue 53

shock syndrome (DSS) (9, 12). An estimated 50 – 100 million individuals are infected 54

with DENV each year mostly in tropical and subtropical areas of Southeast Asia, 55

resulting in nearly 500,000 severe life-threatening illnesses and 25,000 deaths. The 56

incidence of dengue disease is growing as the mosquito vector spreads due to 57

urbanization, population growth, increased international travel, a decrease in mosquito 58

control efforts, and global warming (11). 59

The existence of four distinct serotypes has made DENV vaccine development 60

challenging. While serotype-specific immunity reduces the rate of reinfection, immunity 61

does not provide complete protection from infection by the other three virus serotypes 62

(18). In fact, a second infection with a different virus serotype can potentially increase 63

the risk of severe disease. This enhanced risk is thought to be due to a combination of 64

viral genetics and heterotypic, non-neutralizing antibodies which enhance virus infection 65

(10). Disease severity has been linked to viral load, and patients with DHF or DSS have 66

viral titers in the blood 10-1000 fold higher than patients with DF (28). Thus, an antiviral 67

drug administered early during the course of infection that inhibits viral replication and 68

decreases viral load might be expected to reduce severity of disease. 69

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

4

DENV belongs to the Flaviviridae family and can be cultured in several 70

transformed cell lines to produce robust cytopathic effects. Upon entry of the virus into 71

the host cell, the positive, single stranded RNA genome is translated into a single 72

polyprotein that is proteolytically processed to produce three structural proteins, capsid 73

(C), pre-membrane (prM) and envelope (E), and seven non-structural proteins, NS1, 74

NS2A, NS2B, NS3, NS4A, NS4B and NS5. The non-structural proteins form the viral 75

replicase that is found within vesicles derived from virus-modified ER membranes (31). 76

Full length, positive stranded viral RNA genomes are synthesized from a negative 77

stranded intermediate (7). The newly synthesized RNA genomes are thought to exit 78

through pores that connect the vesicles to the cytosol (31). The viral core (C) protein 79

associates with the genomic RNA to form the nucleocapsid which buds into the ER 80

lumen to produce the immature virus particle containing viral prM and E glycoproteins 81

(36). The immature virus particles traffic via the secretory pathway and are processed in 82

the late Golgi compartment by a furin protease that cleaves the prM protein to produce 83

infectious virus particles that are released from the cell (34, 35). 84

A number of antiviral compounds have been identified that inhibit DENV 85

replication in vitro and in vivo (reviewed in (20)). Virus-specific inhibitors have been 86

identified that target the viral envelope (22), methyl transferase (16), protease (26), 87

NS4B protein (33), polymerase (6, 19), and virus-specific RNA translation (30). In 88

addition, compounds that target host enzymes such as ER glucosidases (4, 5, 8, 32), 89

dihydroorotate dehydrogenase (30), and an intracellular cholesterol transporter (21) 90

have been shown to have antiviral activity. Although these compounds appear to be 91

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

5

effective at inhibiting DENV replication, there is still no approved antiviral therapeutic for 92

the treatment of DENV infection in humans. 93

To identify potential antiviral therapeutics to treat DENV infection, a high 94

throughput screening assay was developed that measured virus-induced cytopathic 95

effects. This assay was used to screen a chemical compound library composed of over 96

200,000 unique small molecules, to identify inhibitors of DENV replication. A novel 97

compound series with activity against all four DENV serotypes was identified. The lead 98

compound in this series, ST-148, inhibited DENV replication in multiple cell types and 99

reduced viral load in a mouse model of DENV replication. Drug resistance was mapped 100

to the capsid coding region of the virus genome and recombinant DENV containing 101

mutations in this region showed reduced susceptibility to ST-148. The compound 102

altered the intrinsic fluorescence of purified wild-type C protein as well as a mutant C 103

protein containing amino acid changes associated with reduced compound 104

susceptibility. These data suggest that ST-148 inhibits the DENV replication cycle by 105

targeting the C protein. 106

107

108

109

110

111

112

113

114

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

6

METHODS AND MATERIALS 115

116

Compound synthesis 117

ST-148 was purchased from ChemBridge (San Diego, CA) and was dissolved in 118

dimethyl sulfoxide (DMSO) (Sigma Aldrich; St. Louis, MO) to a concentration of 10 mM. 119

120

Cells and viruses 121

Vero, BSC40, BHK, C6/36, L929, MDCK and MDBK cells were obtained from 122

American Type Culture Collection (ATCC; Manassas, VA), and Huh-7 cells were 123

obtained from Japan Health Sciences Foundation (Tokyo, Japan). Vero, BSC40, BHK, 124

and L929 cells were maintained at 37°C with 5% CO2 in Minimal Essential Medium 125

(MEM) supplemented with 10% heat-inactivated fetal bovine serum (FBS), 2 mM L-126

glutamine and 10 µg/ml gentamicin (all culture reagents from Invitrogen; Grand Island, 127

NY). C6/36 cells were maintained at 28°C with 5% CO2 in MEM as described above. 128

MDBK cells were maintained in MEM as described above and supplemented with 10% 129

horse serum (HS) (Invitrogen) instead of FBS. MDCK cells were maintained in Eagle’s 130

Modified Essential Medium (EMEM) (ATCC) with 10% FBS and 100U/ml Penicillin and 131

100 µg/ml Streptomycin (Invitrogen). Dengue-1 (DENV-1) strain TH-Sman (VR-344), 132

dengue-2 (DENV-2) strain New Guinea C (NGC) (VR-1255), dengue-3 (DENV-3) strain 133

H87 (VR-1256), and dengue-4 (DENV-4) strain H241 (VR-1257) were purchased from 134

ATCC. DENV-2 K0049 was obtained from Dr. Rebecca Rico-Hesse (Texas Biomedical 135

Research Institute; San Antonio, TX). DENV-2 S221 was obtained from Dr. Sujan 136

Shresta (La Jolla Institute for Allergy and Immunology; La Jolla, CA). The DENV were 137

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

7

propagated on Vero or C6/36 cell cultures. Bovine viral diarrhea virus (BVDV) strain 138

NADL (VR-534), Modoc virus strain M544 (VR-415), influenza A strain A/PR/8/34 (VR-139

1469), Sindbis virus strain AR-339 (VR-1585), and herpes simplex virus (HSV) strain 140

KOS (VR-1493) were purchased from ATCC. Recombinant vaccinia virus expressing 141

the green fluorescent protein (vvGFP) is described elsewhere (3). All cell culture 142

incubations, except for C6/36 cell culture, were performed at 37°C with 5% CO2. 143

144

High throughput screening assay 145

Vero cell monolayers were seeded on 96-well plates at 4.0 x 103 cells per well 146

and incubated overnight. The cells were then infected with DENV-2 at a multiplicity of 147

infection (moi) of 0.1 and incubated for 5 days with 5 µM compound added by a 148

Multiprobe II high-throughput system (PerkinElmer; Waltham, MA). Infected cell 149

monolayers were fixed with 5% glutaraldehyde (JT Baker; Central Valley, PA) in 150

phosphate-buffered saline (Fisher; Pittsburgh, PA), and stained with 0.1% crystal violet 151

(Sigma) in 5% methanol for 30 min. Virus-induced cytopathic effects (CPE) were 152

quantified by measuring absorbance at 570 nm on an EnVision Multilabel Reader 153

(PerkinElmer). 154

155

Inhibitory potency 156

Vero cell monolayers were seeded in 96-well plates at 4.0 x 103 cells per well 157

and incubated overnight. Dose-response curves were generated by measuring virus-158

induced CPE in the presence of a range of compound concentrations. Eight compound 159

concentrations (25, 8, 2.5, 0.8, 0.25, 0.08, 0.025, and 0.008 µM) were used to generate 160

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

8

inhibition curves suitable for calculating the EC50 from virus-induced CPEs. Compound 161

dilutions were prepared in DMSO prior to addition to the cell culture medium. The final 162

DMSO concentration in all samples was 0.5%. Cell monolayers were infected with 163

DENV-2 at a moi of 0.1. At 5 days post-infection (dpi), the assay was terminated by 164

fixing and staining the cells as described above. The level of CPE was quantified by 165

measuring absorbance at 570 nm. EC50 values were calculated by fitting the data to a 166

four-parameter logistic model (variable-slope, nonlinear regression model) to generate a 167

dose-response curve using XLfit 4.1 (IBDS; Emeryville, CA). From this curve, the 168

concentration of compound that inhibited virus-induced CPE by 50% was calculated. 169

170

Cytotoxicity assays 171

For measurement of compound cytotoxicity, Vero, BSC40, and Huh-7 cells were 172

seeded at either 3.5 x 103 or 1.0 x 104 cells per well in MEM supplemented with 5% FBS 173

in 96-well plates, incubated overnight, and then incubated with various concentrations of 174

ST-148 for 48 hr. Cell viability was measured using a metabolic activity assay where the 175

absorbance at 570/600 nm was measured on an EnVision Multilabel Reader at 5-6 hr 176

post-addition of 470 µM resazurin (Sigma) at 10% of the assay volume and incubation 177

at 37°C with 5% CO2. BHK cells were seeded at 5.0 x 103 cells per well, and the 178

remainder of the procedure was performed as above. MDBK cells were seeded at 5.0 x 179

103 cells per well in MEM supplemented with 2% HS, incubated overnight as above, 180

and then incubated with various concentrations of ST-148 for 96 hr. Cell viability was 181

measured as above. C6/36 cells were seeded at 4.0 x 104 cells per well, incubated 182

overnight at 28°C with 5% CO2, and then incubated with various concentrations of ST-183

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

9

148 for 48 hr at 28°C with 5% CO2. Cell viability was measured as above. For the five 184

day cytotoxicity assay, the assay was performed as above with the following 185

modifications: Vero cells were seeded at 3.5 x 103 cells per well in 96 well plates, and 186

incubated for 5 days after compound addition. Cell viability was measured using the 187

CellTiter-Glo® Luminescent Cell Viability Assay (Promega; Madison, WI) according to 188

manufacturer’s protocol. Luminescence was measured on an EnVision Multilabel 189

Reader at 700 nm with a 0.1 sec integration time. 190

191

AMES assay for genotoxicity 192

To evaluate the mutagenic potential of ST-148, the compound was tested in the 193

Bacterial Reverse Mutation Assay and the Muta-Chromo Plate Assay (EBPI; 194

Mississauga, Canada). All bacterial tester strains were purchased from Molecular 195

Toxicology, Inc. (Boone, NC). For the Bacterial Reverse Mutation Assay, 0.1 ml of 196

overnight cultures of E. coli tester strain WP-2 uvrA were added to molten top agar (with 197

0.5% NaCl, 0.05 mM histidine, and 0.05 mM biotin) in the presence and absence of 198

10% (v/v) Aroclor-1254-induced rat liver S9 (with 0.1 M phosphate buffer pH 7.4, 4 mM 199

NADP, 6 mM G-6-P, 33 mM MgCl2, and 8 mM KCl) (Molecular Toxicology) and various 200

concentrations of ST-148 with appropriate controls. The top agar mixture was overlaid 201

onto minimal glucose agar plates (Vogel-Bonner medium E with 2% glucose) (Molecular 202

Toxicology), allowed to harden, and incubated at 37°C for 48 to 72 hr. After the 203

incubation, any revertant colonies were scored. The Muta-Chromo Plate Assay was 204

carried out according to manufacturer’s instructions using Salmonella typhimurium 205

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

10

tester strains TA-98, TA-100, TA-1535, and TA-1537, Aroclor-1254-induced rat liver S9 206

with co-factors and various concentrations of ST-148. 207

208

Viral titer reduction assays 209

A viral titer reduction assay was performed to measure the antiviral efficacy of 210

ST-148 in cell culture. Cells (Vero for DENV or Modoc virus, L929 for MHV) were 211

seeded at 1 x 105 cells per well in 12-well plates and incubated overnight. Cells were 212

infected with DENV-1, DENV-2, DENV-3, DENV-4, DENV-2 S221, or Modoc virus at a 213

moi of 0.1 in the presence of various concentrations of ST-148 and incubated for 1.5 hr. 214

The inoculum was removed and replaced with MEM supplemented with 2% FBS 215

containing various concentrations of ST-148 and incubated for 48 hr. Supernatant was 216

harvested and serially diluted across Vero (L929 for MHV) cells seeded at 3.0 x 105 217

cells per well in 6-well plates. After a 1.5 hr infection, plates were overlaid with a 1:1 mix 218

of SeaPlaque agarose (Lonza; Allendale, NJ) and 2X MEM. Plates were incubated (3 219

days for MHC, 5 days for Modoc, 7 days for DENV-2 and DENV-4, 8 days for DENV-2 220

S221, and 10 days for DENV-1 and DENV-3). Plates were fixed and stained as 221

described above with the exception of a shorter stain period (10 seconds) to visualize 222

plaques. 223

224

Time-of-drug-addition assay 225

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

11

One day prior to infection, 3 x 104 Vero cells were seeded in 24-well plates and 226

incubated overnight. Cells were then infected with DENV-2 at a moi of 1.0. After 1.5 hr, 227

the virus inoculum was replaced with MEM supplemented with 1% FBS. ST-148 at a 228

concentration of 25 µM was added to the assay medium at either 2 hr prior to infection 229

or at 0 (i.e. at the time of infection), 2, 4, 6, 8, 12, 24, or 48 hpi. At 48 hpi the 230

supernatant was harvested and plaque titrated on Vero cells as described above. 231

232

Antiviral specificity 233

All assays (except for influenza) were carried out in the appropriate medium 234

containing 2% FBS (HS for MDBK cells). Ninety-six well cell culture plates were seeded 235

24 hr prior to use with 2.0 x 104 (Vero for HSV, BSC40 for Sindbis and vvGFP), 1.5 x 236

104 (Vero for DENV-2 K0049) or 1.0 x 104 (MDBK for BVDV, MDCK for influenza) cells 237

per well. Compound was added to duplicate wells of cells at final concentrations of 50, 238

25, 12.5, 6.3, 3.1, 1.6, 0.8, and 0 µM. The final concentration of DMSO in the assays 239

was 0.5%. Cells were infected at the cell culture infectious dose causing > 90% CPE at 240

1.6 (Sindbis), 2 (HSV or vvGFP), 3 (influenza), or 4 (BVDV) dpi or at titers that 241

generated an enzyme-linked immunosorbent assay (ELISA) signal of 2.5-fold over 242

background for DENV-2 K0049 at the end of the incubation period. For the influenza 243

assay, the cell culture medium (EMEM, supplemented with 10% FBS and Penicillin-244

Streptomycin) was replaced with serum free, Ultra MDCK Chemically Defined, Serum-245

free Renal cell medium with L-glutamine (Lonza) containing 5 µg/ml TPCK-treated 246

trypsin (Amersham; Pittsburgh, PA). Influenza virus was added to the culture medium 247

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

12

and virus-induced CPE was measured at 72 hr post-infection. As controls, uninfected 248

cells and cells receiving virus without compound were included on each assay plate. At 249

the end of the incubation period, virus-infected cells were fixed and stained as 250

described above. DENV-2 K0049 ELISA was carried out using a 1:100 dilution of 251

mouse monoclonal antibody to DENV prM glycoprotein (Abcam; Cambridge, MA) and 252

1:100 dilution of goat anti-mouse IgG (H+L)-HRP conjugate (BioRad; Hercules, CA) as 253

a secondary antibody. 254

255

Selection of drug resistant virus variants 256

DENV variants with reduced susceptibility to ST-148 were isolated by plating 257

wild-type DENV-2 on Vero cell monolayers in the presence of 5 µM ST-148. The 258

inoculum was removed after 2 hr incubation, replaced with media containing ST-148 and 259

incubated at 37°C with 5% CO2. At 6 dpi, the supernatant was harvested and applied to 260

Vero cells in the presence of ST-148, and the infection and incubation were repeated as 261

above. Virus isolates exhibiting reduced susceptibility to ST-148 were plaque purified 262

three times in the presence of compound prior to large-scale stock preparation. The 263

purified virus isolate (v148R) was amplified and viral RNA was extracted from v148R 264

using the QIAamp Viral RNA Mini Kit (Qiagen; Valencia, CA). Viral RNA was reverse-265

transcribed into cDNA using the SuperScript III One-Step RT-PCR System with 266

Platinum Taq (Invitrogen). The entire cDNA genome was sequenced at the Oregon 267

State University Center for Genome Research and Biocomputing Core Laboratories 268

(Corvallis, OR) and analyzed for mutations. 269

270

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

13

Plaque reduction assay 271

A plaque reduction assay was performed to compare the level of resistance of 272

v148R and wild-type DENV-2 against ST-148. Vero cells were seeded at 3.0 x 105 cells 273

per well in 6-well plates and incubated overnight. Growth media was aspirated from the 274

plates and cells were infected with DENV-2 or v148R at 100 pfu per well in MEM 275

supplemented with 1% FBS in the presence of 5 µM ST-148 or an equal volume of 276

DMSO. After 1.5 hr incubation, the inoculum was aspirated and the cells were overlaid 277

with a 1:1 mix of SeaPlaque agarose and 2X MEM in the presence of 5 µM ST-148 or 278

an equal volume of DMSO. The plates were incubated for 7 days, and fixed and stained 279

as described above with the exception of a shorter stain period (10 seconds) to 280

visualize plaques. 281

282

Multi-cycle growth curve 283

A multi-cycle growth curve was performed to compare the replication fitness of 284

v148R and wild-type DENV-2. Vero cells were seeded at 3.0 x 104 cells per well in 12-285

well plates and incubated overnight. Growth media was aspirated from the plates and 286

cells were infected with DENV-2 or v148R at a moi of 0.1. After 1.5 hr incubation, the 287

inoculum was removed, replaced with fresh media, and incubated. The supernatant was 288

harvested at various time points and virus levels quantified by plaque assay on Vero 289

cells. 290

291

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

14

Reverse genetics 292

DENV-2 cDNA-Tonga/74 and Escherichia coli BD1528 ung-1 nadB7 (BD1528) 293

cells were provided by Dr. Steve Whitehead at the National Institutes of Health 294

(Bethesda, MD). The S34L mutation identified in v148R was reverse engineered into 295

cDNA and transfected to obtain an infectious virus (vcDNA-148R) to confirm the 296

presence of the resistance-generating mutation. The S34L mutation was generated in 297

DENV-2 cDNA-Tonga/74 using QuikChange II XL Site-Directed Mutagenesis Kit 298

(Agilent Technologies; Santa Clara, CA) and primers DenMut-Cap-S34L-F (5’ TTGACA 299

AAGAGATTCTTACTTGGAATGCTGCAG) and DenMut-Cap-S34L-R (5’ CTGCAGCAT 300

TCCAAGTAAGAATCTCTTTGTCAA) following the manufacturer’s instructions, 301

transformed into competent BD1528 cells and plated on LB plates containing 10 µg/ml 302

tetracycline. Colonies were picked and cultured and cDNA was extracted using the 303

QIAquick MiniPrep Kit (Qiagen). The cDNA was sequenced to confirm the presence of 304

the mutation of interest. The cDNA-S34L was linearized and transcribed with the 305

AmpliCap SP6 High Yield Message Maker Kit (Epicentre; Madison, WI) and purified 306

with RNeasy Mini Kit (Qiagen). In 6-well plates 24 hr prior to infection, Vero cells were 307

seeded at 3.0 x105 cells per well in MEM with 10% FBS. Five hr prior to infection, the 308

medium was removed, washed three times with Dulbecco’s Phosphate Buffered Saline 309

without calcium and magnesium (DPBS) (Cellgro; Manassas, VA), and replaced with 310

Opti-Pro Serum Free Media (Invitrogen). Transcribed RNA (1 µg) diluted in 20 mM 311

HEPES/saline was added to diluted DOTAP Liposomal Transfection Reagent (Roche; 312

Indianapolis, IN) and HEPES/saline in a polystyrene tube, and incubated for 10 min at 313

room temperature. The DOTAP mixture was added to 1 ml of Opti-Pro media on Vero 314

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

15

cells and incubated for 48 hr, after which 2 ml of Opti-Pro was added. After six days, the 315

transfection was serially diluted on Vero cells as above, incubated for 7 days, and then 316

fixed and stained as above to determine the presence of infectious vcDNA-148R viral 317

particles. 318

319

C protein purification and expression 320

The DENV mature capsid domain (position 1 to 101) was amplified by PCR using 321

cDNA-Tonga/74 (2) as a template and primers DenCap (5’phosphorylated- 322

AATAACCAACGGAAAAAGGCGAG) and DenCapRevHindIII 323

(5’TCAGTCAAGCTTCATACAGTTCTACGTCTCCTGTTTAAG). The PCR product was 324

digested with StuI and HindIII and cloned into pQE-30Xa vector (Qiagen). The PCR-325

amplified region was verified by sequencing. Plasmids containing the correct insert were 326

transformed into the E. coli M15 [pREP4] expression system (Qiagen) according to the 327

manufacturer’s protocol. Cultures were incubated at 37°C with shaking until they 328

reached an OD of 0.75, were induced with the addition of IPTG to a final concentration 329

of 1 mM, and incubated at 37°C for 5 hr with shaking. Cultures were harvested by 330

centrifugation at 8,000 x g for 20 minutes, and cell pellets were frozen overnight. Pellets 331

were resuspended in a pH 7.8 binding buffer (20 mM imidazole, 2 mM TCEP, 0.5% 332

CHAPS, 1 M NaCl and 20 mM HEPES with a complete EDTA-free Protease Inhibitor 333

Cocktail Tablet (Roche)), microfluidized, centrifuged at 20,000 x g for 30 minutes, and 334

filtered. Supernatant was passed through a pre-equilibrated 5 ml HisTrap HP column 335

(GE Healthcare; Waukesha, WI) and eluted in the same buffer as above except 336

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

16

containing 500 mM imidazole. Fractions containing histidine-tagged dimerized C protein 337

were frozen at -80°C. 338

339

Intrinsic fluorescence 340

Fluorescence measurements were taken using a Fluorolog-3 fluorescence 341

spectrophotometer (HORIBA Jobin Yvon; Edison, NJ) equipped with a mercury-arc 342

lamp. A scan was performed on C protein samples diluted in DPBS using an excitation 343

wavelength of 295 nm and emission wavelengths from 310 to 380 nm in the presence 344

and absence of compound. C protein was diluted until the peak fluorescence count 345

measured was between 1 and 2 million counts which were not saturating for the 346

detection system. Background DPBS and drug fluorescence measurements were also 347

performed and the signals were subtracted from each sample so that the specific C 348

protein signal was measured in the presence and absence of compound. Samples were 349

prepared in 500 µl volumes for use in a quartz microcuvette. Each experiment was 350

performed in triplicate. Data analysis and collection was performed using DataMax 351

software, version 2.20 installed on a SpectraAcq processor (HORIBA Jobin Yvon). 352

353

In vivo studies 354

All studies involving vertebrate animals were approved by the Institutional Animal 355

Care and Use Committee of Oregon State University, a fully AAALAC-accredited facility, 356

and followed all federal guidelines as outlined in the Guide for the Care and Use of 357

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

17

Laboratory Animals (National Research Council, 2011). Strain 129 (129S2/SvPasCrl) 358

mice were obtained from Charles River Laboratories (Wilmington, MA) and were 359

approximately 6 weeks of age for the pharmacokinetic studies. AG129 mice (IFN-α/β 360

and -γ receptor deficient on the Strain 129 background) were purchased from B & K 361

Universal (Hull, UK) and were 5 to 6 weeks of age for antiviral efficacy studies. 362

363

Pharmacokinetic analysis 364

ST-148 was formulated for oral (p.o.), intraperitoneal (i.p.), and intravenous (i.v.) 365

dosing of mice (Strain 129), as an aqueous solution in 32% hydroxypropyl-beta-366

cyclodextrin (HPβCD) (Roquette; Lestrem, France). ST-148 concentrations were 367

measured in plasma harvested from mice at approximately 0.08, 0.25, 0.5, 1, 2, 3, 4, 6, 368

8, and 24 hr after i.v. administration and 1, 2, 3, 4, 6, 8, and 24 hr after p.o. and i.p. 369

administration. Compound concentrations were measured by liquid chromatography-370

tandem mass spectrometry (LC-MS/MS) using a 3200 Q TRAP LC-MS/MS system 371

(Applied Biosystems / MDS Sciex; Foster City, CA). LC was performed with a 372

Phenomenex Synergi JAX-RP column (4-µm particle size; 50 mm by 2 mm) at a flow 373

rate of 0.5 ml/min with a mobile phase containing 20% acetonitrile, 80% 10 mM 374

ammonium formate, pH 3.5 in water. WinNonlin software (Pharsight; Sunnyvale, CA) 375

was used to estimate pharmacokinetic values. 376

377

Antiviral efficacy in dengue murine model 378

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

18

DENV-2 strain NGC was used in the model and was propagated in C6/36 379

mosquito cells. Five to six-week old female AG129 mice were housed appropriately in 380

labeled cages (4 mice per cage), identified by ear-notching, and weighed prior to any 381

other procedures. Mice were injected i.p. with 0.5 ml of virus suspension in PBS (1.3 x 382

106 pfu per mouse). Mice were dosed immediately post-challenge with either 50 mg/kg 383

of ST-148 or the same volume of vehicle. Weight measurements and clinical 384

observations were made daily. On day 3 post-infection, mice were sacrificed by CO2 385

asphyxiation. Spleen and liver samples were taken from each animal and transferred to 386

pre-weighed homogenization tubes (FastPrep-24 Lysing Matrix C with additional ¼ inch 387

ceramic sphere added [MP Biomedical; Solon, OH]) with 0.5 ml of DPBS. Blood was 388

collected into eppendorf tubes containing sodium citrate for plasma isolation. Plasma 389

viremia and tissue viral load were determined by plaque-titration. Plasma cytokine (IL-6, 390

IL-10, IL-12p70, IFN-γ, TNF-α, and MCP-1) levels were assessed using the BD CBA 391

Mouse Inflammation Kit per the manufacturer’s instructions (BD Biosciences, San Jose, 392

CA). 393

394

Statistical analysis 395

For comparison of viral load in the plasma, spleen, and liver as well as cytokine 396

levels in the plasma among vehicle and once daily (QD) and twice daily (BID) ST-148-397

treated groups, one-way analysis of variance (ANOVA) with a Holm-Sidak correction 398

was used. For comparison of these parameters between vehicle and TID ST-148-399

treated groups, an unpaired Student t test was used and P-values <0.05 were 400

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

19

considered statistically significant. SigmaPlot for Windows Version 11.0 was used for 401

performing all statistical analyses (Systat Software, Inc., San Jose, CA). 402

403

404

405

406

407

408

409

410

411

412

413

414

415

416

417

418

419

420

421

422

423

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

20

RESULTS 424

425

ST-148 is a potent inhibitor of DENV replication in vitro 426

ST-148 (Figure 1) was identified through a high throughput screening (HTS) 427

assay of approximately 200,000 chemically diverse compounds. The HTS assay was 428

developed to identify inhibitors of DENV replication as measured by inhibition of virus-429

induced CPE in a 96-well plate format. ST-148 inhibited DENV-2 in a viral titer reduction 430

assay with an EC50 value (the concentration of compound necessary to inhibit 50% of 431

the virus titer) of 0.016 µM (Table 1) and an EC90 value of 0.125 µM. While ST-148 was 432

active against other dengue serotypes, the compound exhibited a broad range of 433

potencies with EC50 values of 0.512, 1.150, and 2.832 for DENV-3,-4, and -1 434

respectively. Analogs of ST-148, with improved potency, were more active against all 435

four serotypes and the range of EC50 values more narrow, suggesting that these 436

analogs were better optimized to interact with the viral target. ST-148 had potent activity 437

against all four serotypes of DENV in various cells types including Vero, C6/36, Huh-7, 438

BHK, and L929 cells. ST-148 was not cytotoxic in any of the cell lines tested (Table 2). 439

440

ST-148 selectively inhibits flaviviruses 441

To determine the spectrum of antiviral activity, ST-148 potency was measured 442

against a variety of viruses from different virus families. As shown in Table 1 and Table 443

3, ST-148 was active against all DENV serotypes and Modoc virus, a murine flavivirus, 444

but showed less activity against other members of the Flaviviridae family, and did not 445

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

21

effectively inhibit viruses outside the Flaviviridae family. These results indicated that ST-446

148 selectively inhibits DENV within the Flavivirus genus. 447

448

ST-148 is not mutagenic 449

To evaluate the mutagenic potential of ST-148, the compound was tested in the 450

Bacterial Reverse Mutation Assay and the Muta-Chromo Plate Assay using Salmonella 451

typhimurium tester strains TA-98, TA-100, TA-1535, and TA-1537 as well as 452

Escherichia coli tester strain WP-2 uvrA in the presence and absence of Aroclor-453

induced rat liver S9. No mutagenic potential was observed with any of these conditions. 454

455

Time-of-drug-addition studies 456

To investigate which stage of the viral life cycle is affected by ST-148, a time-of-457

drug-addition experiment was conducted where 25 µM of ST-148 was added to DENV-2 458

infected Vero cells either just before the time of infection or at several time points after 459

infection. At 48 hr post-infection, viral yield was quantified by plaque assay. The data 460

indicate that ST-148 inhibits virus replication up to 12 hours after infection, consistent 461

with a post-entry mechanism of action (Figure 2A). 462

463

Isolation of drug-resistant virus variants 464

To identify determinants of ST-148 sensitivity, virus variants resistant to the 465

inhibitory effects of the compound were generated by ten serial passages of virus in the 466

presence of ST-148. Virus capable of growing in the presence of 5 µM ST-148 467

(corresponding to a 312-fold higher EC50) was isolated and plaque-purified three times 468

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

22

in the presence of compound. A plaque reduction assay was conducted to measure the 469

ST-148 sensitivity of this variant (designated v148R) relative to wild-type virus. In the 470

presence of ST-148, plaque formation of the wild-type virus was greatly reduced, both in 471

size and number, while equal numbers of plaques, similar in size to the non-drug 472

treated samples, were observed for v148R in the presence of compound (Figure 2B). 473

The plaques are visualized by crystal violet staining with the stain in contact with the 474

monolayer for a very short duration (just a few seconds). The cytopathic effects 475

resulting from a dengue virus replication appear to enhance dye binding relative to the 476

uninfected monolayer resulting in plaques binding more dye and staining darker. This 477

rapid staining technique is used to visualize plaques formed by many types of virus that 478

fail to form obvious plaques on cell monolayers. In multi-cycle growth experiments the 479

replication rate and peak titer of v148R in the absence of compound was similar to wild-480

type virus suggesting that mutations affecting drug susceptibility had no measurable 481

effect on viral fitness (Figure 2C), at least in vitro. A viral titer reduction assay was used 482

to determine the level of susceptibility of the resistant virus to ST-148. The EC50 for 483

inhibition of the resistant variant (8.92 µM) was over 550-fold higher than the EC50 for 484

inhibition of wild-type DENV2 as determined in Vero cells (0.016 µM) (Table 1). 485

486

ST-148 resistance maps to the capsid coding region 487

Genomic RNA from plaque-purified resistant virus was isolated and amplified by 488

reverse transcription PCR to generate complementary DNA. The nucleotide sequence 489

of overlapping segments of the cDNA from the resistant variant was determined and 490

compared to wild-type DENV sequence to identify the changes in the viral genome that 491

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

23

correlated with reduced compound susceptibility. A single nucleotide change (nucleotide 492

101 in the capsid gene, cytosine to thymine) was found in the resistant virus genome 493

that resulted in a predicted serine (Ser) to leucine (Leu) change at amino acid position 494

34 in the capsid gene (Figure 3). Each of the DENV serotypes, but not Modoc virus, has 495

a serine at position 34. This change was reengineered into wild-type DENV-2 cDNA in 496

order to confirm that the single mutation found was responsible for conferring resistance 497

to ST-148. Infectious virus recovered from the engineered cDNA clone (vcDNA-148R) 498

showed reduced susceptibility to ST-148 with an EC50 for inhibition of 8.68 µM, similar 499

to the parental resistant isolate (Table 1). These results support the conclusion that the 500

S34L mutation is sufficient to confer resistance to ST-148. 501

502

ST-148 interacts with C protein 503

Compounds that bind proteins can alter the dielectric properties of aromatic 504

amino acid residues causing a change in the intrinsic fluorescence of the protein. To 505

determine if ST-148 interacts with C protein, its intrinsic fluorescence was measured 506

from 310 to 380 nm in the presence and absence of ST-148. In the presence of ST-148, 507

a dose-dependent decrease in the intrinsic fluorescence was observed consistent with a 508

direct interaction of the compound with C protein (Figure 4A). As controls, the intrinsic 509

protein fluorescence was measured in the presence of DMSO only, or in the presence 510

of ST-610, a DENV inhibitor compound discovered at SIGA that has a different chemical 511

structure and targets the helicase enzyme (manuscript in preparation) (Figure 1 and 512

4C). Neither DMSO nor ST-610 had a substantial effect on the intrinsic protein 513

fluorescence suggesting that the effects on fluorescence induced by ST-148 were 514

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

24

specific. Similar results were observed with a C protein variant isolated from ST-148-515

resistant virus (Figure 4B and D). These results suggest that ST-148 interacts with C 516

protein but amino acid changes that correlate with reduced compound susceptibility do 517

not alter this interaction. 518

519

Pharmacokinetic analyses and tolerability 520

The pharmacokinetic parameters of ST-148 in mice were determined after 521

formulating the compound as an aqueous solution in 32% hydroxypropyl-beta-522

cyclodextrin (HPβCD). Oral or intraperitoneal administration of ST-148 at 50 mg/kg/day 523

in Strain129 mice generated peak plasma concentrations of 3,138 or 52,283 ng/ml (7.44 524

or 124.04 µM) respectively (Table 4), which are 468- and 7,750-fold above the in vitro 525

EC50 value. Intravenous administration of ST-148 at 20 mg/kg/day generated a peak 526

plasma concentration of 41,133 ng/ml (97.58 µM), 6,093-fold above the EC50. The area 527

under the plasma concentration-time curve (AUC) was greatest in mice administered 528

ST-148 via the i.p. route. The absolute oral bioavailability of ST-148 was 9.1%. The 529

compound was rapidly absorbed after oral dosing with the time to maximum plasma 530

concentration measured at 1.0 hr. Intraperitoneal administration produced higher 531

plasma concentrations with rapid absorption, and peak plasma drug concentrations at 532

1.0 hr after dosing. There was no overt toxicity, or changes in physical signs including 533

body weights, observed in mice after repeated oral or i.p. ST-148 administration of 50 534

mg/kg/day for 3 days. These data indicate that ST-148 has limited oral bioavailability 535

and fairly rapid clearance, but good systemic availability following i.p. administration. 536

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

25

537

In vivo antiviral efficacy 538

A murine viremia model (14, 15, 25) using AG129 mice, which are deficient for 539

IFN-α/β and -γ receptors, was used to evaluate the in vivo efficacy of ST-148. Infection 540

of AG129 mice with DENV-2 (strain NGC) via intraperitoneal inoculation with 1.3 x 106 541

pfu produces a non-lethal infection with peak of viremia occurring on day 3 post-542

infection and an elevation in cytokine levels that are characteristic of DF. Immediately 543

after infection, ST-148 or vehicle at 50 mg/kg/dose was administered either once (QD) 544

or twice daily (BID) for three days. There was no additional weight changes in the ST-545

148 treated mice compared to vehicle-treated mice during the course of the experiment. 546

On average, BID treatment with ST-148 reduced peak plasma viremia 52-fold and 547

reduced viral load in the spleen and liver 3- and 20-fold respectively (Figure 5A). This 548

difference in viremia relative to vehicle-treated mice was statistically significant as 549

determined by one-way ANOVA (Table 5). An increase in the level of inflammatory 550

cytokines has been observed in the blood of patients who have DF (13) with a 551

contributing role in vascular leakage and disease severity. In the AG129 model, levels 552

of TNFα, IL-6, MCP-1, and IFN-γ are elevated, and a trend towards lower levels was 553

observed in ST-148 BID mice (Figure 5B and C), which was not statistically significant. 554

We further tested whether ST-148 was active in this model when administered 555

orally. Due to ST-148 p.o. pharmacokinetics that results in plasma levels of ST-148 556

above the in vitro EC90 only for approximately eight hours after administration, the 557

compound was administered three times per day (TID) at 8-hr intervals at 50 558

mg/kg/dose for three consecutive days. On average, treatment with ST-148 TID 559

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

26

reduced peak viremia 3-fold and reduced viral load in the spleen and liver 7- and 9-fold 560

respectively (Figure 6A). This difference in viremia relative to vehicle-treated mice was 561

statistically significant as determined by one-way ANOVA (Table 6). ST-148 562

administered orally showed a trend toward reduced levels of TNF-α, MCP-1, and IL-6, 563

but not IFN-γ, which were not statistically significant (Figure 6B and C). 564

Taken together, these results illustrate that ST-148 can decrease viremia, viral 565

load in tissues, and inflammatory cytokine levels when administered either i.p or orally. 566

567

DISCUSSION 568

A high throughput screen was conducted to identify inhibitors of DENV-induced 569

cytopathic effects. Characterization of compounds from this screen identified a chemical 570

series that inhibited replication of all four DENV serotypes. The lead compound in this 571

series, ST-148, exhibited strong DENV-2 antiviral activity with an EC50 of 16 nM and a 572

CC50 value of > 50 µM in several transformed cell types. ST-148 was inactive against 573

Japanese encephalitis virus and BVDV, but showed substantial activity against Modoc 574

virus (EC50 = 0.36 µM) and weak activity against YFV (EC50 = 6.97 µM) and HCV (EC50 575

= 39.2 µM). The weak activity against HCV was likely non-specific since this assay was 576

conducted using a subgenomic replicon system that lacks C protein. ST-148 was not 577

active against representative viruses from other virus families. The compound was not 578

genotoxic in Ames tests and was well tolerated in mice but exhibited modest oral 579

bioavailability (9.1%). Administration of ST-148 to mice, lacking interferon receptors, via 580

either oral gavage or intraperitoneal injection at 50 mg/kg reduced the viremia and viral 581

load in the liver and spleen by 1 log, suggesting that the compound is active in vivo. 582

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

27

DENV variants with reduced susceptibility to ST-148 were isolated and 583

compound susceptibility determinants mapped to the region of the genome encoding 584

the C protein. An amino acid substitution from serine to leucine at position 34 of the C 585

protein correlated with a loss of compound susceptibility. These results suggest that ST-586

148 interacts with the C protein to inhibit DENV replication. 587

A direct interaction of ST-148 with the C protein was measured by compound-588

induced effects on the intrinsic protein fluorescence. The reduction in intrinsic 589

fluorescence of C protein in the presence of ST-148 was not observed in the presence 590

of an unrelated compound, ST-610, that also inhibits DENV replication but targets the 591

helicase protein. While these data suggest a specific interaction of ST-148 with the C 592

protein, similar results were observed using a mutant C protein that contained the S34L 593

change associated with reduced compound susceptibility. Furthermore, ST-148 showed 594

substantial antiviral activity against Modoc virus, a natural flavivirus pathogen of mice, 595

and yellow fever virus. A comparison of the amino acid residues of DENV C protein with 596

selected flaviviruses including Modoc virus and yellow fever virus did not provide an 597

obvious explanation as to why these viruses would be susceptible to ST-148 while other 598

flaviviruses were not. It is possible that the interaction of ST-148 with the mutant C 599

protein may induce a conformational change that blocks the normal function of the C 600

protein, but not its intrinsic fluorescence properties. It’s also possible that ST-148 601

interacts with another region of the protein to affect intrinsic protein fluorescence and 602

the S34L amino acid change prevents inhibition by ST-148 without blocking this 603

interaction. Taken together, these data suggest that ST-148 interacts with the C protein 604

and blocks an essential activity required for virus replication. 605

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

28

Time-of-drug-addition experiments suggest that ST-148 is maximally effective 606

when added before or at the time of infection (Figure 2A), which might suggest inhibition 607

of uncoating which is consistent with an earlier report on the entry kinetics of DENV 608

(27). Given that the level of DENV released from infected cells is still somewhat reduced 609

when ST-148 is added up to 24 hrs after infection, it is also possible that ST-148 610

interaction with the C protein may have additional effect on later stages of viral 611

morphogenesis, such as nucleocapsid assembly. Alternatively, the assay may be 612

measuring virus spread, as an inhibition event at an early stage may be amplified 613

because of multiple rounds of virus infection and spread. But as it is likely the C protein 614

has multiple functions in uncoating, replication and assembly of progeny virions, more 615

work is needed to understand the overall effect of ST-148. 616

The mature form of the C protein is a basic, 12 kDa protein that forms 617

homodimers in solution consisting of four alpha helices (α1 to α4). They are arranged in 618

the dimer to produce a concave shaped molecule (17). The floor of the C protein dimer 619

is composed of the highly basic α4- α4’ helices which are thought to interact with RNA. 620

At the top of the concave structure, the α2−α2′ and α1−α1′ have been proposed to 621

interact with membranes (17). Based upon this structure, it has been proposed that 622

positively charged residues on α1-α1’ are involved in membrane interactions (17). The 623

amino acid change (S34L) associated with compound susceptibility resides at the end 624

of α1−α1’ (Figure 3B). Perhaps ST-148 interacts with this region of the protein and 625

prevents membrane association or other essential functions required for viral 626

replication. Although the C protein is the least conserved of the flavivirus proteins, the 627

structural properties are very similar and the charge distribution is well conserved. 628

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

29

The C protein binds RNA consistent with a proposed role in encapsidation of 629

newly replicated RNA into virions. A fraction of the cytoplasmic C protein in DENV-630

infected cells accumulates on the surface of ER-derived organelles named lipid droplets 631

(23). A similar phenotype was observed in cell lines expressing HCV C protein (1). 632

Substitutions of amino acid L50 or L54 in the DENV C protein disrupted lipid droplet 633

targeting and impaired viral particle formation. Likewise, mutations that disrupt lipid 634

droplet association of HCV C protein reduced particle formation (24). Moreover, one 635

study proposed that DENV infection increased the number of lipid droplets per cell (23). 636

Taken together, these results suggest that there is a link between lipid droplet 637

metabolism and viral replication (23). It is possible that ST-148 alters the interaction 638

between lipid droplets and the C protein, thereby inhibiting viral replication. While the 639

S34L change did not appear to alter interaction between C protein and ST-148 based 640

upon the results of the intrinsic protein fluorescence experiments, this amino acid 641

change could bypass the inhibitory effects of the compound by facilitating membrane 642

association. Future studies are ongoing to elucidate the mechanism of ST-148 643

inhibition. 644

645

646

647

648

649

650

651

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

30

ACKNOWLEDGEMENTS 652

This work was supported by NIH grants 1R43AI079937-01A1 and 1 653

R01A1093356-01. R. Bartenschlager is supported by grants from the European Union 654

(Silver (grant no. 260644) and Euvirna (grant no. 264286)). This work was performed by 655

employees of SIGA Technologies incorporated who hold stock in the company. We 656

would like to thank Dr. Steve Whitehead for the cDNA, Dr. Sujan Shresta for the S221 657

virus, Dr. Rebecca Rico-Hesse for the K0049 virus, and Dr. Johan Neyts for HCV 658

specificity testing. We would also like to thank Dr. Kady Honeychurch for DENV-specific 659

primer design, Andrew Wieczorek and Thuan Tran for HTS support, Candace Lovejoy 660

for project management, and Dr. Yali Chen for carrying out the AMES assay. Thanks to 661

Dr. Sean Amberg for critical review of the manuscript. 662

663

664

665

666

667

668

669

670

671

672

673

674

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

31

REFERENCES 675

676

1. Barba, G., F. Harper, T. Harada, M. Kohara, S. Goulinet, Y. Matsuura, G. Eder, Z. 677

Schaff, M. J. Chapman, T. Miyamura, and C. Brechot. 1997. Hepatitis C virus core 678

protein shows a cytoplasmic localization and associates to cellular lipid storage droplets. 679

Proc Natl Acad Sci U S A 94:1200-5. 680

2. Blaney, J. E., Jr., C. T. Hanson, K. A. Hanley, B. R. Murphy, and S. S. Whitehead. 681

2004. Vaccine candidates derived from a novel infectious cDNA clone of an American 682

genotype dengue virus type 2. BMC Infect Dis 4:39. 683

3. Byrd, C. M., T. C. Bolken, A. M. Mjalli, M. N. Arimilli, R. C. Andrews, R. Rothlein, T. 684

Andrea, M. Rao, K. L. Owens, and D. E. Hruby. 2004. New class of orthopoxvirus 685

antiviral drugs that block viral maturation. J Virol 78:12147-56. 686

4. Chang, J., W. Schul, T. D. Butters, A. Yip, B. Liu, A. Goh, S. B. Lakshminarayana, 687

D. Alonzi, G. Reinkensmeier, X. Pan, X. Qu, J. M. Weidner, L. Wang, W. Yu, N. 688

Borune, M. A. Kinch, J. E. Rayahin, R. Moriarty, X. Xu, P. Y. Shi, J. T. Guo, and T. 689

M. Block. 2011. Combination of alpha-glucosidase inhibitor and ribavirin for the 690

treatment of dengue virus infection in vitro and in vivo. Antiviral Res 89:26-34. 691

5. Chang, J., W. Schul, A. Yip, X. Xu, J. T. Guo, and T. M. Block. 2011. Competitive 692

inhibitor of cellular alpha-glucosidases protects mice from lethal dengue virus infection. 693

Antiviral Res 92:369-71. 694

6. Chen, Y. L., Z. Yin, J. Duraiswamy, W. Schul, C. C. Lim, B. Liu, H. Y. Xu, M. Qing, A. 695

Yip, G. Wang, W. L. Chan, H. P. Tan, M. Lo, S. Liung, R. R. Kondreddi, R. Rao, H. 696

Gu, H. He, T. H. Keller, and P. Y. Shi. 2010. Inhibition of dengue virus RNA synthesis 697

by an adenosine nucleoside. Antimicrob Agents Chemother 54:2932-9. 698

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

32

7. Cleaves, G. R., T. E. Ryan, and R. W. Schlesinger. 1981. Identification and 699

characterization of type 2 dengue virus replicative intermediate and replicative form 700

RNAs. Virology 111:73-83. 701

8. Courageot, M. P., M. P. Frenkiel, C. D. Dos Santos, V. Deubel, and P. Despres. 702

2000. Alpha-glucosidase inhibitors reduce dengue virus production by affecting the initial 703

steps of virion morphogenesis in the endoplasmic reticulum. J Virol 74:564-72. 704

9. Deen, J. L., E. Harris, B. Wills, A. Balmaseda, S. N. Hammond, C. Rocha, N. M. 705

Dung, N. T. Hung, T. T. Hien, and J. J. Farrar. 2006. The WHO dengue classification 706

and case definitions: time for a reassessment. Lancet 368:170-3. 707

10. Dejnirattisai, W., A. Jumnainsong, N. Onsirisakul, P. Fitton, S. Vasanawathana, W. 708

Limpitikul, C. Puttikhunt, C. Edwards, T. Duangchinda, S. Supasa, K. 709

Chawansuntati, P. Malasit, J. Mongkolsapaya, and G. Screaton. 2010. Cross-710

reacting antibodies enhance dengue virus infection in humans. Science 328:745-8. 711

11. Gubler, D. J. 2002. Epidemic dengue/dengue hemorrhagic fever as a public health, 712

social and economic problem in the 21st century. Trends Microbiol 10:100-3. 713

12. Halstead, S. B. 2007. Dengue. Lancet 370:1644-52. 714

13. Halstead, S. B. 1988. Pathogenesis of dengue: challenges to molecular biology. 715

Science 239:476-81. 716

14. Johnson, A. J., and J. T. Roehrig. 1999. New mouse model for dengue virus vaccine 717

testing. J Virol 73:783-6. 718

15. Kyle, J. L., P. R. Beatty, and E. Harris. 2007. Dengue virus infects macrophages and 719

dendritic cells in a mouse model of infection. J Infect Dis 195:1808-17. 720

16. Lim, S. P., L. S. Sonntag, C. Noble, S. H. Nilar, R. H. Ng, G. Zou, P. Monaghan, K. Y. 721

Chung, H. Dong, B. Liu, C. Bodenreider, G. Lee, M. Ding, W. L. Chan, G. Wang, Y. 722

L. Jian, A. T. Chao, J. Lescar, Z. Yin, T. R. Vedananda, T. H. Keller, and P. Y. Shi. 723

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

33

2011. Small molecule inhibitors that selectively block dengue virus methyltransferase. J 724

Biol Chem 286:6233-40. 725

17. Ma, L., C. T. Jones, T. D. Groesch, R. J. Kuhn, and C. B. Post. 2004. Solution 726

structure of dengue virus capsid protein reveals another fold. Proc Natl Acad Sci U S A 727

101:3414-9. 728

18. Monath, T. P. 1994. Dengue: the risk to developed and developing countries. Proc Natl 729

Acad Sci U S A 91:2395-400. 730

19. Niyomrattanakit, P., Y. L. Chen, H. Dong, Z. Yin, M. Qing, J. F. Glickman, K. Lin, D. 731

Mueller, H. Voshol, J. Y. Lim, S. Nilar, T. H. Keller, and P. Y. Shi. 2010. Inhibition of 732

dengue virus polymerase by blocking of the RNA tunnel. J Virol 84:5678-86. 733

20. Noble, C. G., Y. L. Chen, H. Dong, F. Gu, S. P. Lim, W. Schul, Q. Y. Wang, and P. Y. 734

Shi. 2010. Strategies for development of Dengue virus inhibitors. Antiviral Res 85:450-735

62. 736

21. Poh, M. K., G. Shui, X. Xie, P. Y. Shi, M. R. Wenk, and F. Gu. 2012. U18666A, an 737

intra-cellular cholesterol transport inhibitor, inhibits dengue virus entry and replication. 738

Antiviral Res 93:191-8. 739

22. Poh, M. K., A. Yip, S. Zhang, J. P. Priestle, N. L. Ma, J. M. Smit, J. Wilschut, P. Y. 740

Shi, M. R. Wenk, and W. Schul. 2009. A small molecule fusion inhibitor of dengue 741

virus. Antiviral Res 84:260-6. 742

23. Samsa, M. M., J. A. Mondotte, N. G. Iglesias, I. Assuncao-Miranda, G. Barbosa-743

Lima, A. T. Da Poian, P. T. Bozza, and A. V. Gamarnik. 2009. Dengue virus capsid 744

protein usurps lipid droplets for viral particle formation. PLoS Pathog 5:e1000632. 745

24. Shavinskaya, A., S. Boulant, F. Penin, J. McLauchlan, and R. Bartenschlager. 2007. 746

The lipid droplet binding domain of hepatitis C virus core protein is a major determinant 747

for efficient virus assembly. J Biol Chem 282:37158-69. 748

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

34

25. Shresta, S., J. L. Kyle, H. M. Snider, M. Basavapatna, P. R. Beatty, and E. Harris. 749

2004. Interferon-dependent immunity is essential for resistance to primary dengue virus 750

infection in mice, whereas T- and B-cell-dependent immunity are less critical. J Virol 751

78:2701-10. 752

26. Steuer, C., C. Gege, W. Fischl, K. H. Heinonen, R. Bartenschlager, and C. D. Klein. 753

2011. Synthesis and biological evaluation of alpha-ketoamides as inhibitors of the 754

Dengue virus protease with antiviral activity in cell-culture. Bioorg Med Chem 19:4067-755

74. 756

27. van der Schaar, H. M., M. J. Rust, C. Chen, H. van der Ende-Metselaar, J. Wilschut, 757

X. Zhuang, and J. M. Smit. 2008. Dissecting the cell entry pathway of dengue virus by 758

single-particle tracking in living cells. PLoS Pathog 4:e1000244. 759

28. Vaughn, D. W., S. Green, S. Kalayanarooj, B. L. Innis, S. Nimmannitya, S. 760

Suntayakorn, T. P. Endy, B. Raengsakulrach, A. L. Rothman, F. A. Ennis, and A. 761

Nisalak. 2000. Dengue viremia titer, antibody response pattern, and virus serotype 762

correlate with disease severity. J Infect Dis 181:2-9. 763

29. Vrolijk, J. M., A. Kaul, B. E. Hansen, V. Lohmann, B. L. Haagmans, S. W. Schalm, 764

and R. Bartenschlager. 2003. A replicon-based bioassay for the measurement of 765

interferons in patients with chronic hepatitis C. J Virol Methods 110:201-9. 766

30. Wang, Q. Y., R. R. Kondreddi, X. Xie, R. Rao, S. Nilar, H. Y. Xu, M. Qing, D. Chang, 767

H. Dong, F. Yokokawa, S. B. Lakshminarayana, A. Goh, W. Schul, L. Kramer, T. H. 768

Keller, and P. Y. Shi. 2011. A translation inhibitor that suppresses dengue virus in vitro 769

and in vivo. Antimicrob Agents Chemother 55:4072-80. 770

31. Welsch, S., S. Miller, I. Romero-Brey, A. Merz, C. K. Bleck, P. Walther, S. D. Fuller, 771

C. Antony, J. Krijnse-Locker, and R. Bartenschlager. 2009. Composition and three-772

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

35

dimensional architecture of the dengue virus replication and assembly sites. Cell Host 773

Microbe 5:365-75. 774

32. Whitby, K., T. C. Pierson, B. Geiss, K. Lane, M. Engle, Y. Zhou, R. W. Doms, and M. 775

S. Diamond. 2005. Castanospermine, a potent inhibitor of dengue virus infection in vitro 776

and in vivo. J Virol 79:8698-706. 777

33. Xie, X., Q. Y. Wang, H. Y. Xu, M. Qing, L. Kramer, Z. Yuan, and P. Y. Shi. 2011. 778

Inhibition of dengue virus by targeting viral NS4B protein. J Virol 85:11183-95. 779

34. Yu, I. M., W. Zhang, H. A. Holdaway, L. Li, V. A. Kostyuchenko, P. R. Chipman, R. J. 780

Kuhn, M. G. Rossmann, and J. Chen. 2008. Structure of the immature dengue virus at 781

low pH primes proteolytic maturation. Science 319:1834-7. 782

35. Zhang, Y., J. Corver, P. R. Chipman, W. Zhang, S. V. Pletnev, D. Sedlak, T. S. 783

Baker, J. H. Strauss, R. J. Kuhn, and M. G. Rossmann. 2003. Structures of immature 784

flavivirus particles. Embo J 22:2604-13. 785

36. Zhang, Y., B. Kaufmann, P. R. Chipman, R. J. Kuhn, and M. G. Rossmann. 2007. 786

Structure of immature West Nile virus. J Virol 81:6141-5. 787

788

789

790

791

792

793

794

795

796

797

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

36

LEGENDS TO FIGURES 798

Figure 1. Structures of ST-148 and ST-610. 799

800

Figure 2. (A) Time-of-drug-addition study. DENV-2 infected Vero cells were treated with 801

25 µM ST-148 at various time points and virus levels quantified by plaque assay at 48 802

hpi. The data are mean values ± SD for three independent experiments. (B) Plaque 803

formation of wild-type DENV and ST-148-resistant virus in the presence and absence of 804

ST-148. Vero cell monolayers were infected with DENV-2 or v148R in the presence and 805

absence of 5 µM ST-148 and covered with an agarose overlay. At 7 days post-infection 806

the cultures were fixed in 5% glutaraldehyde and stained with crystal violet. (C) Multi-807

cycle growth curve of wild-type DENV and ST-148-resistant virus in the absence of ST-808

148. Vero cell monolayers were infected with virus, harvested at various time points and 809

plaque titrated. The data are mean values ± SD for two observations. 810

811

Figure 3. (A) A sequence comparison of capsid orthologs. Graphic representation of 812

the DENV genome, illustrating the location of resistance to ST-148 (capsid S34L) and 813

an amino acid sequence comparison of orthologs from the capsid region of other 814

flaviviruses showing the degree of sequence conservation. (B) Structure of the 815

homodimer DENV-2 C protein. Subunit A – light blue, subunit B – dark blue. The 816

location of the serine residue at position 34 is indicated in red for both chains. Protein 817

structure modified using CN3D software (17). 818

819

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

37

Figure 4. The effect of ST-148 on the intrinsic fluorescence of C protein. Intrinsic 820

fluorescence measurements in arbitrary units (a.u.) were made by combining protein 821

and compound, and reading absorbance at an excitation of 295 nm and scanning 822

emissions between 310 and 380 nm. (A) C protein in the presence of ST-148, (B) ST-823

148-resistant mutant C protein (S34L) with ST-148, (C) C protein with ST-610, and (D) 824

mutant C protein with ST-610. 825

826

Figure 5. In vivo efficacy of ST-148 delivered i.p. AG129 mice (8 mice/ treatment 827

group) were infected with 1.3 x 106 pfu DENV-2 (strain NGC) i.p. on day 0. The mice 828

were treated with 50 mg/kg ST-148 i.p. either once (QD) or twice per day (BID). On day 829

3 post-infection (the peak of viremia), the viral load and viremia were quantified by 830

plaque assay. (A) Viral load and viremia. White symbols represent vehicle treated mice, 831

black symbols represent mice treated with ST-148 QD, and gray symbols represent 832

mice treated with ST-148 BID. Circles represent spleen titer, triangles represent liver 833

titer, and squares represent plasma titer. Solid bars represent the arithmetic mean. (B 834

and C) Inflammatory cytokine levels in the plasma. The lower limit of quantification for 835

the CBA assay is 20 pg/ml. The data are mean values ± SD for three observations. 836

837

Figure 6. Oral In vivo efficacy of ST-148. AG129 mice (8 mice/treatment group) were 838

infected with 9 x 106 pfu DENV-2 (strain NGC) i.p. on day 0. The mice were treated with 839

50 mg/kg ST-148 p.o. three times per day (TID). On day 3 post-infection (the peak of 840

viremia), the viral load and viremia were quantified by plaque assay. (A) Viral load and 841

viremia, open symbols represents vehicle-treated mice, filled symbols represent ST-842

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

38

148-treated mice. Circles represent spleen titer, triangles represent liver titer, and 843

squares represent plasma titer. Solid bars represent the arithmetic mean. (B and C) 844

Inflammatory cytokine levels in the plasma. The lower limit of quantification for the CBA 845

assay is 20 pg/ml. The data are mean values ± SD for three observations. 846

847

848

849

850

851

852

853

854

855

856

857

858

859

860

861

862

863

864

865

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

39

Table 1. Antiviral activity of ST-148 against DENV 866

Cell Type Virus EC50(µM)a Vero DENV-1 2.832 ± 1.13 DENV-2 0.016 ± 0.01 DENV-3 0.512 ± 0.42 DENV-4 1.150 ± 0.14 DENV-2 K0049 0.551 ± 0.40 DENV-2 S221 0.023 ± 0.01 v148R 8.918 ± 0.80 vcDNA-148R 8.684 ± 0.75 C6/36 DENV-2 0.039 ± 0.01 Huh-7 DENV-2 0.012 ± 0.01 BHK DENV-2 0.073 ± 0.08 L929 DENV-2 0.016 ± 0.01

aCells were infected with indicated viruses at a moi of 0.1 and treated with ST-148. 867

Culture medium was collected at 48 h post-infection and the viral titer determined by 868

plaque assay. For K0049, activity was measured by ELISA assay. v148R is the tissue 869

culture passaged, drug-resistant DENV-2 isolate, and vcDNA-148R is the virus 870

recovered from the engineered cDNA clone. The data are mean values ± SD for three 871

independent experiments. 872

873

874

875

876

877

878

879

880

881

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

40

Table 2. Cell toxicity of ST-148 882

Cell Type CC50 (µM)a Vero (2d) >100 Vero (5d) >100 BSC40 >100 C6/36 >100 Huh-7 >100 BHK >100 MDBK >100 L929 >50

aCells were incubated with ST-148 at a range of concentrations for 48 hr (also 5 days 883

for Vero; 96 hr for MDBK cells). Cell viability was measured using a resazurin assay 884

where the absorbance was measured on an EnVision Multilabel Reader at 5 hr post-885

addition of resazurin and incubation at 37°C. For the 5 day assay, viability was 886

measured using Cell Titer-Glo Luminescent Cell Viability Assay. The data are mean 887

values for three independent experiments. 888

889

890

891

892

893

894

895

896

897

898

899

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

41

Table 3. ST-148 antiviral spectrum of activity and selectivity 900

Virus Family Genus Group EC50

(µM) Modoc virus Flaviviridae Flavivirus +ssRNA 0.36 Yellow fever virusa Flaviviridae Flavivirus +ssRNA 6.97 Japanese encephalitis virusa Flaviviridae Flavivirus +ssRNA >50 Bovine viral diarrhea virus Flaviviridae Pestivirus +ssRNA >50 Hepatitis C virusb Flaviviridae Hepacivirus +ssRNA 39.2 Sindbis virus Togaviridae Alphavirus +ssRNA >50 Influenza A virus Orthomyxoviridae InfluenzavirusA -ssRNA >25 Vaccinia virus Poxviridae Orthopoxvirus dsDNA >50 Herpes simplex virus 1 Herpesviridae Simplexvirus dsDNA >50 a: In vitro testing done by Integrated Biotherapeutics via a CPE based assay in Vero 901

cells. b:In vitro testing done by Dr. Neyts laboratory at the Rega Institute using a 902

subgenomic HCV replicon (genotype 1b) in Huh 5.2 cells(29). 903

904

905

906

907

908

909

910

911

912

913

914

915

916

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

42

Table 4. Pharmacokinetic parameters of ST-148 in Strain 129 mice 917

Route Dosage (mg/kg)

Tmax

(hr) Cmax (ng/ml)

AUC (hr*ng/ml)

p.o. 50 1.0 3138 5826 i.p. 50 1.0 52283 77011 i.v. 20 0.1 41133 25523 Plasma drug concentrations were measured by LC-MS/MS over a 24-hr time period. 918

Tmax, time of maximal concentration; Cmax, maximal concentration. 919

920

921

922

923

924

925

926

927

928

929

930

931

932

933

934

935

936

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

43

Table 5. Viral titer from mice treated with ST-148 i.p. at 50mg/kg/dose. 937

Treatment

Organ Mean Titer (pfu/g tissue or pfu/mL)

Standard Deviation

P-value vs. vehicle

Vehicle Spleen 1.08 x 107 6.48 x 106 -

Liver 1.21 x 105 1.23 x 105 -

Plasma 1.52 x 105 7.19 x 104 -

ST-148 QD Spleen 3.16 x 106 3.47 x 106 0.002

Liver 1.21 x 104 6.83 x 103 0.004

Plasma 9.20 x 103 6.81 x 103 <0.001

ST-148 BID Spleen 3.59 x 106 2.14 x 106 0.004

Liver 6.12 x 103 4.06 x 103 0.006

Plasma 2.93 x 103 8.33 x 102 <0.001

938

939

940

941

942

943

944

945

946

947

948

949

950

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

44

Table 6. Viral titer from mice treated with ST-148 p.o. at 50mg/kg/dose. 951

Treatment Organ Mean Titer (pfu/g tissue or pfu/mL)

Standard Deviation

P-value vs. vehicle

Vehicle Spleen 6.08 x 106 3.16 x 106 -

Liver 3.98 x 105 4.56 x 105 -

Plasma 1.95 x 106 1.39 x 106 -

ST-148 TID Spleen 8.20 x 105 4.35 x 105 <0.001

Liver 4.22 x 104 4.11 x 104 0.046

Plasma 6.65 x 105 5.44 x 105 0.029

952

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

SN

NN

N

ON

N

O

NN S

O

O N

ST-610

SS

NN

ST-148

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

1 E+03

1.E+04

1.E+05

1.E+06

1.E+07

(pfu

/mL)

A

1.E+00

1.E+01

1.E+02

1.E+03

-6 0 6 12 18 24 30 36 42 48

Tite

r (

Time of addition (hpi)

DENV-2 no drugDENV-2 + ST-148

Time of addition (hpi)

1.E+07

WT Den2 v148R

0 M

B C

1 E+02

1.E+03

1.E+04

1.E+05

1.E+06

Tite

r (pf

u/m

L)

DENV-2

0 µMST-148

1.E+00

1.E+01

1.E+02

0 24 48 72 96 120 144 168Hours post infection (hpi)

v148R

5 µMST-148

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

5’ UTR 3’ UTRC prM E NS1 NS2A NS2B NS3 NS4A NS4B NS5

Capsid aa1 114

A B

NRVSTVQQLT KRFLLGMLQG RGP-LKLFMANRVSTVQQLT KRFSLGMLQG RGP-LKLFMANRVSTVQQLT KRFSLGMLQG RGP-LKLFMA

Capsid aa1-114

S34L

v148R DEN2 NGC DEN2 cDNA

1 NRVSTVSQLA KRFSKGLLSG QGP-MKLVMANRVSTGSQLA KRFSRGLLNG QGP-MKLVMANRVSTPQGLV KRFSTGLFSG KGP-LRMVLA

KKKKNAGRNG KEVPGLALVM GV------IHRLERGKMKIV PKESEKDSKT KPPDATIVVEQDVKFPGGGQ IVGGVYLLPR RGPRLGVRAT

DEN1 TH-Sman DEN3 H87 DEN4 H241

MODOC M544 BVDV NADL HCV 1b/DK1 Q Q

PRVLSLIGLK RAMLS-LIDG KGP-IRFVLA--VRSLSNKI KQKTKQIGNR PGP-SRGVQG

/

WNV KN3829 YFV Uga48

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

1400000

1600000

1800000

(a.u

.)

WTCapsidST-148, 500nMST-148, 2500nM

1400000

1600000

1800000

a.u.

)

WTCapsidDMSOST-610, 2500nM

A C

800000

1000000

1200000

Fluo

resc

necn

e

800000

1000000

1200000Fl

uore

scne

cne

(a

400000

600000

300 320 340 360 380Wavelength (nm)

400000

600000

300 320 340 360 380Wavelength (nm)

1400000 1400000B D

1000000

1200000

1400000

nce

(a.u

.)

MutantCapsidST-148, 500nMST-148, 2500nM

1000000

1200000

1400000

ce (a

.u.)

Mutant CapsidDMSOST-610, 2500nM

B D

600000

800000

Fluo

resc

en

600000

800000

Fluo

resc

en

400000300 320 340 360 380

Wavelength (nm)

400000300 320 340 360 380

Wavelength (nm)

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

50

60

70

Mock-infected QD Mock-infected BIDVehicle BID ST-148 QDST-148 BID

B

10

20

30

40

50pg

/mL

1.E+07

1.E+08

u/m

l)

A

700

Mock-infected QD Mock-infected BIDVehicle BID ST-148 QDST-148 BID

0IL-12 TNFa IL-6

1.E+04

1.E+05

1.E+06

r (pf

u/g

tissu

e or

pfu

C

IL-12 TNFα IL-6

300

400

500

600

pg/m

L

1.E+03

Tite

r

Veh QD BID Veh QD BID Veh QD BIDSpleen Liver Plasma

0

100

200

MCP-1 IFN-gMCP-1 IFN-γ

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from

140160180

Mock-infected Vehicle ST-148B

406080

100120

pg/m

L

1.E+07

1.E+08

pfu/

mL)

A

020

IL-12 TNFa IL-6

800Mock-infected Vehicle ST-1481.E+05

1.E+06

Tite

r (pf

u/g

or p

C

IL-12 TNFα IL-6

300

400

500

600

700

800

pg/m

L

1.E+04Veh ST-148 Veh ST-148 Veh ST-148

Spleen Liver Plasma

0

100

200

300

MCP-1 INF-y

p

MCP-1 IFN-γ

on May 7, 2018 by guest

http://aac.asm.org/

Dow

nloaded from