CVI Accepts, published online ahead of print on 26 November 2014 Clin. Vaccine Immunol. doi:10.1128/CVI.00520-14 Copyright © 2014, American Society for Microbiology. All Rights Reserved.

1

Mapping Broadly Reactive Norovirus Genogroup I and II Monoclonal

2

Antibodies

3

Sue E. Crawford1, Nadim Ajami1, Tracy Dewese Parker1, Noritoshi Kitamoto2,

4

Katsuro Natori3, Naokazu Takeda3, Tomoyuki Tanaka4 , Baijun Kou1, Robert L. Atmar 1,5

5

, and Mary K. Estes1,5

6

1

7

Houston, TX, 77030, USA; 2School of Human Science and Environment, University

8

of Hyogo, Hyogo 670-0092, Japan; 3Department of Virology II, National Institute of

9

Infectious Diseases, Shinjuku, Tokyo,162-8640, Japan; 4Sakai City Institute of Public

10

Health, Sakai, Osaka, 590-0953, Japan; 5 Department of Medicine, Baylor College of

11

Medicine, Houston, TX 77030

12

Running title: Epitope common to Noroviruses

Department of Molecular Virology and Microbiology, Baylor College of Medicine,

13 14

*Corresponding Author. Mailing address: Department of Molecular Virology and

15

Microbiology, Baylor College of Medicine, One Baylor Plaza, Mail Stop BCM-385,

16

Houston, TX 77030. Phone: (713) 798-3585. Fax: (713) 798-3586. E-mail:

17

[email protected].

18

19

ABSTRACT

20

Noroviruses are responsible for most acute nonbacterial epidemic outbreaks of

21

gastroenteritis worldwide. To develop cross-reactive monoclonal antibodies (MAbs)

22

for rapid identification of genogroup I (GI) and genogroup II (GII) noroviruses (NoVs)

23

in field specimens, mice were immunized with baculovirus-expressed recombinant

24

NoV virus-like particles (VLPs). Nine MAbs to the capsid protein were identified that

25

detected both GI and GII NoV VLPs. These MAbs were tested in competition

26

enzyme-linked immunosorbant assays (ELISA) to identify common epitope

27

reactivities to GI and GII VLPs. Patterns of competitive reactivity placed these MAbs

28

into two epitope groups (1 and 2). MAbs NV23 and NS22 in group 1 and F120 in

29

group 2 were mapped to a continuous region in the C-terminal P1 sub-domain of the

30

capsid protein. This domain is within regions previously defined to contain cross-

31

reactive epitopes in GI and in GII viruses suggesting common epitopes are clustered

32

within the P1 domain of the capsid protein. Further characterization in the

33

accompanying manuscript by Kou et al., revealed that MAb NV23, in epitope group 1,

34

was able to detect GI and GII viruses in stool. Inclusion of the GI and GII cross-

35

reactive MAb NV23 in antigen detection assays may facilitate the identification of GI

36

and GII human noroviruses in stool samples as the causative agent of outbreaks and

37

sporadic cases of gastroenteritis worldwide.

38 39

2

40

INTRODUCTION

41

Noroviruses (NoVs) are the major cause of acute, epidemic nonbacterial

42

gastroenteritis in adults and children in both developing and industrialized countries

43

(1-3). In the United States, NoVs cause 19-21 million cases each year (4, 5). NoV

44

outbreaks have been identified in children (6), the elderly (7), military personnel (8,

45

9), immunocompromised individuals (10), restaurant patrons (11, 12), travelers to

46

developing countries (13, 14), passengers of cruise ships (15), residents of

47

healthcare facilities such as nursing homes (16, 17) and hospitals (18), and other

48

populations housed in close quarters (19). The increasing incidence of NoV infections

49

emphasizes the need to quickly detect and identify the causative agent because

50

early diagnosis of NoV infection can be crucial in the effective control of outbreaks

51

and decrease secondary attack rate (20).

52

Currently, only one immunoassay, the RIDASCREEN Norovirus ELISA (3rd

53

generation), is available for NoV diagnosis in the USA and this assay is only

54

approved to be used in outbreak settings due to its low sensitivity of detection. The

55

difficulty in developing broadly detecting NoV diagnostics is due to the diversity of

56

NoV strains. NoVs are classified based on phylogenetic analysis of the viral capsid

57

(VP1) gene into six genogroups (G1-GVI). Viruses within genogroups in GI, GII and

58

GIV cause human infections. Genogroups are further subdivided into genotypes and

59

there are at least 9 GI and 22 GII genotypes, respectively (21, 22). The amino acid

60

sequence diversity is 45% between genogroups (22).

61

Clear relationships between genotypes and antigenicity have not yet been

62

determined due to the lack of a cultivation system.

3

63

Expression of the 3’ end of the genome by using the recombinant baculovirus

64

system results in the formation of virus-like particles (VLPs) that are structurally and

65

antigenically similar to the native virion (23-25). The major capsid protein VP1 is

66

structurally divided into the shell (S) domain that forms the internal structural core of

67

the particle, and the protruding (P) domain, which is exposed on the outer surface of

68

the particle (23). The P domain is further subdivided into the P1 subdomain (residues

69

226 to 278 and 406 to 520 for GI.1 Norwalk virus) and the P2 subdomain (residues

70

279 to 405 for GI.1 Norwalk virus) (23). P2 represents the most exposed surface of

71

the viral particle and is involved in cellular histoblood group antigen (HBGA) binding

72

(26-28).

73

In spite of x-ray crystallographic knowledge of several noroviruses, information

74

is just beginning to emerge defining specific regions of the capsid protein containing

75

cross-reactive epitopes. Most information on the antigenic characteristics of NoVs

76

comes from the study of monoclonal antibodies (MAbs) generated to VLPs from both

77

GI and GII viruses (27, 29-40). The majority of these MAbs are genogroup-specific

78

and only recognize viruses closely related to the immunogen used to generate the

79

MAb. The present study analyzed cross-reactive MAbs that recognize epitopes on

80

viruses in both genogroups GI and GII that may be useful in the development of

81

improved diagnostic assays to detect NoVs.

82

4

83

MATERIALS AND METHODS

84

Development and characterization of monoclonal antibodies. MAbs were

85

isolated as previously described (33). A panel of 9 MAbs (NV23, NV37, NV3, NV57,

86

NV7, NS22, NS941, F8, and F120) were generated to NoV VLPs. MAbs NV23,

87

NV37, and NV3 hybridomas were previously derived from spleen cells of mice

88

immunized orally with recombinant Norwalk virus [NV, GI.1; sequence accession

89

number M87661 (25, 41)] VLPs, while MAbs F8 and F120 hybridomas were obtained

90

from spleen cells of mice immunized orally with recombinant Kashiwa 47 virus [KAV,

91

GII.13; AB078334 (33)] VLPs. MAbs NV57 and NV7 hybridomas were obtained from

92

spleen cells of mice immunized orally with NV VLPs. MAbs NS22, and NS941

93

hybridomas were obtained from spleen cells of mice immunized orally with a mixture

94

of NV and recombinant Snow Mountain Agent [SMA, GII.2 (31)] VLPs. Two

95

previously characterized MAbs, NV3901 and NS14, were also used in this study (35).

96

The binding reactivity of these MAbs was characterized by direct enzyme-

97

linked immunosorbant assay (ELISA) against a panel of 4 GI VLPs and 7 GII VLPs

98

(33). The GI VLPs used were GI.1 Norwalk/1968 (NV; M87661), GI.4 1643/2008

99

(GQ413970), GI.6 TCH-099/2003 (KC998959), and GI.7 TCH-060/2003 (JN005886).

100

The GII VLPs used were GII.2 Snow Mountain/1976 (SMA; AY134748) (31) and

101

TCH-560/2002 (KC998960), GII.3 TCH-577/2004 (KF006265), GII.4 HOV TCH-

102

186/2002 (EU310927) (35) and Grimsby/1995 (GRV; AJ004864) (42), GII.6 E99-

103

13646/1999 (GU930737), GII.7 TCH-134/2003 (KF006266), GII.12 E00-13842/2000

104

(KF006267), and GII.17 Katrina/2005 (DQ438972). Purified NoV particles (100 µl of 1

105

µg/ml in 0.01 M PBS, pH 7.4) were coated onto 96-well polyvinyl chloride plates

5

106

(Dynatech, Chantilly, VA) overnight at 4oC. The plates were blocked with 200 µl 5%

107

BLOTTO (Carnation nonfat milk) in PBS for 2 hours at 37oC. After the plates were

108

washed three times with PBS containing 0.05% Tween 20 (PBS-T), serial two-fold

109

dilutions beginning at 1:100 in 0.5% BLOTTO of each MAb was added to each well

110

and the plates were incubated for 1 h at 37oC. The plates were washed five times

111

with PBS-T and bound antibody was detected by addition of 100 µl of goat anti-

112

mouse IgG-HRP (1:3000 dilution in 0.5% BLOTTO, Sigma-Aldrich, St. Louis, MO) for

113

1 h at 37oC. The plates were then washed five times with PBS-T and developed with

114

100 µl of TMB Microwell Peroxidase Substrate System (A:B=1:1, KPL, Gaithersberg,

115

MD) for 10 min at room temperature. The reaction was stopped by the addition of 100

116

µl of 1 M H3PO4 and the optical density (OD) at 450 nm was read with a SpectraMAX

117

M5 plate reader (Molecular Devices, Sunnyvale, CA). Non-coated wells were used as

118

negative controls. The positive cutoff threshold was calculated as the mean of the

119

negative control wells plus three times the standard deviation of the negative control

120

wells.

121

Competition ELISA. Two methods of competition ELISA were used. For the first

122

competition ELISA, MAbs were biotinylated using an Antibody Biotinylation Kit

123

(American Qualex Manufactures, San Clemente, CA). Briefly, MAbs (5 mg/ml) were

124

dialyzed against 1:10 dilution of carbonate buffer overnight. A long chain N-

125

hydroxysuccinimide ester biotin (NHS-LC-Biotin) solution (0.5 mg biotin/0.5 ml

126

distilled water) was added to the protein solution to a final concentration of 74 µg

127

biotin/mg protein and gently shaken for 1 hour at room temperature. The reaction

128

mixture was then passed over a Sephadex G25 column. The biotin conjugate was

6

129

eluted off the column using PBS buffer, concentrated, and stored at 4°C. Microtiter

130

plates (Sumitomo Bakelite Co Ltd., Tokyo, Japan) were coated overnight at 4°C with

131

50-500 ng/well of GI.1 NV or GII.4 GRV (42) VLPs in 100 µl of 0.05 M carbonate

132

bicarbonate buffer (pH 9.6). Each well was washed twice with PBS-T and blocked

133

with 5% BLOTTO in PBS-T for at least 1 hour at room temperature. In separate

134

tubes, optimized concentrations of biotinylated MAb (which produced an OD of 0.5 to

135

2.0 when no competitor MAb was present) were added to competitor MAb diluted

136

(×64, ×256, ×1024, ×4096, and ×16,384) in PBS (pH 7.2). Following two washes with

137

PBS-T, 100 µl of the MAb mixtures was added to duplicate wells, and the plates were

138

incubated for 2 hours at 37°C. After washing four times with PBS-T, 50 µl of a 1:1000

139

to 1:2000 dilution of HRP-conjugated streptavidin (Cosmo Bio Co., LTD., Tokyo,

140

Japan) was added to all wells and reacted for 2 hours at 37°C. After washing, 50 µl

141

of o-phenylenediamin-H2O2 (0.5 mg/ml o-phenylenediamin, 0.002% H2O2, 0.1 M

142

citrate-phosphate buffer, pH 5.5) was added as a substrate and developed for 10

143

minutes. The color reaction was stopped by the addition of 50 µl of 2 N H2SO4. The

144

optical density (OD) at 405 nm and 630 nm was determined. The OD average of

145

duplicates was calculated and the percentage of competition or enhanced binding

146

was determined for all competitor MAb concentrations (×64, ×256, ×1024, ×4096 and

147

×16384) based on the value of the PBS control. The value of biotinylated MAb

148

binding to coating VLPs in the absence of competitor MAb was defined as zero.

149

A second method from Hale et al. (29) was used to analyze competition

150

between MAbs NV3901, NS14 and NV23. MAbs were purified using protein G

151

columns (Pierce, Rockford, IL) according to the manufacturer’s instructions. One

7

152

MAb, at a concentration of 2 μg/ml in 0.05 M carbonate bicarbonate buffer, pH 9.6,

153

was used to coat flat-bottomed polyvinylchloride microtiter plates (Dynatech

154

Laboratories, Alexandria, VA) overnight at 4°C. In separate tubes, a constant

155

concentration of NV or HOV VLPs (based on each MAb to give an OD450 between 0.5

156

–0.6) was added to decreasing concentrations of competitor MAb (5, 1, 0.5, 0.1, and

157

0.05 μg/ml) in 0.5% BLOTTO in PBS and incubated overnight at 4°C. A control of

158

VLPs in 0.5% BLOTTO without competitor was included in each plate. The antibody-

159

coated microtiter plates were washed three times with PBS-T and blocked with 5%

160

BLOTTO for 1 hour at 37°C. Following six washes with PBS-T, each of the VLP-MAb

161

reaction mixtures was added to triplicate wells, and the plates were incubated for 2

162

hours at 37°C. After washing six times, a 1:5000 dilution of rabbit anti-NV VLP or

163

rabbit anti-HOV hyperimmune serum in 0.5% BLOTTO was added to each well, and

164

the plates were incubated for 1 hour at 37°C. Following incubation, plates were again

165

washed, and a 1:5000 dilution of goat anti-rabbit IgG conjugated to horseradish

166

peroxidase (Sigma, St. Louis, MO) was added. Plates were incubated for an

167

additional hour, and washed again. To develop the ELISA, 100 μl of 3,3’,5,5’-

168

tetramethylbenzidine (Kirkegaard & Perry Laboratories, Inc., Gaithersburg, MD) was

169

added to each well, and the color reaction was stopped by the addition of 100 μl of 1

170

M H3PO4. The optical density at 450 nm was read, and the average of triplicates was

171

calculated. The percentage of competition was determined for the competitor MAb

172

concentrations based on the value of the PBS control, where the value of the VLP

173

binding to coating MAb in the absence of competitor MAb was defined as zero.

174

Homotypic competition was included as a positive control for all coating MAbs.

8

175

Cloning, expression and analysis of norovirus fusion proteins. Characterization

176

of the GST-norovirus fusion protein deletion mutants has been described previously

177

(35). Numbering of the constructs indicates the N-terminal (first) and C-terminal (last)

178

norovirus residue contained within a construct. NoV fusion proteins were expressed

179

in BL-21 cells (Novagen, Madison WI) and purified using glutathione-sepharose 4B

180

beads (Amersham Pharmacia Biotech, Piscataway, NJ).

181

Proteins were separated by SDS-PAGE and transferred onto a nitrocellulose

182

membrane (Hybond-C, Amersham Pharmacia Biotech). The proteins were detected

183

using a polyclonal goat anti-GST antibody (Amersham Pharmacia) at a dilution of

184

1:8000, a mouse hyperimmune anti-NV VLP serum at a dilution of 1:5000, or a rabbit

185

hyperimmune anti-HOV VLP serum at a dilution of 1:5000 in 0.5% BLOTTO. MAb

186

ascites were used for detection at a dilution of 1:1000. All secondary antibodies used

187

were conjugated to horseradish peroxidase (Sigma, St. Louis, MO). Membranes were

188

developed by chemiluminescence using the Western Lightning detection reagent

189

(Perkin-Elmer Life Sciences, Inc., Boston, MA) following the manufacturer’s protocol.

9

190

RESULTS

191

Monoclonal antibodies to norovirus VLPs that recognize both genogroup I and

192

genogroup II viruses can be separated into two epitope groups. We previously

193

reported the characterization of MAbs generated against NoV VLPs with respect to

194

their reactivity to GI and GII VLPs by ELISA or western blot (33). MAbs NV23, NV37,

195

and NV3, obtained from mice administered GI.1 NV VLPs perorally, and F8 and

196

F120, obtained from mice administered GII.13 KAV VLPs perorally, recognized both

197

GI [GI.1 NV by ELISA and GI.1 NV and Seto 124 (SeV; AB031013), GI.2 Funabashi

198

258 (FUV; AB078335) and G1.4 Chiba 407 (CV; AB022679) by western blot] and GII

199

[GII.2 SMA, GII.4 GRV and Narita 104 (NAV; AB078336), GII.6 Ueno 7K (UEV;

200

AB078337), GII.12 Chitta 76 (CHV; AB032758), and GII.13 KAV by ELISA] VLPs

201

(33). Additional MAbs, NV57 and NV7, were obtained from mice administered NV

202

VLPs perorally, and NS22 and NS941, were obtained from mice administered NV

203

and SMA VLPs perorally. Subsequent characterization of the reactivity to a larger

204

panel of GI and GII VLPs by ELISA (Table 1) showed each MAb recognized at least

205

a subset of both GI and GII VLPs by ELISA suggesting each MAb detects a common

206

epitope in both GI and GII VLPs.

207

To determine whether the MAbs detect similar or different epitopes in GI and

208

GII VLPs, each of these MAbs was then tested in competition ELISAs with GI.1 NV

209

and GII.4 GRV VLPs to define them into epitope groups (Fig. 1). In competition

210

ELISAs with GI.1 NV VLPs, homotypic competition for the 9 MAbs was found to vary

211

between 10 and 90% at the highest concentration (×64, about 500 µg/ml) of

212

competitor antibody used in the ELISAs (Fig. 1A, biotinylated MAb NV23, NV37,

10

213

F120 and F8 shown). MAb NV23, NV37, NV3, NV57, NV7, NS22, or NS941 each

214

showed 80% or greater competition with itself and these other 6 MAbs (Fig. 1A, top

215

panels for biotinylated MAb NV23 or NV37). When MAb F8 or F120 was used as the

216

competitor MAb, only 50 to 60% competition was observed (at x64 concentration) for

217

biotinylated MAb NV23, NV37, NV3, NV57, NV7, NS22, or NS941 (Fig. 1A, top

218

panels for biotinylated MAb NV23 or NV37).

219

A reduced level of competition was seen when MAbs NV23, NV37, NV3,

220

NV57, NV7, NS22 and NS941 were used as competitor MAbs with biotinylated MAbs

221

F120 or F8 as compared to homotypic competition (Fig. 1A, bottom panels), although

222

to a lesser extent than seen when F8 or F120 were used as the competitor MAb

223

against these MAbs (Fig. 1A, top panels). This difference may be due to a

224

conformational change in the epitope recognized by the antibody leading to a

225

reduced affinity in binding. Alternatively, the epitopes may overlap such that one MAb

226

causes steric hindrance while the other may not. Similar competition patterns were

227

obtained when GII.4 GRV VLPs were used as a coating antigen with NV23 and F120

228

(Fig. 1B, left and right panels, respectively).

229

The 9 MAbs were classified into two groups according to their competition

230

ELISA patterns as follows: group 1 (NV23, NV37, NV3, NV57, NV7, NS22 and

231

NS941) and group 2 (F8 and F120). The results obtained suggest the 7 MAbs of

232

group 1 recognize the same or almost identical epitope. Group 2 MAbs likely

233

recognize a similar or closely overlapping epitope with group 1.

234

11

235

Groups 1 and 2 monoclonal antibodies recognize an epitope within the C-

236

terminal P1 domain. To identify the binding site(s) for the epitope groups 1 and 2

237

cross-reactive MAbs, a subset of MAbs from each epitope group was tested by

238

western blot analysis for their ability to recognize deletion mutants of the GII.4 HOV

239

VP1 capsid protein. From epitope group 1, MAbs NV23 and NS22 were mapped

240

because these MAbs were identified to react with GI, GII and GIV VLPs by both

241

direct ELISA and by capture ELISA in the companion paper by Kou et al. Both NV23

242

and NS22 produced the same pattern of recognition (Fig. 2) and detected amino

243

acids 453-495 but failed to detect amino acids 473-540, which defined the binding

244

region for these monoclonal antibodies as contained within GII.4 HOV amino acids

245

453–472 (corresponding to GI.1 NV amino acids 437-457). Although similar to NV23

246

and NS22, each of the other MAbs from epitope group 1 (NV3, NV7, NV37, NV57

247

and NV941, and epitope group 2 F8) detected the minimal HOV 453-495 construct

248

(Fig. 3) but these were not further mapped because they lacked reactivity to VLPs in

249

a capture ELISA. Analysis of MAb NV23 binding to GI.1 NV deletion mutants

250

confirmed that NV23 bound to an overlapping sequence within the NV capsid protein

251

with the minimal binding region, based on available constructs, comprised of amino

252

acids 406-466 (Fig. 4).

253

Western blot analysis with the GII.4 HOV VP1 capsid protein deletion mutants

254

was performed to characterize the binding site of the epitope group 2 MAb F120.

255

Similar to the epitope group 1 MAbs, the epitope group 2 MAb F120 also detected

256

the GII.4 HOV amino acids 453-495 and failed to detect amino acids 473-540

257

indicating the epitopes for groups 1 and 2 map to amino acids 453-472 (Fig. 5).

12

258 259

NoV monoclonal antibodies bind to distinct sites of the norovirus capsid

260

protein. The location of the domain that epitope group 1 MAbs NV23 and NS22 and

261

epitope group 2 MAb F120 detect is shown in the crystal structures of VP1 from GI.1

262

NV (amino acids 437-457) and GII.4 2004 variant (amino acids 453-472) P domains

263

(Fig. 6A, top left and right panels, respectively). Previous work by our group identified

264

the epitopes for MAbs NV3901 and NS14, respectively (35). MAb NV3901 is specific

265

for GI viruses and mapped to NV amino acids 454-520 and specifically to E472 that

266

forms a salt bridge with K514 (35). MAb NS14 recognizes GII viruses and mapped to

267

HOV amino acids 473-495. Alignment of the binding sites of each of these MAbs

268

shows that all of the MAbs recognize overlapping or adjacent regions of the NoV

269

capsid proteins (Fig. 6B). Alignment of the epitope group 1 and group 2 MAb binding

270

domain from the test VLPs used in this and the companion study of Kou et al. shows

271

a number of conserved amino acids with only the alanine completely conserved

272

within this domain (Fig. 6C).

273

MAb NV23 is broadly reactive and detects GI and GII strains of virus (43),

274

whereas NV3901 and NS14 are GI- and GII-specific, respectively. To further

275

characterize MAb NV23 and determine whether the binding site for NV23 is distinct

276

from the sites for NS14 and NV3901, we performed competition ELISAs using MAbs

277

NV23 (Fig. 7A), NS14 (Fig. 7B) and NV3901 (Fig. 7C). Because we found that the

278

VLP conformation changes upon direct coating onto ELISA plates, we performed

279

competition ELISAs by capturing the VLPs using MAbs. Neither MAb NV3901 nor

280

NS14 competed for VLP binding to MAb NV23 when used as either the coating or

13

281

competitor antibody (Fig. 7). However, NV23 did show some level of competition with

282

NV3901 (although not greater than 50%), which suggests NV23 may inhibit NV3901

283

binding as a result of steric hindrance. These results confirm that MAbs NV23, NS14

284

and NV3901 recognize distinct epitopes within the C terminal P1 domain of VP1.

285 286

14

287

DISCUSSION

288 289

Noroviruses are a major cause of sporadic cases and epidemic outbreaks of

290

gastroenteritis. To effectively implement infection control measures following

291

outbreaks, sensitive and rapid diagnostic assay are needed to identify the causative

292

agents of viral gastroenteritis. Containment of outbreaks relies on rapid diagnosis;

293

identification of causative agent, within 3 days versus 4 or more days following the

294

first case, resulted in outbreak containment an average of 6 days sooner (7.9

295

compared to 15.4 days) (20). Many currently available immunologic reagents used to

296

study noroviruses are genotype-specific, which limits the usefulness to identify

297

antigenically distinct viruses. Broadly genogroup cross-reactive antibodies are

298

needed for rapid identification of NoVs in field specimens. Identification of cross-

299

reactive epitopes on the NoV capsid proteins also provides important information

300

regarding the antigenic characteristics of these viruses.

301

The present work extends our previous report (33, 44) that oral immunization

302

with different VLPs can generate NoV cross-reactive MAbs. These MAbs were found

303

to compete for binding on GI.1 and GII.4 VLPs and MAbs NV23, NS22 and F120

304

map to the C-terminal P1 sub-domain of the capsid protein that includes amino acids

305

453-472 in the GII.4 HOV sequence. Although the competition ELISA segregated

306

MAbs NV23 and NS22 into epitope group 1 and F120 into epitope group 2, these

307

MAbs map to the same region of the capsid suggesting the epitopes for these MAbs

308

are overlapping but not the same domain and may have competed for binding due to

309

steric hindrance. A full understanding of this observation may be obtained by

15

310

structural studies of VLP-Fab complexes, which is outside the scope of this

311

manuscript.

312

Interestingly, the amino acids 453-472 domain overlaps or is adjacent to the

313

region we previously described that contains epitopes for genogroup specific MAbs

314

NV3901 (GI-specific) and NS14 (GII-specific) (35). The epitopes for NV23

315

characterized in this study and for NV3901 and NS14 appear to be distinct based on

316

competition ELISAs. This C-terminal domain, while not any more highly conserved

317

than the P1 sub-domain overall, contains conserved residues suitable to act as NoV

318

cross-reactive epitopes (Fig. 6C). For the MAbs mapped in this study, the domain

319

analogous to amino acids 453-472 in the GII.4 HOV structure are amino acids 437-

320

457 in the GI.1 NV structure (26) (Fig 6A). In the context of the NV VLP structure, the

321

surface-exposed residues in this domain can be seen within the 5-fold axis in the P1

322

subdomain (23) (Fig. 6A bottom panel, surface-exposed residues labeled in

323

magenta). Interestingly, the conserved glutamic acid/glutamine at position 448

324

(numbered according to the NV sequence) is not surface exposed. However, the

325

proline at position 451 (numbered according to the NV sequence) is conserved in all

326

GI and GII sequences with the exception of the GII.7 virus strain. Modeling the

327

threonine residue in GII.7 in place of the proline in the context of the GII.4 2004

328

variant P domain indicates that the threonine is retained on the surface of the

329

structure. The remainder of surface exposed residues following position 451

330

(numbered according to the NV sequence) are surface exposed in both the NV and

331

GII.4 2004 variant structures (Fig. 6A, top panel) suggesting this domain may be

332

responsible for the common GI and GII reactivity.

16

333

The nine MAbs described in this study detected both GI and GII VLPs by

334

direct ELISA when the VLPs were coated onto ELISA plates in PBS. However, Kou

335

et al., observed an increase or decrease in reactivity to a subset of VLPs in the direct

336

ELISA with MAbs NV3 and NV57 when the VLPs were plated in water (43).

337

Differences in GI.1 and GII.4 attachment efficiency in the presence of phosphate has

338

been previously described and was attributed to differential responses of the unique

339

arrangement of exposed amino acid residues on the capsid surface of each VLP

340

strain, which may explain the different reactivity of MAbs to VLPs plated in water

341

versus PBS (45). Furthermore, the reactivity of these MAbs differed in a capture

342

ELISA compared to the reactivity in the direct ELISA. The reactivity in the capture

343

ELISA correlated with analysis by surface plasmon resonance, which revealed that

344

NV23 and NS22 bound GI and GII VLPs with high affinity, while NV57 only bound GII

345

test VLPs. In contrast, NV37 bound only GI.1 VLPs and NV3 did not bind any test

346

VLP. Additionally, F120 and F8, similar to the epitope group 1 MAbs NV3, NV7,

347

NV37, NV57 and NS941, do not detect GI or GII VLPs in a capture ELISA indicating

348

these MAbs interact with amino acids that are not surface exposed on VLPs in

349

solution but interact with amino acids that are exposed due to binding to the ELISA

350

plate. These results support that the conformation of the VLP changes, either due to

351

the buffer used to plate the VLPs or to direct coating of VLPs onto the wells in ELISA

352

plates, thus exposing residues that are buried within the VLP in solution.

353

The presence of broadly cross-reactive epitopes on VLPs was demonstrated

354

by the reactivity of polyclonal antiserum as well as monoclonal antibodies generated

355

against NoV VLPs that detected both GI and GII VLPs (29, 33, 35, 46-48). Additional

17

356

evidence that the C-terminal P domain contains cross-reactive epitopes is the

357

identification of monoclonal antibody MAb14-1. This MAb, which detects 15

358

recombinant virus-like particles GI.1, GI.4, GI.8, and GI.3 and GII.1-7, GII.12-14, and

359

GII.16, genotypes [renumbered according to Zheng et al. (22) and Kroneman et al.

360

(21)], was mapped to a conformational epitope involving amino acids 418 to 426 and

361

526 to 534, regions surrounding amino acids 453-472 identified as the site for the

362

cross-reactive MAbs characterized in this study (Fig. 6B) (48). Additionally, the

363

crystal structure of monoclonal antibody 5B18, which is currently in use in a

364

commercial norovirus ELISA detection kit (Denka Seiken, Japan) and binds

365

numerous GII genotypes but not GI NoVs, was found to interact with amino acids

366

Val433, Glu496, Asn530, Tyr533, Thr534, and Leu535 on the GII.10 P domain (30).

367

However, this region of the protruding domain was determined to be occluded in the

368

crystal structure and led to the suggestion that NoV particles are capable of extreme

369

conformational flexibility to allow this monoclonal antibody to bind to its epitope (30).

370

The only norovirus diagnostic immunoassay currently approved for use in the

371

United States is the RIDASCREEN Norovirus 3rd Generation Antigen EIA (enzyme-

372

linked immunosorbent assay). However, it is currently approved for use only in

373

outbreak settings because it lacks sensitivity (49-52). The increasing evidence that

374

noroviruses are undergoing antigenic variation highlights the need for diagnostic

375

assays that detect broadly cross-reactive epitopes (53-55). We have identified a

376

broadly cross-reactive epitope in the C terminal P1 domain that is surface exposed;

377

the majority of these residues are dissimilar between human norovirus strains

378

suggesting that this region may remain invariant while other domains of the virus

18

379

undergoing antigenic variation. Therefore, inclusion of MAb NV23 in antigen

380

detection assays may facilitate the identification of GI and GII human noroviruses in

381

stool samples as the causative agent of outbreaks and sporadic cases of

382

gastroenteritis worldwide.

383

19

384

ACKNOWLEDGMENTS

385 386

This work was supported by Public Health Service grants NIH R01 AI38036 and P01

387

AI57788, training grant T32 A107471 (T.D.P.), and P30 CA125123 that funds the

388

Recombinant Protein and Monoclonal Antibody Production Shared Resource at

389

Baylor College of Medicine from the National Institute of Allergy and Infectious

390

Disease, and the John S. Dunn Research Foundation (to RLA). M.K.E. is named as

391

an inventor on patents related to cloning of the Norwalk virus genome and she, TNT

392

and NK receive royalties from commercialization activities through Baylor College of

393

Medicine for NoV monoclonal antibody reagents.

394 395

20

396

FIGURE LEGENDS

397 398

Figure 1. Competition ELISAs for biotinylated MAbs using GI.1 and GII.4 VLPs.

399

Biotinylated monoclonal antibodies (A) NV23, NV37, F120 and F8 or (B) NV23 and

400

F120 were mixed with competitor monoclonal antibodies NV23 (−●−), NV37 (−○−),

401

NV3 (−▲−), NV57 (−Δ−), NV7 (−■−), NS22 (−□−), NS941 (−♦−), F8 (--●--), and F120

402

(--○--) and then used to detect (A) GI.1 NV or (B) GII.4 VLPs bound to microtiter

403

plates. The 50% cutoff for significant competition is indicated by the dotted line.

404

Homotypic and heterotypic competition were observed.

405 406

Figure 2. Mapping epitope group 1 monoclonal antibodies NV23 and NS22. (A) HOV

407

VLPs, GST and purified GST-tagged HOV capsid protein deletion mutants containing

408

the indicated residues were analyzed by western blot with anti-GST antiserum,

409

polyclonal rabbit anti-HOV VLP antiserum, MAbs NV23 or NS22 as indicated below

410

each blot. (B) Schematic representation of the location of the constructs relative to

411

the full-length VP1 protein and a summary of recognition by NV23 and NS22. GST,

412

purified GST protein; HOV VLPs, purified Houston virus GII.4 VLPs.

413 414

Figure 3. Monoclonal antibodies detect HOV amino acids 453-495. GII.4 HOV VLPs

415

and purified GST-tagged HOV capsid construct amino acids 453-495 were analyzed

416

by western blot with polyclonal rabbit anti-HOV VLP antiserum, MAbs NV3, NV37,

417

NS941, NV57, NV7 and F8 as indicated below each blot.

418

21

419

Figure 4. Mapping epitope group 1 MAb NV23. (A) GI.1 NV VLPs and purified GST-

420

tagged NV capsid protein deletion mutants containing the indicated residues were

421

analyzed by western blot with polyclonal rabbit anti-NV VLP antiserum and MAb

422

NV23 as indicated below each blot. (B) Schematic representation of the location of

423

the constructs relative to the full-length VP1 protein and a summary of recognition by

424

NV23.

425 426

Figure 5. Mapping epitope group 2 MAb F120. (A) Purified GST-tagged HOV capsid

427

protein deletion mutants containing the indicated residues were analyzed by western

428

blot with polyclonal rabbit anti-HOV VLP antiserum (top panel), or F120 (bottom

429

panel). (B) Schematic representation of the location of the constructs relative to the

430

full-length VP1 protein and a summary of recognition by F120. GST, purified GST

431

protein; HOV VLPs, purified Houston virus VLPs.

432 433

Figure 6. Location of binding site for epitope group 1 MAbs NV23 and NS22 and

434

epitope group 2 MAb F120. (A) The location of the amino acids that correspond to

435

the epitope binding sites for epitope groups 1 MAbs NV23 and NS22 and 2 MAb

436

F120 on the crystal structure of GI.1 NV [amino acids 437-457, top right panel;

437

Protein Data Bank (PDB) accession number 1IHM] and GII.4 HOV (amino acids 453-

438

472, top left panel; PDB accession number 3SKB) P domain dimer prepared using

439

PyMOL. The ribbon structure for P1 is shown in grey, P2 in blue, and the residues

440

corresponding to the minimal binding domain are indicated in green with the surface-

441

exposed residues in magenta. Bottom panels show (left panel) the NV VLP crystal

22

442

structure (VIPER data base accession number 1IHM) with the A-B dimers in blue, the

443

C-C dimers in green, and the surface-exposed residues in the MAb minimal binding

444

domain in magenta using Chimera. The boxed area (white) is magnified (right panel)

445

and shows the P2 domain surrounding the 5-fold axis with the MAb surface-exposed

446

binding residues in magenta. (B) Alignment of binding sites for cross-reactive MAbs.

447

The binding site for each MAb is shown relative to its position within the P domain.

448

Amino acid numbers for the MAbs characterized in this study and NS14 correspond

449

to GII.4 HOV sequence (GenBank accession number EU310927); amino acid

450

numbers for NV3901 correspond to GI.1 NV sequence (M87661); amino acid

451

numbers for MAb 14-1 correspond to GII.4 1207 sequence (DQ975270); amino acid

452

numbers for 8C7 correspond to GI.1 Norwalk virus (NV 96-908, AB028247); amino

453

acid numbers for 5B18 correspond to GII.10 Vietnam026 (AF504671). (C) Alignment

454

of the MAb-binding domain in the P1 capsid sequences for the indicated virus strains.

455

The amino acid numbering corresponds to each virus strain|GenBank assession

456

number. The minimal binding domain for MAbs NV23, NS22 and F120 on the GI.1

457

NV and GII.4 2004 HOV capsid region are indicated in green with the surface-

458

exposed residues in magenta. The symbols under each column denote (*) identical

459

amino acids, (:) different but highly conserved amino acids, (.) amino acids that are

460

somewhat similar, and ( ) dissimilar amino acids.

461 462

Figure 7. Competition ELISAs between GI-specific (3901), GII-specific (NS14) and GI

463

and GII-specific (NV23) monoclonal antibodies. Competition ELISAs showing

464

competition between monoclonal antibodies (A) NV23, −♦−, (B) NS14, −■−, and (C)

23

465

3901, −▲−. Coating antibody is indicated at the top of the graph, and the competitor

466

MAb in the box. The 50% cutoff for significant competition is indicated by the dotted

467

line.

24

468

Reference List

469 470

1. Glass, R. I., U. D. Parashar, and M. K. Estes. 2009. Norovirus gastroenteritis. N. Engl. J Med 361:1776-1785.

471 472

2. Atmar, R. L. and M. K. Estes. 2006. The epidemiologic and clinical importance of norovirus infection. Gastroenterol. Clin North. Am. 35:275-90, viii.

473 474

3. Koo, H. L., N. Ajami, R. L. Atmar, and H. L. Dupont. 2010. Noroviruses: The leading cause of gastroenteritis worldwide. Discov. Med 10:61-70.

475 476 477

4. Hall, A. J., B. A. Lopman, D. C. Payne, M. M. Patel, P. A. Gastanaduy, J. Vinje, and U. D. Parashar. 2013. Norovirus disease in the United States. Emerg. Infect Dis 19:1198-1205.

478 479 480

5. Scallan, E., R. M. Hoekstra, F. J. Angulo, R. V. Tauxe, M. A. Widdowson, S. L. Roy, J. L. Jones, and P. M. Griffin. 2011. Foodborne illness acquired in the United States-major pathogens. Emerg. Infect Dis 17:7-15.

481 482 483

6. Patel, M. M., M. A. Widdowson, R. I. Glass, K. Akazawa, J. Vinje, and U. D. Parashar. 2008. Systematic literature review of role of noroviruses in sporadic gastroenteritis. Emerg. Infect Dis 14:1224-1231.

484 485 486

7. Green, K. Y., G. Belliot, J. L. Taylor, J. Valdesuso, J. F. Lew, A. Z. Kapikian, and F. Y. Lin. 2002. A predominant role for Norwalk-like viruses as agents of epidemic gastroenteritis in Maryland nursing homes for the elderly. J Infect Dis 185:133-146.

487 488 489

8. Hyams, K. C., J. D. Malone, A. Z. Kapikian, M. K. Estes, J. Xi, A. L. Bourgeois, S. Paparello, R. E. Hawkins, and K. Y. Green. 1993. Norwalk virus infection among Desert Storm troops. J Infect Dis 167:986-987.

490 491 492 493

9. Sharp, T. W., K. C. Hyams, D. Watts, A. F. Trofa, G. J. Martin, A. Z. Kapikian, K. Y. Green, X. Jiang, M. K. Estes, M. Waack, and . 1995. Epidemiology of Norwalk virus during an outbreak of acute gastroenteritis aboard a US aircraft carrier. J Med Virol. 45:61-67.

494 495 496 497

10. Roddie, C., J. P. Paul, R. Benjamin, C. I. Gallimore, J. Xerry, J. J. Gray, K. S. Peggs, E. C. Morris, K. J. Thomson, and K. N. Ward. 2009. Allogeneic hematopoietic stem cell transplantation and norovirus gastroenteritis: a previously unrecognized cause of morbidity. Clin Infect Dis 49:1061-1068.

498 499 500 501 502

11. Daniels, N. A., D. A. Bergmire-Sweat, K. J. Schwab, K. A. Hendricks, S. Reddy, S. M. Rowe, R. L. Fankhauser, S. S. Monroe, R. L. Atmar, R. I. Glass, and P. Mead. 2000. A foodborne outbreak of gastroenteritis associated with Norwalk-like viruses: first molecular traceback to deli sandwiches contaminated during preparation. J Infect Dis 181:1467-1470.

503 504 505 506

12. Smith, A. J., N. McCarthy, L. Saldana, C. Ihekweazu, K. McPhedran, G. K. Adak, M. Iturriza-Gomara, G. Bickler, and E. O'Moore. 2012. A large foodborne outbreak of norovirus in diners at a restaurant in England between January and February 2009. Epidemiol. Infect 140:1695-1701.

25

507 508 509

13. Ajami, N., H. Koo, C. Darkoh, R. L. Atmar, P. C. Okhuysen, Z. D. Jiang, J. Flores, and H. L. Dupont. 2010. Characterization of norovirus-associated traveler's diarrhea. Clin Infect Dis 51:123-130.

510 511 512 513

14. Koo, H. L., N. J. Ajami, Z. D. Jiang, F. H. Neill, R. L. Atmar, C. D. Ericsson, P. C. Okhuysen, D. N. Taylor, A. L. Bourgeois, R. Steffen, and H. L. Dupont. 2010. Noroviruses as a cause of diarrhea in travelers to Guatemala, India, and Mexico. J Clin Microbiol 48:1673-1676.

514 515 516 517

15. Wikswo, M. E., J. Cortes, A. J. Hall, G. Vaughan, C. Howard, N. Gregoricus, and E. H. Cramer. 2011. Disease transmission and passenger behaviors during a high morbidity Norovirus outbreak on a cruise ship, January 2009. Clin Infect Dis 52:11161122.

518 519 520 521

16. Trivedi, T. K., T. DeSalvo, L. Lee, A. Palumbo, M. Moll, A. Curns, A. J. Hall, M. Patel, U. D. Parashar, and B. A. Lopman. 2012. Hospitalizations and mortality associated with norovirus outbreaks in nursing homes, 2009-2010. JAMA. 308:16681675.

522 523 524

17. Calderon-Margalit, R., R. Sheffer, T. Halperin, N. Orr, D. Cohen, and T. Shohat. 2005. A large-scale gastroenteritis outbreak associated with Norovirus in nursing homes. Epidemiol. Infect 133:35-40.

525 526 527

18. Rhinehart, E., S. Walker, D. Murphy, K. O'Reilly, and P. Leeman. 2012. Frequency of outbreak investigations in US hospitals: results of a national survey of infection preventionists. Am. J Infect Control. 40:2-8.

528 529 530 531 532

19. Yee, E. L., H. Palacio, R. L. Atmar, U. Shah, C. Kilborn, M. Faul, T. E. Gavagan, R. D. Feigin, J. Versalovic, F. H. Neill, A. L. Panlilio, M. Miller, J. Spahr, and R. I. Glass. 2007. Widespread outbreak of norovirus gastroenteritis among evacuees of Hurricane Katrina residing in a large "megashelter" in Houston, Texas: lessons learned for prevention. Clin Infect Dis 44:1032-1039.

533 534 535

20. Lopman, B. A., M. H. Reacher, I. B. Vipond, D. Hill, C. Perry, T. Halladay, D. W. Brown, W. J. Edmunds, and J. Sarangi. 2004. Epidemiology and cost of nosocomial gastroenteritis, Avon, England, 2002-2003. Emerg. Infect Dis 10:1827-1834.

536 537 538

21. Kroneman, A., E. Vega, H. Vennema, J. Vinje, P. A. White, G. Hansman, K. Green, V. Martella, K. Katayama, and M. Koopmans. 2013. Proposal for a unified norovirus nomenclature and genotyping. Arch. Virol 158:2059-2068.

539 540 541

22. Zheng, D. P., T. Ando, R. L. Fankhauser, R. S. Beard, R. I. Glass, and S. S. Monroe. 2006. Norovirus classification and proposed strain nomenclature. Virology 346:312-323.

542 543 544

23. Prasad, B. V., M. E. Hardy, T. Dokland, J. Bella, M. G. Rossmann, and M. K. Estes. 1999. X-ray crystallographic structure of the Norwalk virus capsid. Science 286:287290.

545 546

24. Green, K. Y., J. F. Lew, X. Jiang, A. Z. Kapikian, and M. K. Estes. 1993. Comparison of the reactivities of baculovirus-expressed recombinant Norwalk virus capsid antigen

26

547 548

with those of the native Norwalk virus antigen in serologic assays and some epidemiologic observations. J Clin Microbiol 31:2185-2191.

549 550

25. Jiang, X., M. Wang, D. Y. Graham, and M. K. Estes. 1992. Expression, selfassembly, and antigenicity of the Norwalk virus capsid protein. J Virol. 66:6527-6532.

551 552 553

26. Choi, J. M., A. M. Hutson, M. K. Estes, and B. V. Prasad. 2008. Atomic resolution structural characterization of recognition of histo-blood group antigens by Norwalk virus. Proc Natl Acad Sci U. S. A 105:9175-9180.

554 555 556

27. Higo-Moriguchi, K., H. Shirato, Y. Someya, Y. Kurosawa, N. Takeda, and K. Taniguchi. 2013. Isolation of cross-reactive human monoclonal antibodies that prevent binding of human noroviruses to histo-blood group antigens. J Med Virol 86:558-67.

557 558 559

28. Shanker, S., J. M. Choi, B. Sankaran, R. L. Atmar, M. K. Estes, and B. V. Prasad. 2011. Structural analysis of histo-blood group antigen binding specificity in a norovirus GII.4 epidemic variant: implications for epochal evolution. J Virol 85:8635-8645.

560 561 562

29. Hale, A. D., T. N. Tanaka, N. Kitamoto, M. Ciarlet, X. Jiang, N. Takeda, D. W. Brown, and M. K. Estes. 2000. Identification of an epitope common to genogroup 1 "norwalk-like viruses". J Clin Microbiol 38:1656-1660.

563 564 565 566 567

30. Hansman, G. S., D. W. Taylor, J. S. McLellan, T. J. Smith, I. Georgiev, J. R. Tame, S. Y. Park, M. Yamazaki, F. Gondaira, M. Miki, K. Katayama, K. Murata, and P. D. Kwong. 2012. Structural basis for broad detection of genogroup II noroviruses by a monoclonal antibody that binds to a site occluded in the viral particle. J Virol 86:36353646.

568 569 570

31. Hardy, M. E., S. F. Kramer, J. J. Treanor, and M. K. Estes. 1997. Human calicivirus genogroup II capsid sequence diversity revealed by analyses of the prototype Snow Mountain agent. Arch. Virol 142:1469-1479.

571 572 573

32. Hardy, M. E., T. N. Tanaka, N. Kitamoto, L. J. White, J. M. Ball, X. Jiang, and M. K. Estes. 1996. Antigenic mapping of the recombinant Norwalk virus capsid protein using monoclonal antibodies. Virology 217:252-261.

574 575 576 577

33. Kitamoto, N., T. Tanaka, K. Natori, N. Takeda, S. Nakata, X. Jiang, and M. K. Estes. 2002. Cross-reactivity among several recombinant calicivirus virus-like particles (VLPs) with monoclonal antibodies obtained from mice immunized orally with one type of VLP. J Clin Microbiol 40:2459-2465.

578 579 580

34. Lindesmith, L. C., M. Beltramello, E. F. Donaldson, D. Corti, J. Swanstrom, K. Debbink, A. Lanzavecchia, and R. S. Baric. 2012. Immunogenetic mechanisms driving norovirus GII.4 antigenic variation. PLoS Pathog. 8:e1002705.

581 582 583

35. Parker, T. D., N. Kitamoto, T. Tanaka, A. M. Hutson, and M. K. Estes. 2005. Identification of Genogroup I and Genogroup II broadly reactive epitopes on the norovirus capsid. J Virol 79:7402-7409.

27

584 585 586

36. Parra, G. I., J. Azure, R. Fischer, K. Bok, C. Sandoval-Jaime, S. V. Sosnovtsev, P. Sander, and K. Y. Green. 2013. Identification of a broadly cross-reactive epitope in the inner shell of the norovirus capsid. PLoS One 8:e67592.

587 588 589

37. Parra, G. I., E. J. Abente, C. Sandoval-Jaime, S. V. Sosnovtsev, K. Bok, and K. Y. Green. 2012. Multiple antigenic sites are involved in blocking the interaction of GII.4 norovirus capsid with ABH histo-blood group antigens. J Virol 86:7414-7426.

590 591 592

38. Swanstrom, J., L. C. Lindesmith, E. F. Donaldson, B. Yount, and R. S. Baric. 2013. Characterization of blockade antibody responses in GII.2.1976 SMV infected subjects. J Virol 88:829-37.

593 594 595

39. Yoda, T., Y. Suzuki, Y. Terano, K. Yamazaki, N. Sakon, T. Kuzuguchi, H. Oda, and T. Tsukamoto. 2003. Precise characterization of norovirus (Norwalk-like virus)-specific monoclonal antibodies with broad reactivity. J Clin Microbiol 41:2367-2371.

596 597 598 599 600

40. Yoda, T., Y. Terano, Y. Suzuki, K. Yamazaki, I. Oishi, T. Kuzuguchi, H. Kawamoto, E. Utagawa, K. Takino, H. Oda, and T. Shibata. 2001. Characterization of Norwalk virus GI specific monoclonal antibodies generated against Escherichia coli expressed capsid protein and the reactivity of two broadly reactive monoclonal antibodies generated against GII capsid towards GI recombinant fragments. BMC Microbiol 1:24.

601 602

41. Jiang, X., M. Wang, K. Wang, and M. K. Estes. 1993. Sequence and genomic organization of Norwalk virus. Virology 195:51-61.

603 604 605 606

42. Hale, A. D., S. E. Crawford, M. Ciarlet, J. Green, C. Gallimore, D. W. Brown, X. Jiang, and M. K. Estes. 1999. Expression and self-assembly of Grimsby virus: antigenic distinction from Norwalk and Mexico viruses. Clin Diagn. Lab. Immunol 6:142145.

607 608 609

43. Kou, B., S. E. Crawford, N. J. Ajami, R. Czako, F. H. Neill, T. N. Tanaka, N. Kitamoto, T. Palzkill, M. K. Estes, and R. L. Atmar. 2014. Characterization of crossreactive norovirus-specfic monoclonal antibodies. Submitted for publication.

610 611 612

44. Tanaka, T., N. Kitamoto, X. Jiang, and M. K. Estes. 2006. High efficiency crossreactive monoclonal antibody production by oral immunization with recombinant norwalk virus-like particles. Microbiol Immunol 50:883-888.

613 614 615

45. da Silva, A. K., O. V. Kavanagh, M. K. Estes, and M. Elimelech. 2011. Adsorption and aggregation properties of norovirus GI and GII virus-like particles demonstrate differing responses to solution chemistry. Environ Sci Technol. 45:520-526.

616 617 618 619 620

46. Hansman, G. S., K. Natori, H. Shirato-Horikoshi, S. Ogawa, T. Oka, K. Katayama, T. Tanaka, T. Miyoshi, K. Sakae, S. Kobayashi, M. Shinohara, K. Uchida, N. Sakurai, K. Shinozaki, M. Okada, Y. Seto, K. Kamata, N. Nagata, K. Tanaka, T. Miyamura, and N. Takeda. 2006. Genetic and antigenic diversity among noroviruses. J Gen. Virol. 87:909-919.

621 622 623

47. Li, X., R. Zhou, X. Tian, H. Li, and Z. Zhou. 2010. Characterization of a cross-reactive monoclonal antibody against Norovirus genogroups I, II, III and V. Virus Res 151:142147.

28

624 625 626 627 628

48. Shiota, T., M. Okame, S. Takanashi, P. Khamrin, M. Takagi, K. Satou, Y. Masuoka, F. Yagyu, Y. Shimizu, H. Kohno, M. Mizuguchi, S. Okitsu, and H. Ushijima. 2007. Characterization of a broadly reactive monoclonal antibody against norovirus genogroups I and II: recognition of a novel conformational epitope. J Virol 81:1229812306.

629 630

49. Ambert-Balay, K. and P. Pothier. 2013. Evaluation of 4 immunochromatographic tests for rapid detection of norovirus in faecal samples. J Clin Virol 56:194-198.

631 632

50. Atmar, R. L. and M. K. Estes. 2001. Diagnosis of noncultivatable gastroenteritis viruses, the human caliciviruses. Clin Microbiol Rev 14:15-37.

633 634 635 636

51. Kirby, A., R. Q. Gurgel, W. Dove, S. C. Vieira, N. A. Cunliffe, and L. E. Cuevas. 2010. An evaluation of the RIDASCREEN and IDEIA enzyme immunoassays and the RIDAQUICK immunochromatographic test for the detection of norovirus in faecal specimens. J Clin Virol 49:254-257.

637 638 639 640

52. Morillo, S. G., A. Luchs, A. Cilli, C. D. Ribeiro, S. J. Calux, R. C. Carmona, and M. C. Timenetsky. 2011. Norovirus 3rd Generation kit: an improvement for rapid diagnosis of sporadic gastroenteritis cases and valuable for outbreak detection. J Virol Methods 173:13-16.

641 642 643

53. Debbink, K., L. C. Lindesmith, M. T. Ferris, J. Swanstom, M. Beltramello, D. Corti, A. Lanzavecchia, and R. S. Baric. 2014. Within host evolution results in antigenically distinct GII.4 noroviruses. J Virol 88:7244-55.

644 645 646

54. Lindesmith, L. C., E. F. Donaldson, A. D. Lobue, J. L. Cannon, D. P. Zheng, J. Vinje, and R. S. Baric. 2008. Mechanisms of GII.4 norovirus persistence in human populations. PLoS Med 5:e31.

647 648 649 650

55. Lindesmith, L. C., E. F. Donaldson, and R. S. Baric. 2011. Norovirus GII.4 strain antigenic variation. J Virol 85:231-242.

29

Table 1. Reactivity of MAbs against GI and GII VLPs by ELISA. Reactivity by ELISA Mab NV23 NV37 NV3 NV7 NV57 NS941 NS22 F8 F120

Route Organ Immunogen po SP NV po SP NV po SP NV po SP NV po SP NV po SP NV + SMA po SP NV + SMA po SP KAV po SP KAV Positive reactivity below the limit of detection

GI.1

GI VLPs GI.4 GI.6

GI.7

GII.2

GII.3

GII.4

GII VLPS GII.6 GII.7

GII.12

GII.17

Mapping broadly reactive norovirus genogroup I and II monoclonal antibodies.

Noroviruses are responsible for most acute nonbacterial epidemic outbreaks of gastroenteritis worldwide. To develop cross-reactive monoclonal antibodi...
5MB Sizes 4 Downloads 7 Views