JVI Accepts, published online ahead of print on 9 April 2014 J. Virol. doi:10.1128/JVI.00100-14 Copyright © 2014, American Society for Microbiology. All Rights Reserved.

1

Development of a high yield live attenuated H7N9 influenza vaccine that provides

2

protection against homologous and heterologous H7 wild-type viruses in ferrets

3 4 5 6 7 8 9 10

Running title: Live attenuated H7N9 influenza vaccines in ferrets

11

Celia Santos2, Kanta Subbarao2, Hong Jin1, Yumiko Matsuoka2

12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34

Zhongying Chen1#, Mariana Baz 2, Janine Lu1, Myeisha Paskel2,

1 2

MedImmune LLC, 319 North Bernardo Ave., Mountain View, CA, USA Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, MD 20892, USA

Keywords: Influenza, H7N9, vaccines, ferrets.

#

Corresponding author: Zhongying Chen, PhD MedImmune LLC Mountain View, CA Phone: 650-603-2481 Fax: 650-603-3481 Email: [email protected] Abstract word #: 196 Text word#: 4317

1

35

ABSTRACT

36 37

Live attenuated H7N9 influenza vaccine viruses that possess the hemagglutinin

38

(HA) and neuraminidase (NA) gene segments from the newly emerged wild-type (wt)

39

A/Anhui/1/2013 (H7N9) and six internal protein gene segments from the cold-adapted

40

influenza virus A/Ann Arbor/6/60 (AA ca) were generated by reverse genetics. The

41

reassortant virus containing the original wt A/Anhui/1/2013 HA and NA sequences

42

replicated poorly in eggs. Multiple variants with amino acid substitutions in the HA head

43

domain that improved viral growth were identified by viral passage in eggs and MDCK

44

cells. The selected vaccine virus containing two amino acid changes (N133D/G198E) in

45

the HA improved viral titer by more than 10-fold (reached a titer of 108.6 Fluorescent

46

Focus Units/mL) without affecting viral antigenicity. Introduction of these amino acid

47

changes into an H7N9 PR8 reassortant also significantly improved viral titers and HA

48

protein yield in eggs. The H7N9 ca vaccine virus was immunogenic in ferrets. A single

49

dose of vaccine conferred complete protection of ferrets from homologous wt

50

A/Anhui/1/2013 (H7N9) and near complete protection from heterologous wt

51

A/Netherlands/219/2013 (H7N7) challenge infection. Therefore, this H7N9 LAIV

52

candidate has been selected for vaccine manufacture and clinical evaluation to protect

53

humans from wt H7N9 virus infection.

54 55 56 57

IMPORTANCE In response to the recent avian H7N9 influenza virus infection in humans, we developed a live attenuated H7N9 influenza vaccine (LAIV) with two amino acid

2

58

substitutions in the viral HA protein that improved vaccine yield by 10-fold in chicken

59

embryonated eggs, the substrate for vaccine manufacture. The two amino acids also

60

improved the antigen yield for inactivated H7N9 vaccines, demonstrating that this finding

61

could great facilitate the efficiency of H7N9 vaccine manufacture. The candidate H7N9

62

LAIV was immunogenic and protected ferrets against homologous and heterologous

63

wild-type H7 virus challenge, making it suitable for use in protecting humans from H7

64

infection.

3

65

INTRODUCTION

66 67

Avian influenza A viruses pose a threat of influenza pandemics because most

68

people are serologically naive toward most hemagglutinin (HA) and neuraminidase (NA)

69

subtypes. Avian influenza H7 subtype viruses have caused occasional human infection

70

since 1959 (1-4). In 2003, an outbreak of a highly pathogenic avian influenza (HPAI)

71

H7N7 virus in poultry farms in the Netherlands resulted in 89 cases of human infection

72

including one fatal case and 3 cases of possible human-to-human transmission (5). In

73

2004, an outbreak of HPAI H7N3 virus infection in 57 poultry workers with

74

conjunctivitis or influenza-like symptoms was reported in Canada (6, 7). In 2012, HPAI

75

H7N3 infection in two poultry workers was reported during H7N3 outbreaks in Mexican

76

poultry (8). From February 2013, a novel avian-origin H7N9 subtype influenza virus

77

emerged in China causing severe lower respiratory tract disease in humans (9). A total of

78

135 human cases including 45 deaths occurred in the first wave from February to May

79

2013 (including 2 cases in July). From October 2013 a second wave of human infection

80

has been occurring that caused 240 cases including 70 deaths as of February 28, 2014

81

(http://www.who.int/influenza/human_animal_interface/influenza_h7n9/140225_H7N9R

82

A_for_web_20140306FM.pdf ). Most cases occurred among middle-aged and older

83

adults who had direct exposure to poultry (10, 11). Although cross-reactive antibodies

84

against influenza viruses may exist, the pre-existing antibodies against the novel H7N9

85

virus were not detectable in any age group (12). The H7N9 virus possesses several

86

genetic features contributing to its ability to infect humans (9, 13, 14). Structural and

87

receptor binding analyses have demonstrated that the H7N9 viruses bind to both avian-

4

88

like α2,3-linked sialic acid (SA) receptors and mammalian-like α2,6-linked SA

89

receptors. The Q226L change, which has been associated with reduced binding to α2,3-

90

SA and increased binding to α2,6- SA (15, 16), and other residues in the H7N9 HA

91

contribute to this receptor binding specificity (12, 17-19). Although sustained human-to-

92

human transmission has not been reported, the H7N9 virus can be transmitted via aerosol

93

in ferrets, raising concerns about its pandemic potential (20-22).

94

Several strategies have been used to develop vaccines against avian influenza

95

viruses (23). Live attenuated influenza vaccines (LAIVs) bearing the HA and NA of the

96

viruses of interest and remaining genes from the cold-adapted A/Ann Arbor/6/60 ca virus

97

(AA ca) have several potential advantages as pandemic vaccines. LAIVs are based on

98

licensed technology, can be produced at high yield, and elicit antibodies (systemic and

99

mucosal) and cell-mediated immune responses (24, 25). We have generated an H7N7

100

(A/Netherlands/219/2003, NL03) and H7N3 (A/chicken/British Columbia/CN-6/2004,

101

BC04) LAIV viruses that induced cross-reactive antibody responses in animals (mice,

102

ferrets and monkeys) that conferred protection against challenge with either homologous

103

or heterologous H7 viruses (26, 27). In addition, the H7N3 LAIV evaluated in a Phase I

104

clinical trial was shown to be immunogenic in humans (28). We recently showed that

105

ferret antisera against these H7 ca viruses had cross-reactivity to the H7N9 virus (29).

106

The cross-reactivity between divergent H7 viruses was also reported for the inactivated

107

virus, recombinant protein or virus-like particle (VLP) vaccines studied in mice (30-32)

108

or humans (33). Another Eurasian lineage H7N3 LAIV reassortant with an alternative

109

internal gene backbone was reported to induce cross-reactive antibodies to H7N9 (34),

110

indicating an H7 LAIV might be protective against a divergent H7 strain.

5

111

In this study, we describe the generation of a live attenuated H7N9 vaccine

112

candidate (H7N9 ca) containing the HA and NA gene segments of the recently emerged

113

H7N9 A/Anhui/1/2013 wt virus and six internal protein gene segments of the AA ca virus

114

by reverse genetics and the identification of critical residues in the HA that improved

115

vaccine virus yield in eggs. The final selected LAIV candidate demonstrated high yield in

116

eggs, good immunogenicity and protection against challenge infection with wt

117

homologous and heterologous H7 viruses in ferrets.

118

6

119

MATERIALS AND METHODS

120 121

Viruses. The HPAI A/Netherlands/219/2003 (NL03, H7N7) and the A/Anhui/1/2013

122

(AH13, H7N9) wt influenza viruses used for the evaluation of the efficacy of the vaccine

123

candidate were kindly provided by Dr. Nancy Cox, Influenza Division, Centers for

124

Disease Control and Prevention (CDC), Atlanta, GA and David Swayne at South East

125

Poultry Research Laboratories (USDA). Virus stocks were propagated in the allantoic

126

cavity of 9- to 11- day-old specific-pathogen-free embryonated hen’s eggs (Charles River

127

Laboratories, North Franklin, CT) at 35°C. The allantoic fluid from eggs was harvested

128

24 h post-inoculation and tested for hemagglutinating activity, and stored at -80°C. The

129

50% tissue culture infectious dose (TCID50) for each virus was determined by titration of

130

serially diluted virus in Madin-Darby canine kidney (MDCK) cells and calculated by the

131

Reed and Muench method (35).

132 133

Generation of H7N9 reassortant viruses by reverse genetics. Viral RNA isolated from

134

egg amplified A/Anhui/1/2013 wt was received from the CDC. The HA and NA gene

135

segments of A/Anhui/1/2013 were amplified from viral RNA by reverse transcription-

136

polymerase chain reaction (RT-PCR) using the primers that are universal to the HA and

137

NA gene end sequences and cloned into the plasmid vector pAD3000 (36). Site-directed

138

mutagenesis was performed to introduce specific changes into the HA genes using the

139

QuikChange® Site-Directed Mutagenesis kit (Agilent Technologies, Santa Clara, CA)

140

and the HA sequence was confirmed. The 6:2 reassortant vaccine viruses were generated

141

by co-transfecting eight cDNA plasmids encoding the HA and NA of the H7N9 virus and

7

142

the 6 internal protein gene segments of the AA ca master donor strain into co-cultured

143

293T and MDCK cells. At 3 to 5 days post-transfection, the transfected cell supernatants

144

were inoculated into 10- to 11-day-old embryonated hen’s eggs and incubated at 33°C for

145

64±4 hours. Virus titers were determined by the fluorescence focus assay using an anti-

146

NP monoclonal antibody and expressed as log10FFU (fluorescent focus units)/mL or by

147

plaque assay in MDCK cells as previously described (37). The HA and NA sequences of

148

the rescued viruses were verified by sequencing cDNAs amplified from viral RNA by

149

RT-PCR.

150

The 6:2 PR8 reassortant viruses, containing the HA and NA protein gene

151

segments from the H7N9 virus and the 6 internal protein gene segments from the PR8

152

strain, were generated by plasmid rescue as described above.

153 154

Ferret studies. The ferret studies were conducted in AAALAC certified facilities under

155

protocols approved by Institutional Animal Care and Use Committee (IACUC) at

156

MedImmune for the vaccine virus replication and immunogenicity studies and Southern

157

Research Institute for the H7 wt challenge studies.

158

To evaluate the immunogenicity of the H7N9 ca variants, groups of 3 individually

159

housed 8-12 week old male or female ferrets from Simonson (Gilroy, CA) were

160

inoculated intranasally (i.n.) with 107 FFU of virus in 0.2 mL. Ferrets were bled 14 days

161

post-immunization (p.i.) and sera were assessed for antibody titers by a hemagglutination

162

inhibition (HAI) assay (37).

163

To assess the replication of the H7N9 ca vaccine candidate, ferrets were

164

inoculated with the vaccine virus as described above. Three days after inoculation, virus

8

165

titers in the nasal turbinates (NT) and lungs were determined by egg infectivity and

166

expressed as 50% egg infectious dose (EID50) per gram of tissue.

167

To evaluate the protective efficacy of the H7N9 vaccine candidate, groups of 15-

168

to 16-week-old female ferrets (N=4) that were seronegative for antibodies to circulating

169

H3N2 and H1N1 human influenza viruses were immunized i.n. with 1 (Day 28) or 2

170

doses (Day 0 and day 28) of 107 FFU of H7N9 ca or PBS (mock immunized) in 0.2 mL

171

and serum samples were collected on days 0, 14, 28, 42 p.i.. The animals were transferred

172

to an animal biological safety level 3 (ABSL3) facility (Southern Research Institute,

173

Frederick, MD) for challenge infection with wt H7 viruses. On day 55 or day 56 p.i, sera

174

were collected and the ferrets were challenged i.n. with 107 TCID50 of the homologous

175

A/Anhui/1/2013 (H7N9) or heterologous A/Netherlands/219/2003 (H7N7) wt viruses,

176

respectively. The serum antibody response against homologous and heterologous wt H7

177

viruses was determined in a microneutralization assay (26). The animals were euthanized

178

5 days post-challenge and NTs and lungs (right middle lobe and the caudal portion of the

179

left cranial lobe) were harvested. Tissue homogenates of NTs and lungs were titrated in

180

MDCK cells and the virus titers were expressed as TCID50 per gram of tissue.

181 182

Assessment of viral protein yield. The H7N9 PR8 reassortant viruses were propagated

183

in 25 embryonated hen’s eggs as described above. Virus in allantoic fluid was purified by

184

sucrose gradient centrifugation. The virus band was collected, pelleted by

185

ultracentrifugation and resuspended in 1 mL of the NTE buffer (10mM TrisCl, 100mM

186

NaCl, 10mM EDTA, pH7.5). Total protein was quantitated with a BCA assay kit from

9

187

Pierce (Rockford, IL). An equal volume (25 μl) of each purified virus was

188

electrophoresed on a 4-20% SDS-PAGE followed by Coomassie Blue staining.

189

10

190

RESULTS

191 192 193

Generation of A/Anhui/1/2013 reassortant vaccine variants The HA and NA of vRNA isolated from egg grown wt A/Anhui/1/2013 virus was

194

sequenced. The HA gene contained egg adaptation sequence changes at positions 133,

195

135 and 158 (H3 numbering throughout the paper) compared to the HA sequence of wt

196

A/Anhui/1/2013 from a human specimen (GISAID accession # EPI439507) (Table 1).

197

The sequence of the NA gene was identical to that from the same specimen (GISAID

198

accession # EPI439509). The RT-PCR amplified HA and NA gene segments were cloned

199

and each clone was sequenced. From 20 HA clones analyzed, 6 variants were isolated:

200

V1 (5%) contained the same sequence as the original wt sequence, V2 (45%) had a

201

N158D change, V3 (25%) had a N133D/N158D double mutation, V4 (5%) had a N133D

202

change, V5 (15%) and V6 (5%) contained single mutations of A135T and N199D,

203

respectively. Reassortant vaccine viruses with the HA plasmid of each HA variant

204

together with the NA plasmid of A/Anhui/1/2013 and the 6 internal protein gene

205

plasmids from A/Ann Arbor/6/60 (AA ca) were obtained. The variants had different

206

levels of replication in eggs, with titers ranging from 107.2 to 108.3 FFU/mL (Table 1). V1

207

with the wt HA sequence grew poorly in eggs with a titer of only 107.2 FFU/mL and

208

formed tiny plaques in MDCK cells (Figure 1). V2 (N158D) and V3 (N133D/N158D)

209

with the indicated egg adaptation changes in the HA had the highest titers in eggs (108.2

210

and 108.3 FFU/mL respectively), indicating that the N158D change greatly improved the

211

vaccine virus growth in eggs. V4 and V6 with a single mutation of N133D or N199D in

11

212

the HA had titers higher than V1 but lower than 108 FFU/mL. The V5 variant (A135T),

213

which acquired a potential glycosylation site at N133, reached a titer of 108.1 FFU/mL.

214

V1, V4 and V6 had lower titers than V2, V3 and V5, and formed small or mixed

215

plaques in MDCK cells (Figure 1). After another round of egg passage, the titer of V4

216

improved by acquiring an additional G198E mutation in the HA. Further egg passages of

217

V1 and V6 produced mixed plaque sizes. Viruses with larger plaque size were isolated,

218

amplified in eggs and the HA sequences were determined. The following amino acid

219

changes in the HA were identified: N224D in V6, A160T, R220S or K193N in V1. Each

220

of these identified mutations was introduced into the HA of V4, V6 or V1 and additional

221

vaccine variants (V7-V11) were rescued. All the variants exhibited higher titers (>108

222

FFU/mL) than the parental viruses, and V7 (N133D/G198E) had the highest titer of 108.6

223

FFU/mL (Table 1).

224 225

HA N133D/G198E changes significantly improved the yield of H7N9 PR8

226

reassortant in eggs

227

A PR8 reassortant (A/Shanghai/2/2013, RG32A) was generated by CDC for

228

manufacture of inactivated H7N9 vaccines. This reassortant contained the 6 internal

229

protein gene segments from A/Puerto Rico/8/34 (PR8) and the HA and NA gene

230

segments from A/Shanghai/2/2013 (H7N9). The HA amino acid sequence of

231

A/Shanghai/2/2013 is identical to A/Anhui/1/2013. To evaluate whether the HA protein

232

yield of the PR8-H7N9 reassortant could be improved by introduction of the

233

N133D/G198E (V7) amino acid substitutions in the HA, 6:2 reassortant influenza viruses

234

comprising the 6 internal protein gene segments from PR8, the NA gene segment from 12

235

A/Anhui/1/2013, and the HA gene segment from A/Anhui/1/2013-V1 or V7 were

236

generated by plasmid rescue and resulting viruses were amplified in eggs. The PR8-V7

237

virus had a titer of 108.9, which was significantly higher than PR8-V1 (108.1) and the CDC

238

reassortant RG32A (107.9). Corresponding to the virus titers in eggs, the PR8-V7 virus

239

had higher viral protein yield than the PR8-V1 and RG32A reassortants (Figure 2). These

240

results demonstrate that the two amino acid substitutions in V7 (N133D/G198E) greatly

241

improved the viral growth and HA protein yield of a PR8 reassortant and the PR8-V7

242

variant could be a candidate for manufacture of inactivated H7N9 vaccines.

243 244

Immunogenicity and antigenicity of the vaccine variants

245

The sequence changes identified in the HA variants are located in the head region

246

of the HA trimer structure which may impact the immunogenicity and antigenicity of the

247

vaccine viruses (Figure 3). To examine the immunogenicity and antigenicity of the

248

variants, ferrets were inoculated intranasally with 107 FFU of each variant in 0.2 mL.

249

Post-immunization serum samples were collected on day 14 and antibody titers were

250

evaluated by HAI assay against the homologous virus and the reference virus V1 using

251

chicken erythrocytes (Table 2). V1 was selected as a reference virus for antigenicity

252

assessment of each virus because V1 contained the wt HA sequence and was confirmed

253

to be antigenically identical to BPL-inactivated wt A/Anhui/1/2013 in HAI (data not

254

shown). The V1, V2, V6, V7 and V11 variants all elicited good antibody titers (GMT

255

HAI titers ≥ 64) against the homologous virus. The V3, V5, V8, V9 and V10 variants

256

induced lower HAI antibody titers (GMT HAI titers 19-40). V2, V3, V10 and V11

257

variants cross-reacted with V1 with titers that were ≥ 4-fold than the titers to the

13

258

homologous viruses, indicating that the N158D, R220S or K193N changes affected viral

259

antigenicity. V5, V6, V7, V8 and V9-post-infection sera cross-reacted well with V1,

260

indicating that the changes of A135T, N199D, N133D, G198E, N224D or A160T in HA

261

did not significantly change viral antigenicity. Based on its high growth in eggs, authentic

262

antigenicity and good immunogenicity, the A/Anhui/1/2013 ca-V7 containing the

263

N133D/G198E substitutions in HA was selected as the final H7N9 ca vaccine seed for

264

manufacture.

265 266

The A/Anhui/1/2013 ca vaccine is attenuated and offers protection against

267

homologous and heterologous wt virus challenge infection in ferrets

268

The A/Anhui/1/2013 ca-V7 (AH13 ca) vaccine virus replicated in the nasal

269

turbinates (NT) of ferrets on day 3 post-vaccination with an average titer of 104.9

270

EID50/mL. A low level of viral titers (less than 2.0 TCID50/mL) was detected in nasal

271

washes within 3 days of vaccination; no titer was detected after 3 days (data not shown).

272

The vaccine virus was not detected in ferret lungs (Fig. 4A). Lung tissues from

273

vaccinated ferrets showed no abnormal histopathology findings (data not shown). These

274

data demonstrated that the AH13 ca vaccine virus had the desired attenuation phenotype.

275

One dose of the AH13 ca vaccine elicited a robust neutralizing antibody response

276

with a GMT of 220 (ranging from 63 to 640) and 63 (ranging from 10 to 202) against the

277

homologous A/Anhui/1/2013 wt and heterologous A/Netherlands/219/2003 (NL03) wt viruses,

278

respectively, on day 28 p.i. (Table 3). In the group of ferrets that received 2 doses of the

279

vaccine virus, the first dose elicited homologous neutralizing antibody with a GMT of 63

280

(ranging from 10 to 113) on day 28 p.i.. The second dose of vaccine further boosted the

14

281

neutralizing antibody response to a GMT of 294 (ranging from 202 to 453). The

282

difference in the neutralizing antibody titers following one dose of vaccine in the two

283

groups (GMT 220 versus 63), may be due to the fact that ferrets are outbred or that at the

284

time of immunization the ferrets that received only one dose of vaccine were a month

285

older than the ferrets that received 2 doses of vaccine. The GMT of cross-neutralizing

286

antibodies against the heterologous NL03 wt virus following one and two doses of the

287

AH13 ca virus were 16 and 87, respectively (Table 3). Overall the anti-AH13 ca ferret

288

antisera cross-reacted to NL03 with titers that were 2-4 fold lower than the homologous

289

neutralizing antibody titers. An HAI assay showed that the anti-AH13 ca antisera

290

similarly cross-reacted to NL03, an NL03 mutant (T135A) without the glycosylation site

291

at HA position 133, as well as a North American H7N3 strain A/British Columbia/CN-

292

6/2004 (BC04), with 2-4 fold titer reduction compared to homologous HAI titers (data

293

not shown), indicating that the unique glycosylation site in NL03 did not affect viral

294

antigenicity and the AH13 ca virus induced broadly cross-reactive antibodies to H7

295

viruses. Consistently, anti-NL03 and anti-BC04 ferret antisera cross-reacted with

296

A/Anhui/1/2013 with approximately 3-fold reduction in neutralization titers (29),

297

demonstrating the cross-reactivity of divergent H7 viruses.

298

The protective efficacy of 1 or 2 doses of the AH13 ca vaccine in preventing the

299

replication of the homologous and heterologous wt challenge viruses in the respiratory

300

tract of ferrets was evaluated on day 5 following challenge, when the challenge viruses

301

were predicted to be present at a high titer in the lungs of mock-immunized ferrets (21).

302

The H7 wt challenge viruses A/Anhui/1/2013 (H7N9) and A/Netherlands/219/2003

303

(H7N7) replicated well in the respiratory tract of mock-vaccinated ferrets with mean

15

304

titers of 106.4 and 107.1 TCID50/g, respectively in the NT, and mean titers of 105.8 and 104.8

305

TCID50/g, respectively in the lungs. Immunization with one or two doses of the AH13 ca

306

vaccine fully or nearly fully protected ferrets from replication of the homologous and

307

heterologous wt, respectively, in both NTs and lungs. In the homologous H7N9 wt

308

challenge group, none of the immunized ferrets had detectable titer in both NTs and

309

lungs. In the heterologous H7N7 wt challenge group, only three ferrets had a titer of 102.0

310

TCID50/g in the NT and one ferret had a titer of 102.0 TCID50/g in the lung (Figure 4B).

311 312

16

313

DISCUSSION

314 315

The novel avian-origin H7N9 virus has been associated with significant morbidity

316

and mortality in humans. This virus possesses several genetic features, including binding

317

to human-like 2,6-SA receptors, a deletion in the NA protein and the E627K mutation in

318

the PB2 protein, that raise concerns about its pandemic potential (9, 12, 14). In contrast to

319

the H7N7 and H7N3 ca vaccine viruses that grew to high titer in eggs (26, 27), the novel

320

H7N9 ca virus did not grow well in eggs. In order to respond to the potential need to

321

immunize people against the H7N9 virus, we identified the HA variants that can be used

322

to produce a high yield vaccine for manufacture. The candidate H7N9 ca vaccine is

323

highly immunogenic and cross-reacts well to divergent H7 viruses. A single dose

324

provides complete protection against wt H7N9 and H7N7 challenge infection in ferrets.

325

The HA residues identified in H1N1pdm and seasonal influenza viruses that

326

improve vaccine virus growth in eggs are generally at or near the receptor binding site

327

(RBS) (38-40). The changes identified in the H7N9 HA at residues 133, 135, 158, 160,

328

193, 198, 199, 220, 224 following egg and MDCK cell passage are also located in

329

proximity of the RBS (Figure 2). Residues 133 and 135 are located in the 130 loop on the

330

side of the RBS, residues 193 and 198 are located in the 190 helix, and the 211 and 215

331

residues are located in the 220 loop (18, 41). We speculate that these changes create an

332

optimized receptor binding structure for host specific viral replication. It was noticed that

333

the N133D, N158D, G198E, N199D and N224D changes resulted in the presence of

334

negatively charged acidic residues aspartate (D) or glutamate (E) on the surface of the

335

HA. The K193N and R220S changes reduce positive charge on the HA trimer surface.

17

336

Similar changes were also identified in the HA of the H1N1pdm viruses, K122E, A189D,

337

N128D, D130E and K212E (H3#), that improved vaccine virus growth in eggs and

338

MDCK cells. Further studies indicated that the acidic residue substitutions in H1N1pdm

339

did not affect viral entry and replication but greatly improved viral spread in the host

340

cells (40). These negatively charged residues possibly decrease HA and sialic acid

341

interaction and thus facilitate the release of progeny viruses from infected cells for

342

efficient multi-cycle replication.

343

Glycosylation of the HA protein affects receptor binding, fusion and antigenicity

344

and has been shown to play important roles in virus replication, host restriction, virulence

345

and transmission (42, 43). The A135T and A160T changes create potential N-linked

346

glycosylation sites at the N133 and N158 residues that improve virus growth in eggs. We

347

speculate the glycosylation may improve virus receptor binding and infectivity in eggs

348

based on similar findings reported for the HPAI A/Netherlands/219/2003 (H7N7) in

349

which glycosylation at N133 increased its binding affinity to avian-type α2-3-linked

350

sialosides (44). It was also reported that glycosylation at HA position 188 (H3# 197) near

351

the receptor binding site increased the virulence of an avian H7N7 strain in chicken (45).

352

On the contrary, the loss of a glycosylation site at HA position 133 of human H3N2

353

viruses is associated with better viral growth in MDCK cells (46). For the H5N1 viruses,

354

the loss of the glycosylation site at HA residue 158 improved viral transmission and

355

vaccine immunogenicity in ferrets (47-49). Thus, the effect of HA glycosylation is strain

356

and host-specific.

357

Amino acid changes at antigenic sites on the surface of the HA molecule could

358

alter viral antigenicity. Thus, each vaccine variant was evaluated for antigenicity in the

18

359

HAI assay. The N158D (V2, V3) and the K193N (V11) changes at antigenic site B, and

360

the R220S (V10) change at antigenic site D affected viral antigenicity of the H7N9 virus.

361

The 158 residue has previously been shown to alter the viral antigenicity in seasonal

362

H3N2 and H1N1pdm viruses (37, 50).

363

One dose of the H7N9 ca vaccine virus induced neutralizing antibody in ferrets

364

that cross-reacted with the H7N7 virus in ferrets. A second dose of the H7N9 ca vaccine

365

greatly boosted serum antibody titers. The immunogenicity of the H7N9 ca vaccine virus

366

was as great or greater than the immunogenicity of the H7N3 or H7N7 ca vaccines that

367

were previously generated and evaluated in our laboratory (26, 27). The poorer

368

immunogenicity of the A/Netherland/219/2003 (H7N7) vaccine virus could be attributed

369

to its 2,3-SA receptor binding specificity. The introduction of Q226L/G228S that

370

increased the 2,6-SA receptor binding improved immunogenicity of the H7N7 ca vaccine

371

virus in ferrets (29). The novel H7N9 HA contains L226, which may contribute to its

372

higher replication in the upper respiratory tract and greater immunogenicity in ferrets.

373

Pandemic vaccines are evaluated for safety and immunogenicity in clinical trials

374

but efficacy data can only come from studies in experimentally infected animals. In this

375

study, we demonstrated that a single dose of the H7N9 ca vaccine conferred complete

376

protection, in both NT and lungs, against homologous wt virus challenge infection and

377

near complete protection against the heterologous wt virus. This finding correlated with

378

the robust neutralizing antibody response induced after one dose of the vaccine. In our

379

previous studies, one dose of the H7N3 or H7N7 ca vaccines conferred protection from

380

pulmonary replication of the homologous and heterologous wt virus challenge but not

381

replication in the upper respiratory tract (26, 27).

19

382

In summary, we generated a high-yield H7N9 vaccine candidate that is highly

383

immunogenic and efficacious in a ferret challenge study. The candidate vaccine with

384

amino acid changes at HA residues 133 and 198 maintained the attenuation phenotype

385

conferred by the six internal protein gene segments of AA ca. Based on this promising

386

preclinical data, this vaccine is currently being evaluated in phase I clinical studies.

387

20

ACKNOWLEDGEMENTS

388 389 390

This study was funded in part by federal funds from Biomedical Advanced

391

Research and Development Authority (BARDA) of the U.S. Department of Health and

392

Human Services (HHS) under the contract No HHSO100201200012I and by the

393

Intramural Research Program of NIAID, NIH. The work is conducted under a

394

Cooperative Research and Development Agreement (CRADA) between MedImmune and

395

NIAID/NIH. We thank Drs. Michael Shaw and Nancy Cox at CDC for providing the

396

H7N9 virus and viral RNA, the staff of the animal care facilities at MedImmune and SRI

397

for their assistance with ferret studies, MedImmune’s strain variant team for their

398

support, Dr. Christopher Cotter for technical assistance, Dr. JoAnn Suzich for critical

399

review of the manuscript.

400

21

401

REFERENCES

402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445

1. 2. 3. 4. 5.

6.

7.

8.

9.

10.

DeLay PD, Casey HL, Tubiash HS. 1967. Comparative study of fowl plague virus and a virus isolated from man. Public Health Rep 82:615-20. Webster RG, Geraci J, Petursson G, Skirnisson K. 1981. Conjunctivitis in human beings caused by influenza A virus of seals. N Engl J Med 304:911. Banks J, Speidel E, Alexander DJ. 1998. Characterisation of an avian influenza A virus isolated from a human--is an intermediate host necessary for the emergence of pandemic influenza viruses? Arch Virol 143:781-7. Kurtz J, Manvell RJ, Banks J. 1996. Avian influenza virus isolated from a woman with conjunctivitis. Lancet 348:901-2. Fouchier RA, Schneeberger PM, Rozendaal FW, Broekman JM, Kemink SA, Munster V, Kuiken T, Rimmelzwaan GF, Schutten M, Van Doornum GJ, Koch G, Bosman A, Koopmans M, Osterhaus AD. 2004. Avian influenza A virus (H7N7) associated with human conjunctivitis and a fatal case of acute respiratory distress syndrome. Proc Natl Acad Sci U S A. 101:1356-61. Hirst M, Astell CR, Griffith M, Coughlin SM, Moksa M, Zeng T, Smailus DE, Holt RA, Jones S, Marra MA, Petric M, Krajden M, Lawrence D, Mak A, Chow R, Skowronski DM, Tweed SA, Goh S, Brunham RC, Robinson J, Bowes V, Sojonky K, Byrne SK, Li Y, Kobasa D, Booth T, Paetzel M. 2004. Novel avian influenza H7N3 strain outbreak, British Columbia. Emerg Infect Dis 10:2192-5. Tweed SA, Skowronski DM, David ST, Larder A, Petric M, Lees W, Li Y, Katz J, Krajden M, Tellier R, Halpert C, Hirst M, Astell C, Lawrence D, Mak A. 2004. Human illness from avian influenza H7N3, British Columbia. Emerg Infect Dis 10:2196-9. Lopez-Martinez I, Balish A, Barrera-Badillo G, Jones J, Nuñez-García TE, Jang Y, Aparicio-Antonio R, Azziz-Baumgartner E, Belser JA, RamirezGonzalez JE, Pedersen JC, Ortiz-Alcantara J, Gonzalez-Duran E, Shu B, Emery SL, Poh MK, Reyes-Teran G, Vazquez-Perez JA, Avila-Rios S, Uyeki T, Lindstrom S, Villanueva J, Tokars J, Ruiz-Matus C, Gonzalez-Roldan JF, Schmitt B, Klimov A, Cox N, Kuri-Morales P, Davis CT, Diaz-Quiñonez JA. 2013. Highly pathogenic avian influenza A(H7N3) virus in poultry workers, Mexico, 2012. Emerg Infect Dis. 19:1531-1534. Gao R, Cao B, Hu Y, Feng Z, Wang D, Hu W, Chen J, Jie Z, Qiu H, Xu K, Xu X, Lu H, Zhu W, Gao Z, Xiang N, Shen Y, He Z, Gu Y, Zhang Z, Yang Y, Zhao X, Zhou L, L iX, Zou S, Zhang Y, Li X, Yang L, Guo J, Dong J, Li Q, Dong L, Zhu Y, Bai T, Wang S, Hao P, Yang W, Zhang Y, Han J, Yu H, Li D, Gao GF, Wu G, Wang Y, Yuan Z, Shu Y. 2013. Human infection with a novel avian-origin influenza A (H7N9) virus. N Engl J Med. 368:1888-1897. Li Q, Zhou L, Zhou M, Chen Z, Li F, Wu H, Xiang N, Chen E, Tang F, Wang D, Meng L, Hong Z, Tu W, Cao Y, Li L, Ding F, Liu B, Wang M, Xie R, Gao R, Li X, Bai T, Zou S, He J, Hu J, Xu Y, Chai C, Wang S, Gao Y, Jin L, Zhang Y, Luo H, Yu H, He J, Li Q, Wang X, Gao L, Pang X, Liu G, Yan Y, Yuan H, Shu Y, Yang W, Wang Y, Wu F, Uyeki TM, Feng Z. 2014. 22

446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491

11.

12.

13.

14.

15. 16. 17. 18. 19. 20.

21.

Epidemiology of Human Infections with Avian Influenza A(H7N9) Virus in China. N Engl J Med 370:520-532. Yu H, Cowling BJ, Feng L, Lau EH, Liao Q, Tsang TK, Peng Z, Wu P, Liu F, Fang VJ, Zhang H, Li M, Zeng L, Xu Z, Li Z, Luo H, Li Q, Feng Z, Cao B, Yang W, Wu JT, Wang Y, Leung GM. 2013. Human infection with avian influenza A H7N9 virus: an assessment of clinical severity. Lancet 382:138-145. Zhou J, Wang D, Gao R, Zhao B, Song J, Qi X, Zhang Y, Shi Y, Yang L, Zhu W, Bai T, Qin K, Lan Y, Zou S, Guo J, Dong J, Dong L, Zhang Y, Wei H, Li X, Lu J, Liu L, Zhao X, Li X, Huang W, Wen L, Bo H, Xin L, Chen Y, Xu C, Pei Y, Yang Y, Zhang X, Wang S, Feng Z, Han J, Yang W, Gao GF, Wu G, Li D, Wang Y, Shu Y. 2013. Biological features of novel avian influenza A (H7N9) virus. Nature 499:500-503. Chen Y, Liang W, Yang S, Wu N, Gao H, Sheng J, Yao H, Wo J, Fang Q, Cui D, Li Y, Yao X, Zhang Y, Wu H, Zheng S, Diao H, Xia S, Zhang Y, Chan KH, Tsoi HW, Teng JL, Song W, Wang P, Lau SY, Zheng M, Chan JF, To KK, Chen H, Li L, Yuen KY. 2013. Human infections with the emerging avian influenza A H7N9 virus from wet market poultry: clinical analysis and characterisation of viral genome. Lancet 381:1916-1925. Kageyama T, Fujisaki S, Takashita E, Xu H, Yamada S, Uchida Y, Neumann G, Saito T, Kawaoka Y, Tashiro M. 2013. Genetic analysis of novel avian A(H7N9) influenza viruses isolated from patients in China, February to April 2013. Euro Surveill. 18:20453. Connor RJ, Kawaoka Y, Webster RG, Paulson JC. 1994. Receptor specificity in human, avian, and equine H2 and H3 influenza virus isolates. Virology 205:1723. Stevens J, Blixt O, Tumpey TM, Taubenberger JK, Paulson JC, Wilson IA. 2006. Structure and receptor specificity of the hemagglutinin from an H5N1 influenza virus. Science 312:404-10. Xiong X, Martin SR, Haire LF, Wharton SA, Daniels RS, Bennett MS, McCauley JW, Collins PJ, Walker PA, Skehel JJ, Gamblin SJ. 2013. Receptor binding by an H7N9 influenza virus from humans. Nature 499:496-499. Yang H, Carney PJ, Chang JC, Villanueva JM, Stevens J. 2013. Structural Analysis of the Hemagglutinin from the Recent 2013 H7N9 Influenza Virus. J. Virol. 87:12433-12446. Dortmans JC, Dekkers J, Wickramasinghe IN, Verheije MH, Rottier PJ, van Kuppeveld FJ, de Vries E, de Haan CA. 2013. Adaptation of novel H7N9 influenza A virus to human receptors. Sci Rep 3:3058. Zhang Q, Shi J, Deng G, Guo J, Zeng X, He X, Kong H, Gu C, Li X, Liu J, Wang G, Chen Y, Liu L, Liang L, Li Y, Fan J, Wang J, Li W, Guan L, Li Q, Yang H, Chen P, Jiang L, Guan Y, Xin X, Jiang Y, Tian G, Wang X, Qiao C, Li C, Bu Z, Chen H. 2013. H7N9 influenza viruses are transmissible in ferrets by respiratory droplet. Science 341:410-4. Zhu H, Wang D, Kelvin DJ, Li L, Zheng Z, Yoon SW, Wong SS, Farooqui A, Wang J, Banner D, Chen R, Zheng R, Zhou J, Zhang Y, Hong W, Dong W, Cai Q, Roehrl MH, Huang SS, Kelvin AA, Yao T, Zhou B, Chen X, Leung GM, Poon LL, Webster RG, Webby RJ, Peiris JS, Guan Y, Shu Y. 2013. 23

492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536

22.

23. 24. 25. 26.

27.

28.

29.

30.

31.

32.

33.

Infectivity, transmission, and pathology of human-isolated H7N9 influenza virus in ferrets and pigs. Science 341:183-6. Belser JA, Gustin KM, Pearce MB, Maines TR, Zeng H, Pappas C, Sun X, Carney PJ, Villanueva JM, Stevens J, Katz JM, Tumpey TM. 2013. Pathogenesis and transmission of avian influenza A (H7N9) virus in ferrets and mice. Nature 501:556-559. Subbarao K, Joseph T. 2007. Scientific barriers to developing vaccines against avian influenza viruses. Nat Rev Immunol 7:267-78. Maassab HF, Bryant ML. 1999. The development of live attenuated coldadapted influenza virus vaccine for humans. Rev. Med. Virol. 9:237-244. Murphy BR, Coelingh K. 2002. Principles underlying the development and use of live attenuated cold-adapted influenza A and B virus vaccines. Viral Immunol 15:295-323. Joseph T, McAuliffe J, Lu B, Vogel L, Swayne D, Jin H, Kemble G, Subbarao K. 2008. A live attenuated cold adapted influenza A H7N3 virus vaccine provides protection against homologous and heterologous H7 viruses in mice and ferrets. Virol. 378:123-132. Min JY, Vogel L, Matsuoka Y, Lu B, Swayne D, Jin H, Kemble G, Subbarao K. 2010. A live attenuated H7N7 candidate vaccine virus induces neutralizing antibody that confers protection from challenge in mice, ferrets, and monkeys. J. Virol. 84:11950-11960. Talaat KR, Karron RA, Callahan KA, Luke CJ, DiLorenzo SC, Chen GL, Lamirande EW, Jin H, Coelingh KL, Murphy BR, Kemble G, Subbarao K. 2009. A live attenuated H7N3 influenza virus vaccine is well tolerated and immunogenic in a Phase I trial in healthy adults. Vaccine 27:3744-3753. Xu Q, Chen Z, Cheng X, Xu L, Jin H. 2013. Evaluation of Live Attenuated H7N3 and H7N7 Vaccine Viruses for Their Receptor Binding Preferences, Immunogenicity in Ferrets and Cross Reactivity to the Novel H7N9 Virus. PLoS One. 8:e76884. Goff PH, Krammer F, Hai R, Seibert CW, Margine I, García-Sastre A, Palese P. 2013. Induction of cross-reactive antibodies to novel H7N9 influenza virus by recombinant Newcastle disease virus expressing a North American lineage H7 subtype hemagglutinin. J. Virol. 87:8235-8240. Klausberger M, Wilde M, Palmberger D, Hai R, Albrecht RA, Margine I, Hirsh A, García-Sastre A, Grabherr R, Krammer F. 2014. One-shot vaccination with an insect cell-derived low-dose influenza A H7 virus-like particle preparation protects mice against H7N9 challenge. Vaccine 32:355-362. Smith GE, Flyer DC, Raghunandan R, Liu Y, Wei Z, Wu Y, Kpamegan E, Courbron D, Fries III LF, Glenn GM. 2013. Development of influenza H7N9 virus like particle (VLP) vaccine: homologous A/Anhui/1/2013 (H7N9) protection and heterologous A/chicken/Jalisco/CPA1/2012 (H7N3) crossprotection in vaccinated mice challenged with H7N9 virus. Vaccine 31:43054313. Krammer F, Albrecht RA, Tan GS, I. M, Hai R, Schmolke M, Runstadler J, Andrews SF, Wilson PC, Cox RJ, Treanor JJ, García-Sastre A, Palese P.

24

537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581

34. 35. 36. 37.

38. 39.

40.

41.

42. 43. 44. 45. 46. 47.

2014. Divergent H7 immunogens offer protection from H7N9 challenge. J. Virol. :Epub ahead of print. Rudenko L, Isakova-Sivak I, Donina S. 2013. H7N3 live attenuated influenza vaccine has a potential to protect against new H7N9 avian influenza virus. Vaccine 31:4702-4705. Reed LJ, Muench H. 1938. A simple method of estimating fifty percent endpoints. Am. J. Hyg. 27:493-497. Hoffmann E, Neumann G, Kawaoka Y, Hobom G, Webster RG. 2000. A DNA transfection system for generation of influenza A virus from eight plasmids. Proc Natl Acad Sci U S A. 97:6108-13. Chen Z, Wang W, Zhou H, Suguitan ALJ, Shambaugh C, Kim L, Zhao J, Kemble G, Jin H. 2010. Generation of live attenuated novel influenza virus A/California/7/09 (H1N1) vaccines with high yield in embryonated chicken eggs. J. Virol. 84:44-51. Chen Z, Zhou H, Jin H. 2010. The impact of key amino acid substitutions in the hemagglutinin of influenza A (H3N2) viruses on vaccine production and antibody response. Vaccine 28:4079-4085. Lu B, Zhou H, Ye D, Kemble G, Jin H. 2005. Improvement of influenza A/Fujian/411/02 (H3N2) virus growth in embryonated chicken eggs by balancing the hemagglutinin and neuraminidase activities, using reverse genetics. J. Virol. 79:6763-6771. Wang W, Lu J, Cotter CR, Wen K, Jin H, Chen Z. 2013. Identification of critical residues in the hemagglutinin and neuraminidase of influenza virus H1N1pdm for vaccine virus replication in embryonated chicken eggs. J. Virol. 87:4642-4649. Shi Y, Zhang W, Wang F, Qi J, Wu Y, Song H, Gao F, Bi Y, Zhang Y, Fan Z, Qin C, Sun H, Liu J, Haywood J, Liu W, Gong W, Wang D, Shu Y, Wang Y, Yan J, Gao GF. 2013. Structures and receptor binding of hemagglutinins from human-infecting H7N9 influenza viruses. Science 342:243-247. Schulze IT. 1997. Effects of glycosylation on the properties and functions of influenza virus hemagglutinin. J. Infect. Dis. 176 S24-28. Cheng X, Zengel JR, Xu Q, Jin H. 2012. Surface glycoproteins of influenza A H3N2 virus modulate virus replication in the respiratory tract of ferrets. Virology 432:91-98. Yang H, Carney PJ, Donis RO, Stevens J. 2012. Structure and receptor complexes of the hemagglutinin from a highly pathogenic H7N7 influenza virus. J. Virol. 86:8845-8852. Perdue ML, Latimer JW, Crawford JM, Perdue ML, Latimer JW, Crawford JM. 1995. A novel carbohydrate addition site on the hemagglutinin protein of a highly pathogenic H7 subtype avian influenza virus. Virol. 213:276-281. Asaoka N, Tanaka Y, Sakai T, Fujii Y, Ohuchi R, Ohuchi M. 2006. Low growth ability of recent influenza clinical isolates in MDCK cells is due to their low receptor binding affinities. Microbes Infect. 8:511-9. Herfst S, Schrauwen EJ, Linster M, Chutinimitkul S, de Wit E, Munster VJ, Sorrell EM, Bestebroer TM, Burke DF, Smith DJ, Rimmelzwaan GF,

25

582 583 584 585 586 587 588 589 590 591 592 593 594 595 596

48. 49.

50.

Osterhaus AD, Fouchier RA. 2012. Airborne transmission of influenza A/H5N1 virus between ferrets. Science 336:1534-1541 Imai M, Kawaoka Y. 2012. The role of receptor binding specificity in interspecies transmission of influenza viruses. Curr Opin Virol. 2:160-167. Wang W, Lu B, Zhou H, Suguitan ALJ, Cheng X, Subbarao K, Kemble G, Jin H. 2010. Glycosylation at 158N of the hemagglutinin protein and receptor binding specificity synergistically affect the antigenicity and immunogenicity of a live attenuated H5N1 A/Vietnam/1203/2004 vaccine virus in ferrets. J. Virol. 84:6570-6577. Jin H, Zhou H, Liu H, Chan W, Adhikary L, Mahmood K, Lee MS, Kemble G. 2005. Two residues in the hemagglutinin of A/Fujian/411/02-like influenza viruses are responsible for antigenic drift from A/Panama/2007/99. Virol. 336:113-119.

26

597 598

Table 1. HA sequences and virus titers of A/Anhui/1/2013 (H7N9) ca variants Amino acid at the HA position H3# (H7#) 6:2 variant

133 (123)

135 (125)

wt

N

A

N

vRNA

N/D

A/T

D/N

158 160 193 198 199 220 (149) (151) (184) (189) (190) (211)

A

K

G

N

R

224 (215)

Titer in eggs (log10FFU/mL)

N

n/a n/a

V1

7.2

V2 V3

D

V4

D

V5

D

8.2

D

8.3 7.7

T*

8.1

V6 V7

D D

E

V8 V9

599 600 601 602 603 604

8.6 D

D

T*

8.2 8.4

V10 V11

7.8

S N

8.3 8.4

The wt virus sequence was downloaded from GISAID database. The vRNA was isolated from egg grown A/Anhui/1/2013. The titers represent the average of at least two independent experiments. a. The amino acid changes from the wt virus are shown. *A135T and A160T changes introduce potential N-glycosylation sites.

27

605 606

Table 2. Immunogenicity and antigenicity of A/Anhui/1/2013 ca HA variants Serum HAI titers (GMT) of ferrets immunized with A/Anhui/1/2013 ca

HA variants: Virus

V1

V2

V3

V5

V6

V7

V8

V9

V10

V11

V1 (wt)

81

16

10

16

32

32

10

32

5

32

Homologous Virus

n/a

128

40

19

64

64

20

40

20

256

607 608

Ferrets (n=3) were inoculated i.n. with 107FFU of the indicated A/Anhui/1/2013 ca HA

609

variants (V1-V11). Serum was collected 14 days p.i. and the serum antibody titers were

610

determined by HAI assay against the homologous virus and the V1 reference virus. The

611

values represent the geometric mean titers (GMT) from three ferrets. n/a: not applicable.

28

612 613 614 615

Table 3. Serum neutralizing antibody responses in ferrets following one or two doses of AH13 ca vaccine

Test antigen

616 617 618 619 620 621 622

Serum neutralizing antibody titers (GMT) in ferrets immunized with the AH13 ca vaccine 2 dosesa

1 dose D14

D28

D14

D28

D42

D56

AH13 (H7N9) wt

30

220

22

63

281

294

NL03 (H7N7) wt

11

63

11

16

99

87

Ferrets (n=8) were inoculated i.n. with 1 or 2 doses of 107 FFU of the AH13 ca vaccine. Serum was collected at the indicated days after the first immunization and assessed by the microneutralization assay. Antibodies were not detected in preimmunization sera and in sera from mock-immunized ferrets. a Ferrets received 2 doses of vaccine administered 28 days apart.

29

623 624

FIGURE LEGEND

625

Figure 1. Plaque morphology of A/Anhui/1/2013 ca variants

626

Viruses were rescued by reverse genetics and propagated once in embryonated hen’s

627

eggs. Plaque assay was performed in MDCK cells and incubated at 33°C for 4 days and

628

stained with crystal violet.

629

(A) A/Anhui/1/2013 ca variants (V1-V6) isolated from vRNA

630

(B) A/Anhui/1/2013 ca variants (V7-V11) with introduced HA sequence changes

631 632

Figure 2. HA protein yield of PR8 reassortants. PR8-A/Anhui/1/2013-V1, PR8-

633

A/Anhui/1/2013-V7 and PR8-A/shanghai/2/2013 (RG32A) were propagated in eggs and

634

the virus titers were indicated on the bottom of each lane. Viral harvest from 25 eggs was

635

purified by sucrose gradient. Purified virus was resuspended in 1mL of NTE and total

636

viral protein was measured and expressed as mg/100eggs. An equal volume of each

637

purified virus was loaded onto a SDS-PAGE for electrophoresis and stained with

638

Coomassie Blue.

639 640

Figure 3. The location of the identified HA residues that improve the growth of H7N9

641

viruses on the HA 3D structure (PDB# 4KOL, only one monomer shown). RBS: receptor

642

binding site.

643 644

Figure 4. A. Attenuation study. Ferrets were inoculated with AH13 ca intranasally with a

645

dose of 107 FFU. After 3 days, the nasal turbinate (NT) and lung tissues were collected

646

and viral titers were expressed as EID50 per gram of tissue. The dashed lines indicate the

647

limit of detection. B and C. Wild-type virus challenge study. Ferrets were inoculated

648

with AH13 ca (H7N9 ca) intranasally with one or two doses of 107 FFU. One month after

649

the final dose of vaccine was administered, ferrets were challenged with wt

650

A/Anhui/1/2013 (H7N9) (B) or wt A/Netherlands/219/2003 (H7N7) (C) viruses. After 5

651

days, the NT and lung tissues were collected and viral titers were expressed as TCID50

652

per gram of tissue. Vaccinated groups had a statistically significant reduction in virus

30

653

titers of the homologous H7N9 (P ≤0.0003) and heterologous H7N7 (P ≤0.05) compared

654

to the titers in the mock-immunized group.

31

Development of a high-yield live attenuated H7N9 influenza virus vaccine that provides protection against homologous and heterologous H7 wild-type viruses in ferrets.

Live attenuated H7N9 influenza vaccine viruses that possess the hemagglutinin (HA) and neuraminidase (NA) gene segments from the newly emerged wild-ty...
2MB Sizes 0 Downloads 3 Views