JVI Accepts, published online ahead of print on 12 March 2014 J. Virol. doi:10.1128/JVI.03101-13 Copyright © 2014, American Society for Microbiology. All Rights Reserved.

1

Mucosal immunization with a candidate universal influenza vaccine

2

reduces virus transmission in a mouse model

3 4

Graeme E. Price1, Chia-Yun Lo1, Julia A. Misplon1 and Suzanne L. Epstein1

5 6

1

7

Food and Drug Administration, Rockville, Maryland, USA.

Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research,

8 9

Address correspondence to Graeme E. Price, [email protected]

10 11

Running Title:

12

NP+M2 vaccine reduces influenza transmission in mice

13

1

14

Word Counts:

15

Running Title (with spaces):

52 characters

17

Abstract:

208 words

18

Main Text:

5179 words

19

References:

64

20

Display Items:

4 tables, 4 figures

21

Supplementary Material:

None

16

22

2

23

ABSTRACT

24

Pandemic influenza is a major public health concern, but conventional strain-matched vaccines

25

are unavailable early in a pandemic. Candidate “universal” vaccines targeting viral antigens

26

nucleoprotein (NP) and matrix 2 (M2), which are conserved among all influenza A virus strains

27

and subtypes, could be manufactured in advance for use at the onset of a pandemic. These

28

vaccines do not prevent infection, but can reduce disease severity, deaths, and virus titers in the

29

respiratory tract. We hypothesized that such immunization may reduce virus transmission from

30

vaccinated, infected animals. To investigate this hypothesis, we studied mouse models for direct

31

contact and airborne transmission of H1N1 and H3N2 influenza viruses. We established

32

conditions under which virus transmission occurs, and showed that transmission efficiency is

33

determined in part at the level of host susceptibility to infection. Our findings indicate that virus

34

transmission between mice has both airborne and direct contact components. Finally, we

35

demonstrated that immunization with recombinant adenovirus vectors expressing NP and M2

36

significantly reduced transmission of virus to co-housed, unimmunized mice in comparison to

37

controls. These findings have broad implications for the impact of conserved antigen vaccines,

38

not only in protecting the vaccinated individual but also in protecting others by limiting

39

influenza virus transmission and potentially reducing the size of epidemics.

40 41

IMPORTANCE

42

Using a mouse model of influenza A virus transmission, we demonstrate that a candidate

43

“universal” influenza vaccine both protects vaccinated animals from lethal infection and reduces

44

transmission of virus from vaccinated to non-vaccinated mice. This vaccine induces immunity

45

against proteins conserved between all known influenza A virus strains and subtypes, so it could 3

46

be used early in a pandemic before conventional strain-matched vaccines are available and

47

could potentially reduce spread of infection in the community.

4

48

INTRODUCTION

49

The possibility that newly emergent influenza viruses may become efficiently transmissible

50

among humans is a major public health concern. Thus, studies of influenza virus transmission

51

and development of medical countermeasures, including antiviral drugs and vaccines, are of

52

high priority. Current influenza vaccines generate strain-specific antibodies against the highly

53

variable viral hemagglutinin and neuraminidase. However, production of strain-matched

54

vaccines requires several months, limiting their utility against new, rapidly spreading viruses. In

55

contrast, cross-protective vaccines targeting conserved antigens such as nucleoprotein (NP) and

56

matrix 2 (M2) do not rely on strain identification and could be used off the shelf early in an

57

outbreak. They protect animals from lethal challenge with a broad range of influenza A virus

58

strains and subtypes, including H1N1, H3N2 and H5N1 (1-8). While these vaccines do not

59

prevent infection, they significantly reduce morbidity, mortality, and viral titers in the

60

respiratory tract (6,7). A potential concern about infection-permissive vaccines is that although

61

they would protect the vaccinee, they might not control transmission to others.

62 63

A recent epidemiological modeling study suggests that reducing virus transmission could

64

dramatically reduce the size of influenza pandemics or seasonal epidemics, and slow the

65

antigenic evolution of seasonal influenza viruses (9). Thus a critical question to test empirically is

66

whether cross-protective vaccines provide that reduction in transmission.

67 68

Animal models for influenza virus transmission include ferrets and guinea pigs (10-13), which

69

efficiently transmit infection to either co-housed animals (direct contact) or across a perforated 5

70

barrier (either aerosol or respiratory droplet transmission). While experiments in these species

71

have elucidated viral and environmental factors affecting host-range and transmission (14-17),

72

studies have been limited by space requirements, expense, and lack of reagents for

73

immunological studies. For practical reasons it is generally not possible to use sufficient

74

numbers of ferrets or guinea pigs for statistical analyses in transmission experiments (18).

75 76

In contrast, mice have been critical for defining host defenses against influenza viruses, and

77

many immunological reagents are available. A mouse influenza transmission system was

78

developed by Schulman and Kilbourne 50 years ago (19-24). Mice have since been seldom used

79

to study influenza virus transmission, although a recent report described contact-dependent

80

transmission in mice (25). Here we have established a model allowing transmission experiments

81

with H1N1 and H3N2 influenza viruses, and accommodating group sizes that permit statistical

82

analyses. We demonstrate that transmission efficiency is in part dependent on the susceptibility

83

of contact animals, and show that limiting exposure to the airborne route alone results in a

84

significantly lower frequency of transmission than when mice are co-housed. We then use this

85

model to demonstrate that immunization with a combination of recombinant adenovirus

86

vectors expressing NP and M2, a candidate universal influenza vaccine, reduces viral

87

transmission from vaccinated to co-housed, unvaccinated animals in the absence of neutralizing

88

antibodies.

6

89

MATERIALS AND METHODS

90

Experimental animals and animal housing. CFW mice [Crl:CFW(SW)] were obtained from

91

Charles River Laboratories, DBA/2J mice were purchased from the Jackson Laboratory (Bar

92

Harbor, ME) and BALB/cAnNCR mice were purchased from the National Cancer Institute

93

(Frederick, MD). All animals were females aged 8-15 weeks at time of use. For experiments using

94

mechanically ventilated housing, mice were housed 5 per cage in standard shoebox cages with

95

microisolator lids (191 × 292 × 127 mm), in a Smart Bio-Pak rack (model SB4100, Allentown

96

Caging Co., Allentown, NJ) with water supplied by an automated system and positive pressure

97

ventilation rate set at a factory-defined 60 air changes per hour. For non-ventilated housing,

98

mice were housed in the same sized shoebox cages with water bottles and microisolator lids on

99

an open-sided shelf with no exhaust system, just room ventilation.

100 101

For experiments examining airborne transmission (defined here as transmission by either

102

aerosols or droplets), contact and donor mice were separated using dividers manufactured to

103

our specifications by Ancare (Belmore, NY), and further modified to ensure a tight fit to the sides

104

of the cage (D.P. Cook Construction, Gaithersburg, MD). Each divider consisted of two

105

perforated stainless steel sheets held 5 mm apart by spacers, with angled flanges at either end

106

of the divider to prevent direct contact between mice at the corners. Each sheet had 532

107

perforations 3.5 mm in diameter and 71 perforations 7 mm in diameter allowing air to exchange

108

between the two compartments. The 7 mm diameter perforations were arranged so as to be

109

inaccessible to mice when the cages were assembled. The divider runs the length of a standard

110

mouse, separating it into two compartments of ~90 × 295 mm. Dividers were held in place with

7

111

a wire grid (Ancare) providing water bottles and feed for each of the compartments. Cages were

112

topped with standard microisolator lids and further secured with elastic bands.

113 114

As a humane end point, mice losing >25% of body weight were euthanized. All experiments used

115

standard mouse bedding (Paperchip, Shepherd Specialty Papers, Watertown, TN), which was

116

not changed during the transmission period. Food and water were provided ad libitum. All

117

animal experiments were conducted under temperature- and humidity-controlled (mean daily

118

values: 22.3 ± 0.1 °C, 51.02 ± 0.1 % humidity) ABSL-2 conditions in animal facilities accredited by

119

the Association for Assessment and Accreditation of Laboratory Animal Care International. All

120

animal protocols and procedures were approved by Institutional Animal Care and Use

121

Committees at the Center for Biologics Evaluation and Research

122 123

Influenza viruses. Mouse-adapted A/FM/1/47-ma (H1N1) [A/FM] originally provided by Earl

124

Brown (University of Ottawa, Canada) (26), the mouse-adapted A/Philippines/2/82 × A/PR/8/34

125

(H3N2) reassortant [X-79] (27), A/PR/8/34 (H1N1) [PR8], and A/Udorn/307/72 (H3N2) [A/Udorn]

126

were prepared as previously described (28). Mice were infected intranasally (i.n.) under

127

isoflurane anesthesia in a 50 µl volume with a dose of 104 50% Tissue Culture Infectious Doses

128

(TCID50), unless stated otherwise.

129 130

rAd vaccines. Production and titration of recombinant adenovirus (rAd) vectors expressing

131

influenza A nucleoprotein (NP), influenza B nucleoprotein (B/NP) or consensus M2 has been 8

132

described previously (3,4). For immunization experiments, a dose of 5×109 particles each of NP-

133

rAd and M2-rAd, or 1×1010 particles of B/NP-rAd in a 50 µl volume were given i.n. under

134

isoflurane anesthesia.

135 136

Lung sampling, BAL, and nasal washing. Mice were euthanized with ketamine/xylazine. Nasal

137

washes were obtained by inserting an intravenous catheter (24 gauge × ¾”, Terumo Medical

138

Corp., Elkton MD) into the nasopharynx via a slit in the trachea and injecting PBS into the nasal

139

cavity until 200 µl of flow-through was collected from the nares. Lungs were then harvested. To

140

prevent cross-contamination at the harvest stage, dissections were performed on contact

141

animals before donor animals in each cage. Instruments were cleaned and sterilized by

142

immersion of contact surfaces in 70% ethanol between each animal. Mucosal sampling for lung

143

cells and bronchoalvelolar lavage (BAL) was otherwise as described (6).

144 145

Virologic analyses. Virus titers in lung tissues and nasal washes were determined by TCID50

146

assay on MDCK cells (29), read out by hemagglutination with 0.5% chicken erythrocytes as

147

described (6). Titers were calculated using the method of moving averages and Weil’s tables

148

(30,31). Assay limits of detection were 102.68 TCID50/organ and 102.19 TCID50/ml for lung and

149

nasal washes, respectively. Contact animals were considered positive for virus transmission if

150

virus was detected by TCID50 assay in either lung homogenate, nasal wash, or both.

151

9

152

50% Mouse Infectious Dose (MID50) determination. Groups of 5 mice were intranasally

153

inoculated with each dilution of virus in a 50 µl volume under isoflurane anesthesia and

154

euthanized 3 days later for collection of lung and nasal wash samples. Animals were considered

155

infected if virus was detected by TCID50 assay in either lung homogenate, nasal wash, or both.

156

MID50 with 95% confidence intervals were calculated using the method of moving averages as

157

described above.

158 159

Immunologic analyses. ELISAs for antibody against the M2 ectodomain (M2e) and recombinant

160

NP have been previously described (5,6). Quantitation of interferon-γ (IFN-γ) secreting cells by

161

ELISPOT was performed as described (4), except that peptide pools covering the M2 and NP

162

sequences of PR8 were used. The M2 peptide pool was a series of 13-15-mer peptides with 11

163

amino acid overlap. The NP peptides (15-17-mer with 11-13 amino acid overlap) were divided

164

into 3 pools of 27-29 peptides each. Peptides were synthesized by Genscript (Piscataway, NJ).

165 166

Hemagglutination inhibition assay. Sera were incubated overnight at 37ºC with 3 volumes of

167

receptor-destroying enzyme (RDE; Denka Seiken Co. Ltd, Tokyo, Japan), and heat inactivated at

168

56ºC for 30 minutes before addition of 6 volumes 0.85% saline. Two-fold dilutions of sera were

169

made in microtiter plates for a final volume of 25 μl and an equal volume of A/FM diluted to 4

170

HAU/25 µl was added to each well and mixed thoroughly. One hour later, 50 µl of 0.5% chicken

171

erythrocytes were added and mixed well. Plates were scored after 45 min, and

172

hemagglutination inhibition (HAI) titers were calculated. Polyclonal anti-A/Fort Monmouth/1/47

10

173

(H1N1) antiserum (NR-3099) obtained through the NIAID, NIH Biodefense and Emerging

174

Infections Research Resources Repository was used as a positive control.

175 176

Microneutralization Assay. Serial two-fold dilutions of RDE treated sera in PBS were made in a

177

96 well U-bottom plate. An equal volume of A/FM at 1×104 TCID50/ml was added to each well,

178

gently mixed, and incubated at 37 ºC. After 2 hours, 20 µl of the virus-antibody mixture was

179

transferred to plates of MDCK cells plus virus growth medium, so as to give 100 TCID50 per well.

180

Plates were then incubated for 48 hours and read out by hemagglutination with 0.5% chicken

181

erythrocytes.

182 183

Statistical Analyses. SigmaPlot 12 (Systat Software, Point Richmond, CA) was used for statistical

184

analyses as follows: Survival analysis used the log rank method, virus titer comparisons used the

185

t- test, transmission rate comparisons used the Chi-Square test.

11

186

RESULTS

187

Establishment of transmission system. In preliminary experiments, CFW outbred mice (as used

188

by Schulman and Kilbourne (21)) were infected with a virulent mouse-adapted influenza H1N1

189

strain, A/FM. Twenty-four hours later, 2 infected donor mice were placed in the same cage as 3

190

uninfected contact animals. 3 days later, all mice were euthanized and lungs and nasal washes

191

collected for virus titration. During the contact period, donors but not contacts progressively

192

lost weight. While all donors had virus recoverable from lungs and most from nasal wash, A/FM

193

was detectable in only 2/18 (11%) contacts when cages were held on a mechanically ventilated

194

cage rack (data not shown), but this rose to 6/18 (33%) of contacts when cages were housed on

195

an open-sided shelf with room ventilation only (Table 1). There was no obvious correlation in

196

either experiment between individual donor lung or nasal wash virus titers on day 4 and the

197

number of co-housed contacts that became infected. This suggests that virus shedding from

198

donors earlier than day 4 is more important for virus transmission, and/or that other factors are

199

involved. Because of the higher rate of transmission observed, all further experiments used

200

cages housed under room ventilation conditions.

201 202

In a further experiment using CFW mice, contacts were monitored for weight loss and survival.

203

All A/FM infected donors died between days 3 and 5 (Fig. 1). Significant weight loss was seen in

204

2 contacts (both in the same cage), but all survived. One month post contact, 4/18 contacts

205

(including the two that lost weight) had A/FM-specific antibody responses with HAI titers of 160-

206

640. Using seroconversion as a marker for transmission, only half the transmitted infections

207

caused clinically apparent disease and insufficient virus was transmitted to initiate a lethal

12

208

infection. This experiment also suggests that secondary transmission (i.e. between infected and

209

uninfected contacts) is either inefficient or absent in the case of A/FM.

210 211

Effect of mouse and virus strain. As different mouse strains vary in their susceptibility to

212

influenza viruses, we extended our studies to investigate direct contact transmission in two

213

additional mouse strains: DBA/2J, which are highly susceptible to lethal influenza virus infection,

214

and BALB/c, which are relatively resistant (32,33). Transmission experiments in these two mouse

215

strains using 104 TCID50 of A/FM were conducted as described above. Transmission of A/FM

216

between DBA/2J mice occurred to a similar extent as seen between CFW mice, but transmission

217

between BALB/c mice was not seen (Table 1).

218 219

To investigate whether the lack of transmission of A/FM in BALB/c mice was due to decreased

220

susceptibility to infection or because of an impaired ability of BALB/c mice to transmit the virus,

221

a “criss-cross” transmission experiment was performed, in which BALB/c mice were used as

222

donors with CFW mice as recipients, and vice versa. Limited transmission of A/FM was seen for

223

CFW donors with BALB/c contacts, but significantly higher levels of transmission were seen for

224

BALB/c donors with CFW contacts (Table 1). A similar experiment examining transmission

225

between the two susceptible stains, DBA/2J and CFW, showed similar rates of transmission with

226

both donor/contact combinations (Table 1). Taken together, these findings suggest that

227

establishment of infection in contact animals is at least in part dependent on the relative

228

sensitivity of the contact animal. However, donor traits also seem to be involved, as

229

transmission of A/FM is more efficient from BALB/c donors to CFW contacts than from CFW 13

230

donors to CFW contacts. Interestingly, virus titers in lungs and nasal washes of infected BALB/c

231

and CFW mice were very similar over the first 4 days of infection (data not shown).

232 233

Limited transmission (2/18 contacts becoming infected; data not shown) between BALB/c mice

234

occurred for two different mouse-adapted viruses (PR8 and X-79). These viruses were also

235

inefficiently transmitted between CFW mice (1/18 contacts becoming infected; data not shown).

236 237

Taken in combination, our data suggest that transmission efficiency is multifactorial and

238

dependent on sensitivity of contacts, donor characteristics, and biological properties of the

239

specific virus strain.

240 241

Mode of transmission. To investigate whether transmission was via a direct contact-dependent

242

route or via the airborne route (by either droplet or aerosol), we developed a removable

243

perforated divider running the length of the cage, as described in Materials and Methods. This

244

prevents direct contact between donor and contact mice while permitting air exchange between

245

the two cage compartments. Using CFW mice and the non-mouse adapted strain A/Udorn,

246

which has been reported to efficiently transmit between BALB/c mice by direct contact but not

247

via the airborne route (25), we found that direct contact resulted in efficient transmission (Table

248

2, Experiment 1). However, when donor and contact mice were physically separated by the

249

perforated divider, a significantly lower proportion of contacts became infected (approximately

250

6-fold, P

Mucosal immunization with a candidate universal influenza vaccine reduces virus transmission in a mouse model.

Pandemic influenza is a major public health concern, but conventional strain-matched vaccines are unavailable early in a pandemic. Candidate "universa...
3MB Sizes 2 Downloads 3 Views