JGV Papers in Press. Published February 9, 2015 as doi:10.1099/vir.0.000083

Journal of General Virology Recent vaccine development for human metapneumovirus --Manuscript Draft-Manuscript Number:

JGV-D-14-00071R1

Full Title:

Recent vaccine development for human metapneumovirus

Article Type:

Review

Section/Category:

Animal - Negative-strand RNA Viruses

Corresponding Author:

Xiaoyong Bao University of Texas Medical Branch Galveston, TX UNITED STATES

First Author:

Junping Ren

Order of Authors:

Junping Ren Thien Phan Xiaoyong Bao

Abstract:

Human metapneumovirus (hMPV) and its close family member respiratory syncytial virus (RSV) are two major causes of lower respiratory tract infection in the pediatrics population. hMPV is also a common cause of morbidity and mortality worldwide in the immunocompromised patients and older adults. Repeated infections occur often demonstrating a heavy medical burden. However, there is currently no hMPV-specific prevention treatment. This review focuses on the current literatures on hMPV vaccine development. We believe that a better understanding of the role(s) of viral proteins in host responses might lead to efficient prophylactic vaccine development.

Powered by Editorial Manager® and ProduXion Manager® from Aries Systems Corporation

Manuscript Including References (Word document) Click here to download Manuscript Including References (Word document): hMPV virus review JGV-3.docx

1

Recent vaccine development for human metapneumovirus

2

Junping Rena, Thien Phana & Xiaoyong Baoa,b,c *

3

Running Title: hMPV vaccine development

4

Author Affiliations

5

a

6

&Immunity, University of Texas Medical Branch, Galveston, TX.

7

*Correspondence should be addressed to Xiaoyong Bao, Ph.D..

8

Department of Pediatrics, Division of Clinical and Experimental Immunology & Infectious

9

Diseases, 301 University Blvd., Galveston, TX 77555-0372; Fax (409)772-0460;

10

Department of Pediatrics, bInstitute for Translational Science, CInstitute for Human Infections

Tel. (409) 772-1777; Email: [email protected]

11 12 13 14 15 16 17 18

Key Words: hMPV, vaccine, and recombinant hMPV.

19

20

Abstract

Human metapneumovirus (hMPV) and its close family member respiratory syncytial

21

virus (RSV) are two major causes of lower respiratory tract infection in the pediatrics population.

22

hMPV is also a common cause of morbidity and mortality worldwide in the

23

immunocompromised patients and older adults. Repeated infections occur often demonstrating a

24

heavy medical burden. However, there is currently no hMPV-specific prevention treatment. This

25

review focuses on the current literatures on hMPV vaccine development. We believe that a better

26

understanding of the role(s) of viral proteins in host responses might lead to efficient

27

prophylactic vaccine development.

28

Introduction

29

Human metapneumovirus (hMPV), a negative sense single-stranded RNA virus, belongs

30

to the Paramyxoviridae family that also includes respiratory syncytial virus (RSV) and

31

parainfluenza virus (van den Hoogen et al., 2001). Soon after its discovery in 2001, hMPV has

32

been quickly recognized as a frequent cause for lower respiratory tract infections in young

33

children, the immunocompromised patients and older adults (Edwards et al., 2013;Englund et

34

al., 2006;Esper et al., 2004;Falsey et al., 2003). Although hMPV is a clinical important

35

pathogen, no vaccine is currently available.

36

hMPV comprises two genetic groups, A and B. Each group has two sub-genetic classes,

37

i.e. A1 and A2; B1 and B2. Currently, there are five contemporary circulating clades of hMPV,

38

which have existed for decades (Gaunt et al., 2011;Papenburg et al., 2013). Its antigenome

39

contains nine open reading frames for viral protein expression: 3’-N-P-M-F-M2-1-M2-2-SH-G-

40

L-5’. Among them, the nucleoprotein N, phosphoprotein P, and large protein L are essential for

41

RNA synthesis, which is comprised of two independent events: viral replication and gene

42

transcription. Upon entry, the viral RNA-dependent RNA polymerase (RdRp), formed by the L

43

and P proteins, binds the N-encapsidated genome at the leader region, then sequentially

44

transcribes each genes by recognizing start and stop signals flanking viral genes. mRNAs are

45

capped and polyadenylated during synthesis. Replication presumably starts when enough

46

nucleoprotein is present to encapsidate neo-synthetized antigenomes and genomes. The negative-

47

sense genome is replicated into a positive-sense antigenome, which serves as a template for

48

replication of many copies of the viral genome (Bermingham & Collins, 1999;Schildgen et al.,

49

2011;van den Hoogen et al., 2002). In addition to P and L, the M2-2 protein is important for viral

50

RNA synthesis as well. Whether hMPV M2-2 is a key regulatory factor involved in the switch of

51

the viral RNA polymerase from viral gene transcription to viral genome replication like RSV

52

M2-2 protein is still controversial (Buchholz et al., 2005;Kitagawa et al., 2010;Ren et al., 2012).

53

The phosphoprotein P, glycoprotein G, small hydrophobic protein SH, and M2-2 protein have

54

been shown to modulate host immune responses(Bao et al., 2008a;Bao et al., 2008b;Goutagny et

55

al., 2010;Ren et al., 2012;Ren et al., 2014). Recombinant hMPV, lacking G, SH and M-2,

56

individually or in combination, or having P replaced with avian P, are attenuated (Biacchesi et

57

al., 2005;Pham et al., 2005). Fusion protein F is essential for hMPV entry and also induces a

58

strong humoral immune response (Cox et al., 2014). In this review, we will discuss the recent

59

status and efforts for hMPV vaccine development based on the functions of these proteins. We

60

hope all these studies will eventually decrease the medical burden caused by such a clinical

61

important pathogen- hMPV.

62

Inactivated vaccines

63

Formalin-inactivated influenza is commonly used for mass immunization because it is in

64

good stability, easy for manufacturing, and biologically safe due to the absence of viral

65

replication, (http://www.cdc.gov/vaccines/hcp/vis/vis-statements/flu.html). However, the

66

vaccination of a formalin-inactivated human RSV vaccine (FI-hRSV) led to enhanced disease

67

upon natural infection (Kapikian et al., 1969;Kim et al., 1969). Enhanced disease probably

68

resulted from 1) a Th2-biased T cell-memory responses (Boelen et al., 2000;Hussell et al.,

69

1997;Openshaw et al., 1992), 2) formaldehyde hypersensitivity (Moghaddam et al., 2006),

70

and/or 3) immature antibody production and its associated weak recognition of hRSV epitopes

71

from natural infections(Delgado et al., 2009). Recently, decrease in FI-hRSV enhanced disease

72

by RSV G glycoprotein peptide was reported, suggesting the antibody specific to RSV G is

73

critical for RSV pathogenesis control (Rey et al., 2013). Similarly, vaccine-enhanced pulmonary

74

disease and Th2 response following hMPV challenge were also observed in animals vaccinated

75

with formalin-inactivated hMPV (Hamelin et al., 2007;Yim et al., 2007), suggesting that

76

formalin-inactivated hMPV may not be a suitable vaccine candidate.

77

Other inactivation methods are also being investigated for safe vaccine development. For

78

example, a nanoemulsion-adjuvanted inactive RSV has been demonstrated to be able to induce

79

durable RSV-specific humoral responses, decrease mucus production and increase viral

80

clearance, without evidence of Th2 immune mediated pathology (Lindell et al., 2011). In the

81

meanwhile, vaccinated mice exhibited an enhanced Th1/Th17 response. Although the safety of

82

nanoemulsion-adjuvanted inactive RSV vaccine candidate needs to be further investigated

83

(Mukherjee et al., 2011), nanoemulsion inactivation could be applied to hMPV, if nanoemulsion

84

inactivated hMPV is immunogenic and protective, and has balanced immune responses.

85

Viral protein-based vaccines

86

Subunit vaccines are purified or expressed viral proteins, full-length or partial. The

87

expressed proteins are usually in a form of virus-like particles (VLPs), nanoparticles, or with

88

immune-enhancing adjuvants (Anderson et al., 2013). The most immunogenic protein among

89

paramyxoviruses is mainly the fusion protein F. In terms of RSV, a close family member of

90

hMPV, its F in a form of nanoparticle is being evaluated in a phase II clinical trial by Novavax

91

(Driscoll, 2013).

92

Several animal studies using hMPV proteins as subunit vaccine candidates have been

93

recently conducted. By using retroviral core particles as a carrier, intraperitoneal injection of

94

hMPV F induces a strong humoral immune response against both homologous and heterologous

95

strains. Moreover, the induced neutralizing antibodies prevented mortality upon subsequent

96

infection of the lungs with both homologous and heterologous viruses, while hMPV glycoprotein

97

G vaccination did not induce neutralizing activity (Levy et al., 2013). Similar results were

98

observed using an alphavirus replicon- or parainfluenza virus type 3 (PIV3)-based hMPV F

99

vaccine (Mok et al., 2008;Tang et al., 2005). It has been also demonstrated that animals

100

vaccinated by intramuscular injection of adjuvanted soluble hMPV F proteins develop humoral

101

immune response. However, such response diminished rapidly over time (Herfst et al., 2008b).

102

Recently, Dr. Williams’ group demonstrated that hMPV VLPs obtained by expressing matrix

103

(M) and F protein in suspension-adapted human embryonic kidney epithelial (293-F) cells

104

provide immune protection against hMPV replication in the lungs of mice, and are not associated

105

with a Th2-skewed cytokine response, Mice immunized with F-M-VLPs mounted an F-specific

106

antibody response, and generated CD8+ T cells recognizing an F-protein derived epitope. VLP

107

immunization also induced a neutralizing antibody response, which was enhanced by the

108

addition of either TiterMax Gold or α-galactosylceramide adjuvant. All of these suggested that

109

fusion protein-based vaccine is a potential candidate for hMPV vaccine development (Cox et al.,

110

2014).

111

Other hMPV proteins which have been used for protein-based vaccine development

112

include P and G proteins (Levy et al., 2013;Palavecino et al., 2014). A recombinant bacillus

113

Calmette-Guerin (BCG, a carrier to promote immune response against antigens from other

114

bacterial, parasitic, and viral pathogens) expressing hMPV P protein is able to confer strong

115

effector phenotypes to both CD4+ and CD8+ T cells, which showed protective hMPV immunity

116

equivalent to actively immunized animals. However, several groups have suggested that hMPV

117

G-based subunit did not develop protective antibodies, suggesting hMPV G is not important for

118

immunogenicity (Levy et al., 2013;Ryder et al., 2010;Skiadopoulos et al., 2006). Interestingly,

119

studies using recombinant hMPV lacking G protein (rhMPV-G) suggested that G protein plays

120

an important role in inducing protective immune responses (Biacchesi et al., 2004b). Although

121

the results on the role of G in immunogenicity are still controversial, there are several

122

possibilities may contribute to unsuccessful immunogenicity of G during the single protein

123

immunization process. One possibility is that hMPV G undergoes certain modification on the

124

level of gene and/or protein during the single protein immunization, similar to what has been

125

described for RSV F protein(Yang et al., 2013).Another possibility is that same carriers may

126

have reduced ability to incorporate G than F (Levy et al., 2013). Overall, whether G is important

127

in immunogenicity still needs to be clarified.

128

Overall, the immunization using hMPV F-based subunit vaccine is promising; but more

129

experiments are needed to determine the combination of inoculation routes, carrier forms, and

130

length of F to induce the best immunogenicity efficacy and duration. Since other hMPV proteins

131

are also important for immunogenicity and immune balance, subunit immunization requires more

132

investigation on the effect of immunization on Th1/Th2/Th17 balance.

133

Live attenuated vaccines:

134

Live attenuated vaccines can be divided into two groups: non-recombinant and

135

recombinant. Non-recombinant live attenuated viruses are usually generated by natural

136

mutations/deletions during viral passages in cells with or without experimental stresses such as

137

chemical mutagenesis and cold passage (Crowe, Jr. et al., 1995;Juhasz et al., 1997;Whitehead et

138

al., 1998).The major risk of non-recombinant live attenuated vaccine is its in vivo reversion and

139

recovery of viral pathogenicity and subsequent disease development. Some non-recombinant

140

live-attenuated RSV vaccines have been evaluated in clinical trials, but showed some side effects

141

and also insufficient attenuation (Wright et al., 2000). Temperature-sensitive hMPV strains have

142

been generated recently by the group of Drs. Fouchier and Osterhaus. Immunized hamsters

143

showed protective immunity (Herfst et al., 2008a).

144

The recombinant live-attenuated viruses are generated from the cells transfected with

145

hMPV cDNA genome, with/without gene modification/deletion, along with plasmids encoding

146

individual proteins essential for forming RdPp complex (Bao et al., 2008b;Biacchesi et al.,

147

2004a;Ren et al., 2012). The attenuation of recombinant hMPV has been achieved by the

148

deletion of certain accessory genes. They are recombinant hMPV lacking G (rhMPV-∆G), G and

149

SH (rhMPV-∆G/SH), and M2-2 (rhMPV-∆M2-2) (Biacchesi et al., 2004b;Biacchesi et al.,

150

2005;Buchholz et al., 2005). In infected hamsters, rhMPV-∆G and rhMPV-∆G/SH were at least

151

40-fold and 600-fold restricted in replication in the lower and upper respiratory tract,

152

respectively, compared to wild-type rhMPV. However, in rodent model, rhMPV lacking SH

153

alone (rhMPV-ΔSH) replicated somewhat more efficiently in hamster lungs when compared to

154

wild-type (WT)-rhMPV, indicating that SH is completely dispensable in vivo and that its deletion

155

does not confer an attenuating effect. In infected African green monkeys, the attenuation of

156

rhMPV-∆M2-2 reached higher level than that of rhMPV-∆G, and had induce comparable

157

immunogenicity and protective efficiency (Biacchesi et al., 2005).There is another attenuated

158

recombinant hMPV whose P protein was replaced with avian MPV P protein (rhMPV-Pavian).

159

Recently, a wild type recombinant hMPV has been approved to be a suitable parent virus for

160

development of live-attenuated hMPV vaccine candidates in experimental human infection trial

161

(Talaat et al., 2013). Although rhMPV-Pavian was found to be poorly infectious in healthy

162

adults (Schmidt, 2011), the mechanisms associated with poor infectivity of rhMPV-Pavian and

163

whether other attenuated recombinant viruses have a similar human infectivity issues need to be

164

investigated in the future.

165

Other factors should be considered in designing future vaccines.

166

Although F protein is believed to be a major factor determining the immunogenicity of

167

hMPV, identification of viral antigens that activate both protective cytotoxic T-lymphocyte

168

(CTL) and humoral responses are still necessary to develop a successful vaccination strategy.

169

Indeed, several CTL peptides have been proved to be important for CD8+ CTL responses to

170

hMPV challenge. These peptides are 164VGALIFTKL172 from N for H-2b mice, 56CYLENIEII64

171

from M2-2 protein for H-2d mice, and 35KLILALLTFL44 from SH protein and 32SLILIGITTL41

172

from G protein for HLA-A*0201 transgenic mice. Vaccination with these hMPV CTL epitopes

173

upregulates expression of Th1-type cytokines in the lungs and peribronchial lymph nodes of

174

hMPV-challenged mice, resulted in reduced viral titers and disease in mouse models (Herd et al.,

175

2006). Recently, dominant and subdominant hMPV H-2d epitopes were screened and identified

176

(Melendi et al., 2007). Among 12 selected predicted epitopes, 81GYIDDNQSI89 of M2-1 and

177

307

178

respectively during primary infection. In addition, a wide diversification of H-2d-restricted

179

epitopes, in the memory CTL response after secondary and third infection were also identified,

180

including a subdominant role for the memory CTL response against 56CYLENIEII64 of M2-2.

181

Given the importance of CTL epitopes in the immunogenicity, the deficiency of such epitope(s)

182

by complete gene deletion in live attenuated rhMPV may lead to the reduced ability of rhMPV to

183

induce the immunogenicity. To prolong immunogenicity of F protein-based vaccination or to

184

enhance the immunogenicity of deletion mutants of rhMPV, co-immunizing the host with

185

peptides containing CTL epitopes may be a good option.

186

SPKAGLLSL315 of N were identified to be dominant and subdominant H-2d epitope

In addition, since the epitopes identified in the N and M2-2 proteins are completely

187

conserved, protection afforded by a CTL epitope vaccine is expected to extend to both hMPV

188

strains. Identifying viral proteins which are important for antiviral signaling regulation is also

189

critical in vaccine design. Recently, we identified that some viral proteins, such as G and M2-2,

190

play significant roles in suppressing hMPV-induced host innate immunity (Bao et al., 2008b;Bao

191

et al., 2013;Kolli et al., 2011;Ren et al., 2012). Regarding M2-2 protein, we and others found

192

that it is a protein with multiple functions. It not only regulates the viral gene transcription and

193

viral RNA replication(Buchholz et al., 2005;Ren et al., 2012), but also contains a CTL epitope

194

and targets central adaptors for RIG-I and TLRs (Herd et al., 2006;Kitagawa et al., 2010;Ren et

195

al., 2012;Ren et al., 2014). In addition, M2-2 also plays a significant role in regulating the

196

expression of miRNAs, some of which are important for the expression of immune related genes

197

(Deng et al., 2014). The multi-functions of viral protein(s) raise the need to identify the domains

198

respectively responsible for their function, as it is important for rational design of live attenuated

199

recombinant virus. Recently, we identified that the regulatory domains of M2-2 for viral gene

200

and genome replication are different (Ren et al., 2012). We also identified M2-2 motifs which

201

are responsible for their inhibition on antiviral signaling (manuscript in preparation). All these

202

pieces of information on M2-2 might provide a foundation to design M2-2-based live attenuated

203

vaccine candidates. For example, mutants containing mutations on 1) M2-2’s viral replication

204

domain for replication attenuation purpose, and 2) protein interactive motifs to abolish M2-2’s

205

suppression on antiviral signaling for immunogenicity enhancement. On the other hand, the

206

domains which are important for the transcription of viral genes should not be modified in order

207

to 1) minimize frequent mutations of other viral proteins (Schickli et al., 2008), 2) prevent

208

skewedTh1/Th2 balance (Becker, 2006), and 3) maintain all naïve CTL epitopes for

209

immunogenicity purpose (Herd et al., 2006), Overall, dissecting the functional domains of viral

210

protein is highly desirable for vaccine development.

211

Discussion

212

An efficient vaccine candidate should ideally be safe and more immunogenic and protective than

213

natural hMPV infection, which only launches incomplete immune protection. Studies in cotton

214

rats revealed that immunization with FI-hMPV enhanced pathology in the lungs of animals after

215

subsequent infection with hMPV (Yim et al., 2007), excluding it as a promising candidate.

216

Subunit vaccines seem to induce short protective in response to primary infection (Herfst et al.,

217

2008b). However, they may be useful to boost the immune response in individuals that have

218

previously been exposed to hMPV. In addition, subunit vaccines are promising and safe,

219

especially in the form of non-infectious carrier, to provide certain duration of protection for the

220

risk groups such as premature infants, immunocompromised individuals and the elderly. Current

221

live attenuated hMPV vaccines are promising especially to those infants and young children.

222

However, the balance between a satisfactory degree of attenuation and a satisfactory level of

223

immunogenicity may be difficult to obtain. We are currently exploring the possibilities to

224

identify major immune regulatory protein(s) and associated functional motifs with an aim to

225

develop vaccine candidates carrying decent attenuation and immunogenicity. Overall, a variety

226

of vaccination strategies have been explored and tested in rodent models and non-human primate

227

models. However, none has yet been tested in human volunteers. Recent experimental infection

228

of adults with recombinant wild-type hMPV have been performed, demonstrating possibilities to

229

test the immune efficiency of live attenuated recombinant hMPV in human in the future

230

231

Acknowledgements

232

All authors concur there are no conflicts of interest associated in this published work. This work

233

was supported by grants from the National Institutes of Health-National Institute of Allergy and

234

Infectious DiseasesR56 AI107033-01A1, Research Pilot Grant from John Sealy Memorial

235

Foundation, UTMB, the American Lung Association RG232529N, and American Heart

236

Association 12BGIA12060008 to X.B.

237

Reference

238 239 240 241

Anderson, L. J., Dormitzer, P. R., Nokes, D. J., Rappuoli, R., Roca, A. & Graham, B. S. (2013).Strategic priorities for respiratory syncytial virus (RSV) vaccine development. Vaccine 31 Suppl 2, B209-B215.

242 243 244

Bao, X., Kolli, D., Liu, T., Shan, Y., Garofalo, R. P. & Casola, A. (2008a).Human metapneumovirus small hydrophobic protein inhibits NF-kappaB transcriptional activity. J Virol 82, 8224-8229.

245 246 247

Bao, X., Kolli, D., Ren, J., Liu, T., Garofalo, R. P. & Casola, A. (2013).Human metapneumovirus glycoprotein G disrupts mitochondrial signaling in airway epithelial cells. PLoS ONE 8, e62568.

248 249 250

Bao, X., Liu, T., Shan, Y., Li, K., Garofalo, R. P. & Casola, A. (2008b).Human metapneumovirus glycoprotein G inhibits innate immune responses. PLoS Pathog 4, e1000077.

251 252 253

Becker, Y. (2006).Respiratory syncytial virus (RSV) evades the human adaptive immune system by skewing the Th1/Th2 cytokine balance toward increased levels of Th2 cytokines and IgE, markers of allergy-a review. Virus Genes 33, 235-252.

254 255 256

Bermingham, A. & Collins, P. L. (1999).The M2-2 protein of human respiratory syncytial virus is a regulatory factor involved in the balance between RNA replication and transcription. Proc Natl Acad Sci U S A 96, 11259-11264.

257 258 259 260

Biacchesi, S., Pham, Q. N., Skiadopoulos, M. H., Murphy, B. R., Collins, P. L. & Buchholz, U. J. (2005).Infection of nonhuman primates with recombinant human metapneumovirus lacking the SH, G, or M2-2 protein categorizes each as a nonessential accessory protein and identifies vaccine candidates. J Virol 79, 12608-12613.

261 262 263

Biacchesi, S., Skiadopoulos, M. H., Tran, K. C., Murphy, B. R., Collins, P. L. & Buchholz, U. J. (2004a).Recovery of human metapneumovirus from cDNA: optimization of growth in vitro and expression of additional genes. Virology 321, 247-259.

264 265 266 267

Biacchesi, S., Skiadopoulos, M. H., Yang, L., Lamirande, E. W., Tran, K. C., Murphy, B. R., Collins, P. L. & Buchholz, U. J. (2004b).Recombinant human Metapneumovirus lacking the small hydrophobic SH and/or attachment G glycoprotein: deletion of G yields a promising vaccine candidate. J Virol 78, 12877-12887.

268 269 270 271

Boelen, A., Andeweg, A., Kwakkel, J., Lokhorst, W., Bestebroer, T., Dormans, J. & Kimman, T. (2000).Both immunisation with a formalin-inactivated respiratory syncytial virus (RSV) vaccine and a mock antigen vaccine induce severe lung pathology and a Th2 cytokine profile in RSV-challenged mice. Vaccine 19, 982-991.

272 273 274 275

Buchholz, U. J., Biacchesi, S., Pham, Q. N., Tran, K. C., Yang, L., Luongo, C. L., Skiadopoulos, M. H., Murphy, B. R. & Collins, P. L. (2005).Deletion of M2 gene open reading frames 1 and 2 of human metapneumovirus: effects on RNA synthesis, attenuation, and immunogenicity. J Virol 79, 6588-6597.

276 277 278

Cox, R. G., Erickson, J. J., Hastings, A. K., Becker, J. C., Johnson, M., Craven, R. E., Tollefson, S. J., Boyd, K. L. & Williams, J. V. (2014).Human Metapneumovirus Viruslike Particles Induce Protective B and T cell Responses in a Mouse Model. J Virol.

279 280 281 282 283

Crowe, J. E., Jr., Bui, P. T., Siber, G. R., Elkins, W. R., Chanock, R. M. & Murphy, B. R. (1995).Cold-passaged, temperature-sensitive mutants of human respiratory syncytial virus (RSV) are highly attenuated, immunogenic, and protective in seronegative chimpanzees, even when RSV antibodies are infused shortly before immunization. Vaccine 13, 847-855.

284 285 286 287 288

Delgado, M. F., Coviello, S., Monsalvo, A. C., Melendi, G. A., Hernandez, J. Z., Batalle, J. P., Diaz, L., Trento, A., Chang, H. Y., Mitzner, W., Ravetch, J., Melero, J. A., Irusta, P. M. & Polack, F. P. (2009).Lack of antibody affinity maturation due to poor Toll-like receptor stimulation leads to enhanced respiratory syncytial virus disease. Nat Med 15, 34-41.

289 290 291

Deng, J., Ptashkin, R. N., Wang, Q., Liu, G., Zhang, G., Lee, I., Lee, Y. S. & Bao, X. (2014).Human metapneumovirus infection induces significant changes in small noncoding RNA expression in airway epithelial cells. Mol Ther Nucleic Acids 3, e163.

292 293 294

Edwards, K. M., Zhu, Y., Griffin, M. R., Weinberg, G. A., Hall, C. B., Szilagyi, P. G., Staat, M. A., Iwane, M., Prill, M. M. & Williams, J. V. (2013).Burden of human metapneumovirus infection in young children. N Engl J Med 368, 633-643.

295 296 297 298

Englund, J. A., Boeckh, M., Kuypers, J., Nichols, W. G., Hackman, R. C., Morrow, R. A., Fredricks, D. N. & Corey, L. (2006).Brief communication: fatal human metapneumovirus infection in stem-cell transplant recipients. Ann Intern Med 144, 344349.

299 300 301

Esper, F., Martinello, R. A., Boucher, D., Weibel, C., Ferguson, D., Landry, M. L. & Kahn, J. S. (2004).A 1-year experience with human metapneumovirus in children aged

Recent vaccine development for human metapneumovirus.

Human metapneumovirus (hMPV) and respiratory syncytial virus, its close family member, are two major causes of lower respiratory tract infection in th...
433KB Sizes 0 Downloads 18 Views