1 a unique and conserved neutralization epitope in h5n1

59
1 A Unique and Conserved Neutralization Epitope in H5N1 Influenza Viruses 1 Identified by a Murine Antibody against the A/goose/Guangdong/1/96 2 Hemagglutinin 3 Running title: H5N1 HAs with a broadly Neutralizing Antibody 4 5 Xueyong Zhu, a Yong-Hui Guo, b Tao Jiang, c Ya-Di Wang, b Kwok-Hung Chan, d 6 Xiao-Feng Li, c Wenli Yu, a Ryan McBride, e James C. Paulson, e,f Kwok-Yung Yuen, d 7 Cheng-Feng Qin, c Xiao-Yan Che, b and Ian A. Wilson a,g 8 9 Department of Integrative Structural and Computational Biology, The Scripps Research 10 Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA a ; Center for Clinical 11 Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, 12 China b ; State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of 13 Microbiology and Epidemiology, Beijing, 100071, China c ; Department of Microbiology, 14 The University of Hong Kong, Hong Kong Special Administrative Region, China d ; 15 Department of Chemical Physiology, e Department of Cell and Molecular Biology, f 16 Skaggs Institute for Chemical Biology, g The Scripps Research Institute, 10550 North 17 Torrey Pines Road, La Jolla, CA 92037, USA 18 19 Address correspondence to Ian A. Wilson, [email protected] , or Xiao-Yan Che, 20 [email protected] . 21 Xueyong Zhu, Yong-Hui Guo and Tao Jiang contributed equally to this article. 22 Ian A. Wilson, Xiao-Yan Che and Cheng-Feng Qin are co-senior authors. 23 24 JVI Accepts, published online ahead of print on 18 September 2013 J. Virol. doi:10.1128/JVI.01577-13 Copyright © 2013, American Society for Microbiology. All Rights Reserved. on April 8, 2018 by guest http://jvi.asm.org/ Downloaded from

Upload: lethien

Post on 10-Feb-2017

220 views

Category:

Documents


0 download

TRANSCRIPT

1

A Unique and Conserved Neutralization Epitope in H5N1 Influenza Viruses 1

Identified by a Murine Antibody against the A/goose/Guangdong/1/96 2

Hemagglutinin 3

Running title: H5N1 HAs with a broadly Neutralizing Antibody 4

5

Xueyong Zhu,a Yong-Hui Guo,

b Tao Jiang,

c Ya-Di Wang,

b Kwok-Hung Chan,

d 6

Xiao-Feng Li,c Wenli Yu,

a Ryan McBride,

e James C. Paulson,

e,f Kwok-Yung Yuen,

d 7

Cheng-Feng Qin,c Xiao-Yan Che,

b and Ian A. Wilson

a,g 8

9

Department of Integrative Structural and Computational Biology, The Scripps Research 10

Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USAa; Center for Clinical 11

Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, 12

Chinab; State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of 13

Microbiology and Epidemiology, Beijing, 100071, Chinac; Department of Microbiology, 14

The University of Hong Kong, Hong Kong Special Administrative Region, Chinad; 15

Department of Chemical Physiology,e Department of Cell and Molecular Biology,f 16

Skaggs Institute for Chemical Biology,g The Scripps Research Institute, 10550 North 17

Torrey Pines Road, La Jolla, CA 92037, USA 18

19

Address correspondence to Ian A. Wilson, [email protected], or Xiao-Yan Che, 20

[email protected]. 21

Xueyong Zhu, Yong-Hui Guo and Tao Jiang contributed equally to this article. 22

Ian A. Wilson, Xiao-Yan Che and Cheng-Feng Qin are co-senior authors. 23 24

JVI Accepts, published online ahead of print on 18 September 2013J. Virol. doi:10.1128/JVI.01577-13Copyright © 2013, American Society for Microbiology. All Rights Reserved.

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

2

Abstract 25

Despite substantial efforts to control and contain influenza H5N1 viruses, ‘bird flu’ 26

viruses continue to spread and evolve. Neutralizing antibodies against conserved epitopes 27

on the viral hemagglutinin (HA) could confer immunity to the diverse H5N1 influenza 28

virus strains and provide information for effective vaccine design. Here, we report on 29

characterization of a broadly neutralizing murine monoclonal antibody H5M9 to most 30

H5N1 clades and sub-clades that was elicited by immunization with viral HA of 31

A/goose/Guangdong/1/96 (H5N1), the immediate precursor of the current dominant 32

strains of H5N1 viruses. Crystal structures of Fab´ H5M9 with H5 HAs of 33

A/Vietnam/1203/2004 and A/goose/Guangdong/1/96 reveal a conserved epitope in the 34

HA1 vestigial esterase subdomain that is some distance from the receptor binding site, 35

and partially overlaps antigenic site C of H3 HA. Further epitope characterization by 36

selection of escape mutant and epitope mapping by flow cytometry analysis of site-37

directed mutagenesis of HA with yeast cell surface display identified four residues that 38

are critical for H5M9 binding. D53, Y274, E83a and N276 are all conserved in H5N1 39

HAs and are not in H5 epitopes identified by other mouse or human antibodies. Antibody 40

H5M9 is effective in protection of H5N1 virus both prophylactically and therapeutically 41

and appears to neutralize by blocking both virus receptor binding and post-attachment 42

steps. Thus, the H5M9 epitope identified here should provide valuable insights into 43

H5N1 vaccine design and improvement, as well as antibody-based therapies for treatment 44

of H5N1 infection. 45

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

3

Keywords: H5N1 influenza virus, epitope, broadly neutralizing murine antibody, crystal 46

structure, escape mutant, site-directed mutagenesis, vaccine design, passive 47

immunotherapy. 48

49

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

4

Introduction 50

The highly pathogenic H5N1 influenza viruses continue to evolve and cause 51

poultry and occasional human infections. In 1996, an avian H5N1 virus, 52

A/goose/Guangdong/1/96 (GD1), was first isolated from a sick farmed goose in 53

Guangdong Province, China (62), and is believed to be the immediate precursor of the 54

current dominant strain of H5N1 virus that is spreading globally. Hemagglutinin (HA) is 55

the surface glycoprotein responsible for viral binding to host cell, internalization of the 56

virus, and subsequent membrane fusion of the viral and host cell membrane within the 57

endosomal pathway inside the infected cell. HA is also the major antigen on the viral 58

surface and provides the primary neutralizing epitopes for antibodies. The HA genes of 59

the subsequent H5N1 viruses are all related to those of GD1 or similar viruses (38), and 60

considerable genetic variation of HA genes have evolved the viruses into over ten distinct 61

phylogenetic clades (numbers 0-9) and second, third and fourth order subclades 62

(http://www.who.int/influenza/gisrs_laboratory/201101_h5fulltree.pdf), but only four 63

clades have been isolated from humans strains (clades 0, 1, 2, and 7) (1). H5N1 virus 64

infection is considered an avian disease, although there is some very limited evidence for 65

direct human-to-human transmission (55). Since 1997, the H5N1 viruses were 66

transmitted to humans mainly by direct contact with sick poultry with a very high fatality 67

rate of about 60% (http://www.who.int). Although the human H5N1 infection is sporadic 68

and rare, there is still great concern about a future H5N1 pandemic due to its high 69

virulence and lethality, its increasing avian reservoir, and the continued evolution and 70

potential reassortment with other human viruses (29, 42). 71

72

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

5

Current strategies against influenza include antiviral treatment and vaccination. 73

Two classes of small molecule drugs, neuraminidase inhibitors and M2 ion-channel 74

blockers, have been used for prophylaxis and treatment of influenza. Neuraminidase is 75

the only other viral surface glycoprotein and cleaves terminal sialic acid moieties from 76

newly formed virions and host cell receptors. Oseltamivir phosphate (Tamiflu), zanamivir 77

(Relenza), and some other NA inhibitors (17) inhibit the NA activity and prevent the 78

budding of new viruses from infected cells, and are effective against both influenza A 79

and B viruses, including H5N1 viruses. These two NA inhibitors are currently used to 80

treat influenza viruses, but resistance to both drugs are emerging, including resistance of 81

H5N1 viruses to treatment by oseltamivir phosphate (33). Amantadine and rimantadine 82

inhibit viral entry and replication by blocking an ion channel formed by the M2 protein, 83

but both drugs are not currently recommended by the Centers for Disease Control and 84

Prevention for treatment of influenza A viruses because of resistance derived from 85

amino-acid substitutions in M2 proteins 86

(http://www.cdc.gov/flu/professionals/antivirals/antiviral-drug-resistance.htm). 87

From a population and global health perspective, vaccination remains the most 88

effective countermeasure against influenza virus. The ideal influenza vaccine would 89

induce cross-protective cellular and humoral responses, and would be safe and 90

immunogenic in all age groups of the population. In 2007, the first H5N1 human vaccine 91

derived from the A/Vietnam/1203/2004 (VN1203) was approved by the U.S. Food and 92

Drug Administration, and a number of other egg-dependent and egg-independent 93

vaccines are at various stages of development (50). Vaccination is also a major strategy 94

to control H5N1 influenza virus in poultry, such as in China, an inactivated vaccine 95

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

6

containing the HA and NA genes of the GD1 virus and internal genes from the A/Puerto 96

Rico/8/34 (H1N1) has been used for domestic poultry since 2004 (35). This GD1 virus-97

based vaccine was demonstrated to be effective against different H5N1 viruses isolated in 98

China, excepted for the recently isolated, low-pathogenic, A/chicken/Shanxi/2/06–like 99

viruses (clade 7.2) (35). 100

A panel of 15 monoclonal antibodies specific for H5N1 viruses was generated by 101

immunizing mice with viral or recombinant HA of the GD1 virus (34). Among them, one 102

antibody H5M9, elicited in mice from H5 HA that was concentrated from GD1 H5N1 103

virus, had broad neutralizing activity against different clades (effective for all tested 104

clades 0, 1, 2.3.4, and 7) of H5N1 influenza viruses isolated from 1997 to 2008 (listed in 105

Table 1 and in our previous report (34)), indicating the presence of a conserved 106

neutralizing epitope in the H5 HA protein recognized by H5M9. Antibody epitope 107

mapping of H5 HA using yeast cell surface display showed that H5M9 binds to the HA1 108

region (34). 109

To define the H5M9 epitope at the atomic level, we determined crystal structures 110

of the H5M9 Fab´ in complex with the VN1203 HA and GD1 HA ectodomains. The 111

H5M9 antibody primarily recognizes a conserved and previously uncharacterized epitope 112

region in the vestigial esterase subdomain of HA1 (22) between the receptor binding site 113

head region and the HA fusion subdomain. The epitope was further refined by selection 114

of escape mutants and flow cytometry analyses of H5M9 binding to HA and by site-115

directed mutagenesis of key HA binding residues. We previously documented that 116

H5M9 is an inhibitor of hemagglutination as it directly blocks virus binding to cellular 117

receptors (34). We show here that H5M9 is also an inhibitor of the pH-induced 118

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

7

conformational change required for virus fusion activity. These properties can be 119

translated into virus neutralization in vivo since H5M9 is also found to be protective 120

against a lethal viral challenge in a mouse model, both prophylactically and 121

therapeutically. 122

123

MATERIALS AND METHODS 124

Ethics statement. The animal experiments with BALB/c mice were carried out in strict 125

accordance with the guidelines of the Animal Experiment Committee of the State Key 126

Laboratory of Pathogen and Biosecurity, Ministry of Science and Technology of the 127

People’s Republic of China, and were approved by the Animal Experiment Committee of 128

the State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology 129

and Epidemiology, Beijing, China. 130

Cloning, expression and purification of the HAs. The ectodomains of VN1203 131

and GD1 HAs were expressed in the baculovirus system essentially as previously 132

described (15, 52). Briefly, HA0 cDNAs corresponding to residues 11-327 of HA1 and 1-133

174 of HA2 (H3 numbering, 17-504 in H5 numbering) of the VN1203 HA (GenBank 134

accession number AY818135) and GD1 HA (GenBank accession number AF144305) 135

were inserted into a baculovirus transfer vector, pFastbacHT-A (Invitrogen) with a N-136

terminal gp67 signal peptide, a C-terminal trimerization domain and a 6-His tag, and a 137

thrombin cleavage site incorporated to separate the HA ectodomain and the trimerization 138

domain and His tag. The constructed plasmids were used to transform DH10bac 139

competent bacterial cells by site-specific transposition (Tn-7 mediated) to form a 140

recombinant Bacmid with allowed blue-white selection. The purified recombinant 141

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

8

VN1203 HA and GD1 HA Bacmids were used to transfect Sf9 insect cells for 142

overexpression. HA protein was produced by infecting suspension cultures of Hi5 cells 143

with recombinant baculovirus at an MOI of 5-10 and incubation at 28ºC shaking at 110 144

RPM. After 72 hours, Hi5 cells were removed by centrifugation and supernatants 145

containing secreted, soluble HA proteins were concentrated and buffer-exchanged into 146

1xPBS, pH 7.4. The VN1203 and GD1 HAs consisted of a mixture of uncleaved HA0 147

and cleaved HA1/HA2, and were recovered from the cell supernatants by metal affinity 148

chromatography using Ni-NTA resin. The HAs were digested with thrombin to remove 149

the trimerization domain and His-tag. The cleaved VN1203 HA or GD1 HA was purified 150

further by size exclusion chromatography on a Hiload 16/90 Superdex 200 column (GE 151

healthcare) in 10 mM Tris pH 8.0, 50 mM NaCl, and 0.02% (v/v) NaN3. 152

H5M9 Fab´ preparation and purification. Antibody H5M9 was elicited by 153

immunization of mice with HA from A/goose/Guangdong/1/96 as described (34). The 154

Fab´ fragment of antibody H5M9 (IgG1ț) was produced by standard protocols (25). The 155

intact H5M9 IgG was digested to (Fab´)2 with 1% (w/w) pepsin for four hours and 156

followed by reduction to Fab´ by 15 mM 2-mercaptoethylamine (MEA) for two hours. 157

The protein was purified to homogeneity by a combination of protein A and protein G 158

affinity chromatography, as well as ion-exchange chromatography (Mono-Q column, 159

Pharmacia) and gel filtration (Superdex 200 column, GE healthcare). 160

Purification of H5M9 Fab´-VN1203 HA and H5M9 Fab´-GD1 HA complexes. 161

The H5M9 Fab´-VN1203 HA and H5M9 Fab´-GD1 HA complexes were generated by 162

incubating the two purified components with an optimal ratio of Fab´ to HA that was 163

estimated by gel-shift using Blue Native PAGE (Invitrogen). H5M9 Fab´ at 1 mg/ml was 164

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

9

titrated into 5 µg of VN1203 HA and GD1 HA in a total volume of 10 µl. The mixtures 165

were incubated overnight at 4ºC before Blue Native PAGE analysis. Following 166

determination of the optimal molar ratio (roughly 3 Fabs per trimer), H5M9 Fab´ and 167

purified VN1203 HA or GD1 HA in the same buffer (10 mM Tris, pH 8.0, 50 mM NaCl) 168

were mixed with that ratio. The mixture solution was incubated overnight at 4ºC before 169

further purification by gel filtration (Superdex 200 column) to remove unbound Fab´ and 170

HAs. 171

Crystallization and structural determination of H5M9 Fab´-VN1203 HA 172

complex. Crystallization experiments were set up using the sitting drop vapor diffusion 173

method. Initial crystallization conditions for the H5M9-VN1203 HA complex were 174

obtained from robotic crystallization trials using the automated Rigaku Crystalmation 175

system at the Joint Center for Structural Genomics (JCSG). Following optimization, 176

diffraction quality crystals were obtained by mixing 0.5 µl of the concentrated protein in 177

6.8 mg/ml in 10 mM Tris, pH 8.0, 50 mM NaCl with 0.5 µl of a reservoir solution 178

containing 0.1 M Hepes, pH 7.5, 2% (w/v) PEG 4000, 2.2 M (NH4)2SO4 at 22ºC. The 179

crystals were flash-cooled in liquid nitrogen using 30% saturated malonate in the mother 180

liquor as cryoprotectant. Diffraction data of the complex crystals were collected at 100K 181

at beamline 23ID-B, Advanced Photon Source (APS), Argonne National Laboratory. 182

HKL2000 (HKL Research, Inc) was used to integrate and scale diffraction data. The 183

crystals diffract to 3.6 Å resolution and the diffraction data were indexed in space group 184

C2 with a Matthews’ coefficient (Vm) of 6.1 Å3/Da and 80% solvent content (Table 2). 185

The structure was determined by molecular replacement using the program Phaser 186

(39). The initial model for H5M9 was constructed from a light chain variable domain 187

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

10

from PDB model 2VL5 and a heavy chain variable domain the Fab´ constant domain 188

from PDB model 1P7K which share high sequence similarity (with 88% and 74% 189

identities, respectively) with no sequence gaps. The previously determined structure of 190

VN1203 HA (PDB codes 2FK0, 3GBM) was used as the HA model in the complex. One 191

HA trimer and three Fabs were found in the asymmetric unit. The initial rigid body 192

refinement and restrained refinement were performed in program REFMAC5 (41). 193

Despite the modest resolution, the data-to-atom ratio is still reasonable in the refinement 194

due to high solvent content (Table 2). The electron density maps fit the structure model 195

very well with continuous density for all main-chain atoms and good density for most 196

side chains. Additional positive electron density was observed near all 6 potential N-197

glycosylation sites in each HA monomer (18 glycosylation sites per HA trimer). 198

Interestingly, strong positive density was also observed near mouse H5M9 Fab´ AsnL63 199

possessing an Asn-Gly-Ser motif, and a GlcNAc residue was built at AsnL63 in all three 200

Fabs in the asymmetric unit. Structural refinement was completed with program Buster 201

(5) and Phenix (2). Final refinement statistics are summarized in Table 2. 202

Crystallization and structural determination of GD1 HA and H5M9 Fab´-203

GD1 HA complex. The purified GD1 HA in 10 mM Tris, pH 8.0, 50 mM NaCl was 204

concentrated to 8.0 mg/ml and subjected to crystallization trials using the Mosquito 205

crystal liquid handler (TTP LabTech). Initial crystal hits were obtained from Initial 206

Screening Reagents CP-CUSTOM-IV from Axygen Biosciences. The crystals for data 207

collection were grown by the sitting drop vapor diffusion method with a reservoir 208

solution containing 0.1 M Tris, pH 8.2, 21% (w/v) MPEG 2000 at 22ºC. The crystals 209

were flash-cooled in liquid nitrogen using 25% ethylene glycol (v/v) in mother liquor as 210

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

11

cryoprotectant. Diffraction data were collected at 100K at beamline 9-2, Stanford 211

Synchrotron Radiation Lightsource. The crystals diffract to 2.6 Å resolution and the 212

diffraction data were indexed in space group P21 with a Matthews’ coefficient (Vm) of 213

3.4 Å3/Da and 64% solvent content (Table 2). 214

Initial crystallization conditions for the H5M9 Fab´-GD1 HA complex were 215

obtained from the automated Rigaku Crystalmation system at the JCSG. Following 216

optimization, diffraction quality crystals were obtained by mixing 0.5 µl of the 217

concentrated protein in 9.7 mg/ml in 10 mM Tris, pH 8.0, 50 mM NaCl with 0.5 µl of a 218

reservoir solution containing 0.1 M Hepes, pH 7.5, 2% (w/v) PEG 4000, 2.0 M 219

(NH4)2SO4 at 22ºC. The crystals were flash-cooled in liquid nitrogen with cryoprotectant 220

40% saturated malonate added to mother liquor. Diffraction data were collected at 100K 221

at beamline 11-1, Stanford Synchrotron Radiation Lightsource. HKL2000 (HKL 222

Research, Inc) was used to integrate and scale diffraction data. The crystals diffract to 7.0 223

Å resolution and the diffraction data were indexed in space group P3121 with a 224

Matthews’ coefficient (Vm) of 4.3 Å3/Da and 71% solvent content (Table 2). 225

GD1 HA and H5M9-GD1 HA structures were both determined by molecular 226

replacement with the program Phaser (39). The unliganded GD1 HA structure was 227

determined with the VN1203 HA structure (PDB codes 3GBM) as a model. The H5M9 228

Fab´-GD1 HA complex structure was subsequently determined using the refined GD1 229

HA structure and the refined H5M9 Fab´ structure from H5M9-VN1203 HA complex as 230

input models. For GD1 HA structure, the initial rigid body refinement and restrained 231

refinement were performed with program REFMAC5 (41). Additional positive electron 232

density was observed near all 5 potential N-glycosylation sites in the various HA 233

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

12

monomers (for a total of 32 sites observed out of 45 possible sites for three HA trimers). 234

The structural refinement for GD1 HA was completed with program Buster (5) and 235

Phenix (2). For H5M9-GD1 HA complex, the molecular replacement solution resulted in 236

two HA trimers and only one H5M9 Fab´. The other five Fab´ molecules were modeled 237

into electron density by superimposing the relevant HA molecules, and further refinement 238

reduced the R values. Due to the low resolution, only rigid-body refinement was 239

performed with program REFMAC5 (41). Final refinement statistics are summarized in 240

Table 2. 241

The quality of the structures described here was analyzed using the JCSG 242

validation suite (www.jcsg.org) including MolProbity, WHAT IF, Resolve and Procheck. 243

All figures were generated with Pymol (www.pymol.org). 244

KD determination. KD’s were determined by bio-layer interferometry using an 245

Octet Red instrument (ForteBio, Inc.). For VN1203 HA and GD1 HA binding with 246

H5M9 Fab´ and IgG, VN1203 HA or GD1 HA at 20 µg/ml in 1x kinetics buffer (1x PBS, 247

pH 7.4, 0.01% BSA, and 0.002% Tween 20) was loaded onto Ni-NTA coated biosensors 248

and incubated with varying concentrations of VN1203 HA or GD1 HA in 1x kinetics 249

buffer. All binding data were measured at 30 ºC. Five or six concentrations in 1:2 serial 250

dilutions of H5M9 Fab´ or H5M9 IgG were used, with the highest concentration being 25 251

nM, except for 100 nM for H5M9 Fab´ in binding with GD1 HA. The KD reported here 252

was determined from the ratio of koff to kon. All binding traces and curves used for fitting 253

are reported in Figure S1. 254

HA glycan microarray receptor binding assay. Protocols for microarray HA 255

analysis and the list of glycans on the array (Fig. S2) were as previously described (6, 256

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

13

63). Briefly, HA-antibody complexes were prepared by mixing HA, mouse anti-His 257

Alexa Fluor 488 and goat anti-mouse IgG Alexa Fluor, and the complex mixture was 258

then added directly to the surface of the array and allowed to incubate for 1 hour at room 259

temperature (~ 22 ºC). After washing with 1x PBS and Tween, the array slides were dried 260

and scanned for fluorescence signal. 261

Cells and Virus. MDCK cells were maintained in DMEM (Life technologies) 262

supplemented with 10% FBS at 37°C in 5% CO2. After infection with influenza virus, the 263

MDCK cells were maintained in DMEM containing 0.2% (w/v) BSA and 0.5 ȝg/ml 264

TPCK-trypsin (Sigma-Aldrich). All the viruses in this study were propagated in 10-day-265

old embryonated eggs and titered by hemagglutination tests and plaque forming assays. 266

Neutralization assay. Virus neutralization titers of the antibody were determined 267

as described previously (34, 46). 268

Selection of neutralization escape mutants. 103 PFU of H5N1 virus strain 269

A/Vietnam/1194/2004 (H5N1) (VN1194) was mixed with purified H5M9 and incubated 270

at 37 °C for 1 h. The virus-antibody mixture was added to the MDCK cell monolayer and 271

incubated at 37 °C for another 1 h. Following removal of the inoculum, the plate was 272

rinsed twice with PBS, then 3 ml of DMEM containing 2 ug/ml TPCK-trypsin and 0.5 273

ȝg/ml of H5M9 were added. After 4 days of incubation in the presence of H5M9, 274

medium containing potential escape viruses was harvested. After three passages in the 275

presence of H5M9, escape mutants were isolated by plaque-to-plaque purification and 276

further amplified in MDCK cells. The HA segments of escape mutants obtained from 277

each passage were sequenced and analyzed. 278

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

14

Plaque reduction neutralization test. Antibody H5M9 was diluted to 500 ȝg/ml, 279

and then serially 10-fold diluted in DMEM. H5M9 IgG was added with an equal volume 280

of approximately 100 PFU of wild-type H5N1 virus or escape mutant. The antibody-virus 281

mixture was incubated at 37˚C for 1 h, and then added to MDCK monolayers in a 6-well 282

plate in duplicate and incubated for another 1 h at 35˚C. The supernatant was removed, 283

and 3 ml of 1.0% (w/v) LMP agarose (Promega) in DMEM plus TPCK-trypsin (2 ȝg/ml) 284

was layered onto the infected cells. After further incubation at 35˚C for another 4 days, 285

the overlays were removed and the cells were stained with 1% (w/v) crystal violet 286

dissolved in 4% (v/v) formaldehyde to visualize the plaques. The percentage of plaque 287

reduction was calculated accordingly. 288

Epitope mapping of H5M9 by yeast cell surface display. The DNA fragment 289

(49–954 nt) coding the HA protein was amplified from a cDNA clone of VN1194 HA by 290

PCR with BamHI andXhoI sites and ligated into the pYD1 vector with the Xpress epitope 291

tag (DLYDDDDK) (Invitrogen), producing the recombinant plasmid pYD1-HA. The HA 292

protein was expressed in the Saccharomyces cerevisiae strain EBY100 as described in 293

our previous report (34). The potential key HA contacting residues within H5M9 binding 294

site were mutated using the QuikChange® Lightning Site-Directed Mutagenesis Kit 295

(Stratagene, CA, USA). Yeasts expressing wild-type HA protein and mutant HA proteins 296

were identified by sequencing. The expression of mutant proteins on the EBY100 yeast 297

cell surface and fluorescent labeling of yeast surface displayed proteins were performed 298

according to the method described previously (34). Briefly, yeast cells were washed with 299

PBS and incubated with purified antibodies at a concentration of 10 ȝg/ml with 1 mg/ml 300

BSA in PBS. After a 30-minute incubation on ice, yeast were washed in PBS and then 301

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

15

incubated with a goat anti-mouse IgG secondary antibody conjugated to FITC (Sigma) at 302

a dilution of 1:150 with 1 mg/ml BSA in PBS. After fixation with 2% (w/v) 303

paraformaldehyde in PBS, yeast cells were analyzed on a Accuri C6 flow cytometer 304

(Becton-Dickinson) using software Flowjo (www.flowjo.com). 305

Protease susceptibility assay. Protocols for trypsin susceptibility analysis were 306

as previously described (15). For VN1203 HA, each reaction contained ~2.5 ȝg of the 307

HA or ~2.5 ȝg of the HA and a two-fold molar excess of H5M9 Fab´ (2 Fabs per HA 308

protomer). Reactions were incubated at 37 °C for one hour at pH values 4.9 and 8.0, 309

respectively. After incubation, the reaction pH was neutralized to pH 8.4. Trypsin was 310

then added to all samples except controls at a final ratio of 1:20 (wt/wt) of trypsin to the 311

HA, and reactions were incubated overnight at 22°C. Samples were then analyzed by 312

reducing SDS-PAGE. 313

Prophylactic and therapeutic efficacy of H5M9 in mice. In the prophylactic 314

model, groups of 4 week-old female BALB/c mice (n=6) were i.p. injected with a dose of 315

2 or 20 mg/kg of H5M9 one day prior to intranasal challenge with 10 MLD50 of VN1194. 316

In the therapeutic model, mice received the same dose of H5M9 one day after the lethal 317

challenge of 10 MLD50 of VN1194. The mice were monitored daily for survival and 318

weight loss until day 14 post-infection (p.i.). All manipulations involving live H5N1 319

virus were carried out in the Biosafety Level 3 (BSL-3) or Animal Biosafety Level 3 320

(ABSL-3) containment facility in Beijing Institute of Microbiology and Epidemiology. 321

Statistical Analysis. Survival curves were generated by the Kaplan-Meier method 322

and analyzed with Log-rank test. P<0.05 is considered significant. 323

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

16

Accession numbers. The atomic coordinates and structure factors are deposited 324

in the Protein Data Bank under accession codes 4MHH for the H5M9 Fab´-VN1203 HA 325

complex, 4MHI for the unliganded GD1 HA, and 4MHJ for the H5M9 Fab´-GD1 HA 326

complex. Nucleotide sequences for the H5M9 IgG have been deposited in GenBank 327

under accession numbers KF499999 for the light chain and KF500000 for the heavy 328

chain. 329

330

RESULTS 331

The overall structure of H5M9 Fab´-VN1203 HA complex. VN1203 virus was 332

originally isolated from a 10-year old Vietnamese boy who died of bird flu, and this virus 333

is among the most pathogenic H5N1 isolates studied to date in mammalian models (37). 334

The VN1203 HA ectodomain was expressed in a baculovirus system, and H5M9 Fab´ 335

was digested and purified from the mouse antibody (34). The H5M9 binds to the 336

VN1203 HA with high affinity with Kd values of 1 nM and 0.008 nM for binding of the 337

immobilized HA to the monovalent Fab´ and bivalent IgG, respectively (Table 3, Fig. 338

S1). The crystal structure of the H5M9 Fab´-VN1203 HA complex was determined to 3.6 339

Å resolution with good refinement statistics for this moderate resolution, which might in 340

part be due to high solvent content in the crystals (Table 2). The mature HA is a 341

homotrimer with multiple glycosylation sites (Fig. 1A). Each HA polypeptide is 342

proteolytically cleaved by host proteases into two disulfide-linked subunits HA1 and 343

HA2. As in the unliganded VN1203 HA (52), the overall fold of the VN1203 HA trimer 344

in the complex is very similar to other published HAs, with HA1 containing primarily the 345

receptor binding subdomain and vestigial esterase subdomain, and HA2 constituting most 346

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

17

of the core fusion machinery in the stalk region (Figs 1A and B). Interpretable density is 347

observed for VN1203 HA1 (residues 11-324) and HA2 (1-174) (H3 numbering), and 348

H5M9 Fab´. In the complex, the VN1203 HA trimer is in its prefusion state with the N-349

terminal fusion peptide in HA2 embedded in the hydrophobic core between the trimer 350

HA2 subunits, with one H5M9 Fab´ binding to each HA monomer. Significantly, unlike 351

most of other structurally defined neutralizing antibodies, H5M9 binds in the vestigial 352

esterase subdomain (22) of HA1 at the base of the membrane distal domain, at a distance 353

from the receptor binding site (Figs. 1A and B). The antibody H5M9 and VN1203 HA 354

paratope and epitope surfaces interact well with a shape complementarity of 0.48, and as 355

expected, no glycosylation sites are found within the epitope (Fig. 1A). 356

A unique and conserved H5N1 HA epitope from the H5M9 Fab´-VN1203 HA 357

complex. Upon complex formation, one H5M9 Fab´ binds to each H5 HA1 protomer of 358

the trimer (three Fab´s per trimer), burying ~ 820 Å2 of HA protein surface with typical, 359

heavy chain-dominant binding, where 60% of the binding surface arises from the heavy 360

chain. A total of 16 residues from each VN1203 HA monomer participate in the 361

intermolecular contacts. 362

The epitope is conformational and consists of mostly HA1 polar residues from 363

fragments 53 to 62, 78 to 83a, 117 to 119, and 273 to 278 in the vestigial esterase 364

domain, as previously defined by Ha et al. (22). The key HA contacting residues are 365

shown in Figure 1C. Five of the six H5M9 complementarity determining regions (CDRs), 366

i.e. not CDR L3, are involved in HA binding (Fig. 1D). The heavy-chain residues 367

contribute 3 out of 4 hydrogen bonds (H-bonds) or salt bridges, as analyzed by HBPLUS 368

(40), and about 60% of the 110 total van der Waals’ interactions. The light chain of 369

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

18

H5M9 interacts with VN1203 HA1 residues with mainly nonpolar contacts, as well as a 370

salt bridge between ArgL50 and Glu83a. The heavy chain of H5M9 makes mainly 371

nonpolar interactions and three H-bonds with VN1203 HA1, including one H-bond each 372

between SerH98 (OȖ) and Asp53 (Oį2), ThrH28 (OȖ1) and Arg62 (NȘ1), as well as 373

ThrH28 (N) and Glu78 (Oİ2). 374

No major changes are observed in the overall structure of VN1203 HA upon 375

complex formation except for a few side-chain rotamers, such as for Asn278 to avoid a 376

steric clash with the antibody. The CĮ root mean square deviation (rmsd) is 1.3 Å 377

between the VN1203 HA in H5M9 Fab´ complex and the uncomplexed VN1203 HA 378

(PDB code 2FK0), while the CĮ rmsd is only 0.5 Å between the VN1203 HA in the 379

H5M9 Fab´ complex and the VN1203 HA in a complex with a stem binding antibody 380

CR6261 (PDB code 3GBM), where the stem epitope is distant from H5M9 binding site 381

(15). Thus, no large structural changes are required for neutralization to occur, as 382

observed in other influenza neutralizing antibodies, such as the H3 virus neutralizing 383

antibodies HC19 and HC45 (19). 384

Antibody H5M9 binds the same epitope in GD1 HA. As the GD1 H5N1 virus 385

is believed to be the immediate precursor of the dominant strains of H5N1 viruses, it was 386

important to compare a crystal structure of GD1 HA with the VN1203 HA in the 387

VN1203 HA-H5M9 complex to establish the conserved nature of the epitope recognized 388

by H5M9. Thus, we determined the crystal structure of apo-form GD1 HA ectodomain to 389

2.6 Å resolution (Table 2). Three GD1 HA protomers form the canonical trimer (Fig. 2A) 390

with a CĮ rmsd of 0.6 Å between GD1 and VN1203 HA protomers, and a CĮ rmsd of 0.5 391

Å between HA1 domains with a total of 16 amino acid differences (Table 4). The 392

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

19

receptor-binding sites (RBS) of GD1 HA and VN1203 HA are almost identical with 393

conserved key residues except for a Ser221 to Pro221 switch in HA1 from VN1203 HA 394

to GD1 HA at its far edge of the RBS (Fig. 3A), implying the similar receptor binding 395

activity and specificity between two viruses. Indeed, direct analysis by glycan microarray 396

showed that both HAs are highly specific for Į2-3 sialosides (Figs. 4 and S2), with the 397

exception that VN1203 HA acquired additional binding to fucosylated Į2-3 linked 398

glycans. 399

The H5M9 binding sites in VN1203 HA and GD1 HA are well conserved (Fig. 400

3B), which is consistent with the similar Kd values of 0.9 nM for the Fab´ binding to GD1 401

HA and 1.0 nM for VN1203 HA, although the Kd of 0.04 nM for IgG binding to GD1 HA 402

is ten-fold higher that the highly potent 0.008 nM binding to VN1203 HA (Table 3, Fig. 403

S1). A conservative variation at position 55 in HA1 (Asp in VN1203 HA and Asn in 404

GD1 HA) is the only difference within the H5M9 footprint between these two H5N1 405

viruses (Fig. 3B, Table 4) and these changes do not appear to affect H5M9 neutralization 406

as H5N1 viruses with Asp55 or Asn55 in HA can all be neutralized by H5M9 (Tables 1 407

and 5). Asp55 of VN1203 HA lies on the edge of H5M9 epitope and makes van der 408

Waals contacts with both PheL28 and LysL30 of CDR L1. Asn55 of GD1 HA is also on 409

the periphery of the epitope and makes contacts with CDR L1. The low-resolution crystal 410

structure of GD1 HA in complex with H5M9 Fab´ (Table 2) also clearly indicates that 411

H5M9 Fab´ binds within the same site in GD1 HA as that of VN1203 HA (Fig. 2B). 412

Sequence analysis and glycosylation prediction (11) indicates that up to seven 413

glycosylation sites can be present in H5N1 HAs, of which six sites are over 95% 414

conserved (N21, N33, N169 and N289 in HA1, and N154 and N213 in HA2); the other 415

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

20

glycosylation site at N158 in HA1 is 47% conserved. In our H5 HA construct (from 416

residue 11 in HA1 to residue 173 in HA2) expressed in a baculovirus expression system 417

in insect cells, all potential glycosylation sites were found to be glycosylated in both 418

VN1203 HA (Fig. 1A) and GD1 HA (Fig. 2A). By comparison, the GD1 HA has five 419

conserved glycosylation sites (N21, N33, N169 and N289 in HA1, and N154 in HA2), 420

while the VN1203 HA has an additional glycosylation site at N158 that is close to the 421

RBS (Fig. 1A). The loss of glycosylation at N158 in recent H5N1 strains has been 422

associated with increased potential to acquire a receptor specificity that supports aerosol 423

transmission in ferrets (45, 51). However, loss of this glycan alone is not sufficient to 424

change receptor specificity since GD1 exhibits strong binding to α2-3 sialosides that are 425

characteristic of avian virus receptor specificity (Fig. 4). 426

Selection and sequence analysis of escape mutants. The HA epitope was further 427

probed by generating and characterizing escape mutants. Neutralization escape mutants 428

were generated by culturing H5N1 virus, A/Vietnam/1194/2004 (H5N1) (VN1194) in the 429

presence of H5M9. VN1194 HA has only one amino-acid substitution T46K compared to 430

VN1203 HA and this change is distant from the epitope (Table 4). After three rounds of 431

passages, escape variants selected by H5M9 in VN1194 virus exhibited mutations in 432

HA1 D53N (with single G-A change at nucleotide position 177), close to the H5M9 433

epitope center (Fig. 1C). The D53N mutant generated similar plaque morphology and 434

peak titers to that of wild-type VN1194 in MDCK cells. Plaque reduction neutralization 435

tests (PRNT) were then performed to compare the relative infectivity of VN1194 and 436

D53N mutant viruses (Fig. 5A). Wild-type VN1194 virus was neutralized by H5M9 at a 437

PRNT50 value of 0.12 µg/ml. In clear contrast, the D53N mutant virus was not 438

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

21

neutralized by H5M9 even at the highest tested concentration of 500 µg/ml (Fig. 5A). 439

The D53N mutation would change the overall electrostatic potential of the HA surface 440

around position 53 from weakly acid to near neutral and reduce the interactions with 441

H5M9 which is weakly basic around D53 binding site (Figs. 5B to D). Otherwise, a 442

similar hydrogen bond is made with Asp/Asn 53 to SerH98. 443

Identification of key epitope residues by yeast cell surface display. To further 444

explore the key epitope residues for H5M9, a panel of HA single mutants of nine H5 445

contacting residues contributing most of the interface interactions was interrogated for 446

specific binding of antibodies by yeast cell surface display. Alanine scanning 447

mutagenesis was carried out for H5M9 epitope residues 53, 57, 62, 78, 83A, 117, 273, 448

274 and 276 on VN1194 HA with only one amino-acid substitution T46K to VN1203 HA 449

(Table 4). Since the amino acids of these residues on VN1194 HA (or VN1203 HA) are 450

among the most frequently found at these positions in all H5N1 HAs (> 85.9% 451

conservation) from our extensive survey of all non-redundant H5N1 HA sequences in the 452

National Center for Biotechnology Information (NCBI) influenza database (Table 6), 453

nine polar mutations for these residues were also made based on the second most-454

frequent amino acid in H5N1 HAs: D53N, K57R, R62K, E78K, E83aK, H117R, E273K 455

and N276D, as well as Y274F (Table 6). Thus, a total of 18 HA mutants and one wild-456

type control were generated from yeast cell surface display and analyzed by FACS of 457

flow cytometry (Fig. 6). Several of these mutants conferred considerable loss of binding 458

to H5M9. Alanine mutants, D53A, E83aA and Y274A abolished and N276A 459

substantially reduced binding of H5M9 (Fig. 6), indicating the importance of these side 460

chains in H5M9 binding. Furthermore, for other mutants at these four positions, D53N, 461

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

22

E83aK abolished binding of H5M9, while Y274F and N276D showed no obvious effect 462

(Fig. 6). Interestingly, although binding of mutant E78A was unchanged, mutant E78K 463

significantly reduced binding of H5M9 (Fig. 6). Alanine scanning mutants at K57, R62, 464

H117 and E273, as well as other mutants at these positions, K57R, R62K, H117R and 465

E273K, cause no obvious loss of H5M9 binding, indicating these positions are not critical 466

for binding to H5M9. Taken together, FACS analysis revealed that D53 and E83a, as 467

well as Y274 and N276 to a lesser extent, constituted critical contact residues for H5M9 468

binding (Fig. 1C). The escape mutant D53N was confirmed once more to be critical and 469

mutations E83aA and E83aK abolish the salt bridge between E83a to ArgL50 of H5M9. 470

In addition, mutation Y274A would eliminate interactions with GlyH97 and SerH98, 471

while mutation N276A abolishes interactions with ThrH33, PheH52 and SerH98 of 472

H5M9. 473

Mechanism of antibody H5M9 neutralization. We previously reported that 474

antibody H5M9 blocked viral entry in a hemagglutination inhibition test, although the 475

inhibition titer is weaker than those of some other antibodies elicited from the same 476

mouse immunization as H5M9 (34). This lower HAI inhibition for H5M might due to the 477

epitope being distant from the receptor binding site and, hence, the antibody only 478

partially blocking receptor binding (Fig. 1). During virus infection, the HA protein is 479

responsible for viral attachment and viral fusion. To further explore the H5M9 480

neutralization mechanism, trypsin digestion of VN1203 HA was performed in which the 481

HA protein was exposed to a low pH (pH 4.9) to trigger the pH-induced conformational 482

changes and acquire sensitivity to cleavage by trypsin (Fig. 7). Convincingly, the low-pH 483

treated HA could be completely digested in its HA1 domain by trypsin in the absence of 484

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

23

H5M9 Fab´ (lane 1 in Fig. 7). However, H5M9 Fab´ was able to protect the HA from 485

degradation by trypsin when treated at pH 4.9 (lane 3 in Fig. 7). These results suggest the 486

binding of H5M9 somehow inhibits the pH-induced conformational changes in HA, 487

indicating its function in preventing virus fusion. 488

Prophylactic and therapeutic efficacy of antibody H5M9. The in vivo 489

protective effects of H5M9 against H5N1 viral infection were tested in mice using 490

prophylactic and therapeutic models, respectively. In the prophylactic model, female 491

BALB/c mice were i.p. injected with a dose of 2 or 20 mg/kg of H5M9 one day prior to 492

intranasal challenge with 10 MLD50 (50% mouse lethal dose) of VN1194 virus. A single 493

treatment of H5M9 provided full protection against lethal challenge of VN1194 virus in 494

mice (Fig. 8A). All H5M9-treated mice showed increase in body weight without any 495

signs of respiratory distress throughout the study. As expected, all mice that received 496

control PBS treatment showed signs of respiratory distress, rapid weight loss, and 497

succumbed to viral infection (Fig. 8A). In the therapeutic model, a single treatment with 498

20 mg/kg of H5M9 one-day post 10 MLD50 of VN1194 challenge provided 66.7% 499

protection, while 16.7% of mice treated with 2 mg/kg of H5M9 survived (Fig. 8B). 500

Moreover, all surviving mice appeared healthy and showed recovery in body weight after 501

initial infection. Overall, H5M9 treatment significantly increased the mean survival time 502

of infected mice from 7.3 ± 0.5 days (PBS control group) to 12.7 ± 1.9 days (20 mg/kg) 503

and 10.2 ± 2.5 days (2 mg/kg), respectively (Fig. 8B). 504

505

506

507

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

24

DISCUSSION 508

Highly pathogenic avian influenza H5N1 virus can cause morbidity and mortality 509

in humans. So far, the H5N1 virus is mainly panzootic in avian species and leads to 510

occasional human infection through close contact with infected birds. However, recent 511

reports have shown that the H5N1 viruses, including VN1203, could become 512

transmissible through airborne droplets by mutations in H5 HAs so that the HA proteins 513

recognize receptors in the upper airways of mammals, particularly ferrets (9, 26, 28). It 514

is, therefore, necessary to prepare for a potential H5N1 human pandemic. Current 515

strategies against influenza include vaccination and antiviral drug treatment. Due to the 516

existence of multiple clades and subclades of the H5N1 virus, it is difficult to predict the 517

major strain that might cause the next pandemic. In this study, from HA-antibody crystal 518

structures, escape mutant analysis and fine epitope mapping by yeast cell surface display, 519

we identified a unique and conserved epitope in H5N1 viruses using a murine antibody 520

elicited from the H5N1 virus immediate precursor GD1 that has prophylactic and 521

therapeutic efficacy to H5N1 virus. 522

The antibody H5M9 epitope is a unique epitope for H5N1 viruses. The antigenic 523

sites of H5 HA were originally mapped into five groups from an H5N9 HA based on the 524

H3 structure (46). From the crystal structures of H5 HA from A/Duck/Singapore/3/97 525

(H5N1) (23) and VN1203 (52), the H5 HA molecule was then antigenically mapped in 526

greater detail first for an H5N2 avian influenza virus (31), and later for VN1203 (30), 527

revealing two antigenic sites, one of which is similar to site A of H3 HA (59), and the 528

other overlapping site B in H3 (59) and site Sa in H1(8). In addition, the fine epitope 529

mapping of an avian subclade 2.2 of the H5N1 viruses found antigenic sites 530

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

25

corresponding to site B in H3 HA, but extending far beyond this area (49). In this study, 531

crystal structures of H5M9 in complex with both VN1203 HA and GD1 HA revealed an 532

epitope located in the vestigial esterase subdomain of the HA1 head, relatively far from 533

the receptor binding site. Escape mutant analysis with the VN1194 virus selected a 534

mutant D53N, and further characterization by flow cytometry of site-directed 535

mutagenesis of the HA with yeast surface display identified four key epitope residues in 536

VN1194 HA, Asp53, Tyr274, as well as E83a and N276, which are located in the lower 537

part of the H5M9 footprint from the crystal structures (Fig. 1C). These key H5 residues 538

do not overlap with any known epitopes described above or detected with other human 539

and mouse antibodies (7, 12, 18, 24, 27, 32, 36, 43, 44, 47, 48, 54, 56, 60). The four 540

H5M9 epitope key residues do not overlap with known H1 antigenic sites (8), but 541

positions 53, 274, and 276 correspond to site C of H3 viruses (59). However, H5M9 did 542

not show cross-subtype neutralization against H1 and H3 viruses in hemagglutination 543

inhibition tests (34), which is likely due to amino-acid differences in this epitope in other 544

virus subtypes (Table 4). 545

The antibody H5M9 epitope appears to be well conserved in H5N1 viruses and is 546

consistent with H5M9’s cross-neutralization activities. In all H5N1 viruses of clades 0, 1, 547

2.3.4 and 7 that were neutralized by H5M9, the H5M9 epitope residues are fully 548

conserved, except for a D55N mutation that does not appear to affect H5M9 binding or 549

neutralization (Tables 1 and 5). The full or almost completely conserved H5M9 epitope 550

can be found in most other clades or subclades of H5N1 viruses, except for some critical 551

mutations, such as D53N, that exist in clades 7.1 and 7.2 or some other viruses (Table 7). 552

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

26

The antibody H5M9 is likely to have neutralizing activity against a broad 553

spectrum of H5N1 influenza viruses. The H5M9 binding surface is highly conserved 554

(>93.6% conservation) across all H5N1 influenza A viruses, except for lower 555

conservation of Asp53 (85.9%), Asp55 (86.1%) and Arg62 (86.1%) (Table 8). H5 viruses 556

with the Asp55 mutation to Asn (14.0 % conservation) may still possess the 557

neutralization activity as confirmed for H5M9 in our neutralization assay (Tables 1 and 558

5), while Arg62 mutation to Lys (16.5% conservation) would not change binding of 559

H5M9 as illustrated by R62K mutant of VN1194 HA in the yeast cell surface display 560

analysis (Fig. 6). However, the Asp53 mutation to Asn (12.5% conservation) in H5 HA 561

might abolish neutralization and binding of H5M9 as seen in the escape mutant and flow 562

cytometry analyses (Figs. 5 and 6). It remains to be seen if H5M9 can neutralize 563

influenza viruses from subtypes H5N2 to H5N9. From the survey of the conservation of 564

H5M9 epitope in HAs from H5N2 to H5N9 (Table 8), the H5M9 epitope is well 565

conserved in H5N5 HAs, but not in other subtypes as one of the key binding residues 566

D53 is rare and is mostly Ser at position 53. 567

Our results suggest antibody H5M9 binding to this previously uncharacterized 568

epitope in H5 HA can play a role in inhibiting both virus binding and virus fusion, 569

although the epitope is distant from the receptor binding site or the fusion peptide. Most 570

neutralizing antibodies against influenza virus recognize epitopes in the highly variable 571

regions in the HA head around receptor binding site and inhibit virus binding to host cells 572

(19-21, 58, 61) or, in one case, prevent membrane fusion (4). Several recently reported 573

broadly neutralizing antibodies from our group and others bind to the stem at the 574

membrane-proximal end of HA and interfere with the pH-induced conformational 575

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

27

changes of the HA that lead to membrane fusion, as seen for H5 and other HAs (13-16, 576

53). Antibody H5M9 inhibited agglutination of erythrocytes in vitro (34), although it 577

binds to an epitope that is distant from the receptor binding site, but close enough to still 578

interfere with receptor binding. Because of their large size, IgGs bound on the 579

membrane-distal surface of HA may still prevent access to the receptor binding site if 580

oriented towards it. So far, only two other structurally-characterized antibodies, HC45 581

(19) and BH151 (20), bind to a similar but not identical epitope on H3 HA of 582

A/Aichi/2/68 (H3N2) (Fig. 9). It was also reported that HC45 neutralized viral infectivity 583

by blocking receptor binding as confirmed by hemagglutination inhibition assay (19). In 584

addition to prevent virus binding, H5M9 appeared to inhibit post-attachment membrane 585

fusion step from our trypsin susceptibility test at low pH 4.9. Another anti-H5N1 586

antibody AVFluIgG01, which bound to HA1 receptor binding subdomain with potential 587

epitope residues 123-125, 128 and 168, was also reported to have a similar dual 588

neutralization mechanism (7). Interestingly, antibody CR8071 neutralizing influenza B 589

viruses also bound to the vestigial esterase domain but showed no obvious 590

hemagglutination inhibition activity; instead, it interfered with release of progeny virions 591

from infected cells (14). In this case, the CR8071 was oriented perpendicular to the long 592

axis of the HA trimer and away from the RBS. 593

Glycosylation of HA can affect the host specificity, virulence and infectivity of 594

influenza virus. The GD1 HA differed by one glycosylation site with VN1203/VN1194 595

HA, which acquired an additional glycosylation site at N158. For H5N1 HA, N158 and 596

N169 are the only two conserved glycosylation sites in the HA head domain. 597

Computational analysis proposed that the presence of N-glycans at N158 would decrease 598

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

28

the affinity of HA for all human and avian type glycan receptors (11). The removal of 599

glycosylation at N158 was reported to result in efficient viral replication in the upper 600

respiratory tract of ferrets and to increase serum antibody response (57). The role of 601

N158 glycosylation was further confirmed in recent reports of H5N1 HA mutants 602

resulting in airborne transmission of H5N1 virus between ferrets and significantly, in all 603

cases, mutation of the sequon for glycosylation at N158 was required for achieving a 604

receptor specificity that supported transmission (9, 26, 28). As N158 glycosylation might 605

mask antigenic epitopes, the removal of N158 glycosylation would be beneficial to 606

improve VN1203-like vaccine-induced immune responses. 607

A GD1-like vaccine has been successfully used in China for poultry since 2004, 608

although a recent study showed reduced efficiency against a clade 7.2 virus 609

A/Chicken/Shanxi/2/06 (35). Passive immunization with antibody H5M9 in a mouse 610

infection model indicated that H5M9 is able to protect mouse from H5N1 influenza virus 611

infection. Most neutralizing antibodies against the HA of H5N1 viruses recognize the 612

epitopes surrounding the receptor-binding site, while only a few are known to interfere 613

with membrane fusion. Here we have shown that a neutralizing antibody elicited from the 614

H5N1 immediate precursor GD1 HA binds to another conserved region located in the 615

vestigial esterase region at the base of the globular head in the HA1 subunit, providing 616

new information on a novel neutralizing epitope that is both highly conserved and 617

immunogenic. This study may shed light on design and development of a vaccine against 618

H5N1 viruses as well as passive antibody therapy. 619

620

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

29

FOOTNOTES 621

Abbreviations 622

HA, hemagglutinin; GD1, A/goose/Guangdong/1/96; VN1203, A/Vietnam/1203/2004; 623

VN1194, A/Vietnam/1194/2004. 624

625

ACKNOWLEDGMENTS 626

We thank Henry Tien of the Robotics Core at the Joint Center for Structural Genomics 627

for automated crystal screening and Dr. Xiang-Lei Yang for access to the Mosquito 628

crystal liquid handler (TTP LabTech). X-ray diffraction data sets were collected at the 629

Stanford Synchrotron Radiation Lightsource (SSRL) beamlines 9-2 and 11-1, and the 630

Advanced Photon Source (APS) beamline 23ID-B. The work was supported in part by 631

NIH grant AI058113 (I.A.W.), R56 AI099275 (I.A.W. and J.C.P.) and the Skaggs 632

Institute for Chemical Biology, by grant 30725031 (X.Y.C.) from the National 633

Outstanding Young Scientist Foundation of China and grants GDUPS 2009 and GDUPS 634

2010 (X.Y.C.), and in part by the National 973 Plan of China (No.2012CB518904). 635

Portions of this research were carried out at the Stanford Synchrotron Radiation 636

Lightsource, a national user facility operated by Stanford University on behalf of the U.S. 637

Department of Energy (DOE), Office of Basic Energy Sciences. The Stanford 638

Synchrotron Radiation Lightsource (SSRL) Structural Molecular Biology Program is 639

supported by the DOE Office of Biological and Environmental Research and by NIH, 640

National Center for Research Resources, Biomedical Technology Program 641

(P41RR001209), and the National Institute of General Medical Sciences. The GM/CA 642

CAT 23-ID-B beamline has been funded in whole or in part with federal funds from 643

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

30

National Cancer Institute (Y1-CO-1020) and NIGMS (Y1-GM-1104). The funders had 644

no role in study design, data collection and analysis, decision to publish, or preparation of 645

the manuscript. This is publication 24015 from The Scripps Research Institute. 646

647

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

31

RRFERENCES 648 649

1. Abdel-Ghafar AN, Chotpitayasunondh T, Gao Z, Hayden FG, Nguyen DH, 650

de Jong MD, Naghdaliyev A, Peiris JS, Shindo N, Soeroso S, and Uyeki TM. 651

2008. Update on avian influenza A (H5N1) virus infection in humans. N Engl J 652

Med 358:261-273. 653

2. Adams PD, Afonine PV, Bunkoczi G, Chen VB, Davis IW, Echols N, Headd 654

JJ, Hung LW, Kapral GJ, Grosse-Kunstleve RW, McCoy AJ, Moriarty NW, 655

Oeffner R, Read RJ, Richardson DC, Richardson JS, Terwilliger TC, and 656

Zwart PH. 2010. PHENIX: a comprehensive Python-based system for 657

macromolecular structure solution. Acta Crystallogr D Biol Crystallogr 66:213-658

221. 659

3. Baker NA, Sept D, Joseph S, Holst MJ, and McCammon JA. 2001. 660

Electrostatics of nanosystems: application to microtubules and the ribosome. Proc 661

Natl Acad Sci U. S. A. 98:10037-10041. 662

4. Barbey-Martin C, Gigant B, Bizebard T, Calder LJ, Wharton SA, Skehel JJ, 663

and Knossow M. 2002. An antibody that prevents the hemagglutinin low pH 664

fusogenic transition. Virology 294:70-74. 665

5. Blanc E, Roversi P, Vonrhein C, Flensburg C, Lea SM, and Bricogne G. 666

2004. Refinement of severely incomplete structures with maximum likelihood in 667

BUSTER-TNT. Acta Crystallogr D Biol Crystallogr 60:2210-2221. 668

6. Blixt O, Head S, Mondala T, Scanlan C, Huflejt ME, Alvarez R, Bryan MC, 669

Fazio F, Calarese D, Stevens J, Razi N, Stevens DJ, Skehel JJ, van Die I, 670

Burton DR, Wilson IA, Cummings R, Bovin N, Wong CH, and Paulson JC. 671

2004. Printed covalent glycan array for ligand profiling of diverse glycan binding 672

proteins. Proc Natl Acad Sci U. S. A. 101:17033-17038. 673

7. Cao Z, Meng J, Li X, Wu R, Huang Y, and He Y. 2012. The epitope and 674

neutralization mechanism of AVFluIgG01, a broad-reactive human monoclonal 675

antibody against H5N1 influenza virus. PLoS One 7:e38126. 676

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

32

8. Caton AJ, Brownlee GG, Yewdell JW, and Gerhard W. 1982. The antigenic 677

structure of the influenza virus A/PR/8/34 hemagglutinin (H1 subtype). Cell 678

31:417-427. 679

9. Chen LM, Blixt O, Stevens J, Lipatov AS, Davis CT, Collins BE, Cox NJ, 680

Paulson JC, and Donis RO. 2012. In vitro evolution of H5N1 avian influenza 681

virus toward human-type receptor specificity. Virology 422:105-113. 682

10. Chen VB, Arendall WB, 3rd, Headd JJ, Keedy DA, Immormino RM, Kapral 683

GJ, Murray LW, Richardson JS, and Richardson DC. 2010. MolProbity: all-684

atom structure validation for macromolecular crystallography. Acta Crystallogr D 685

Biol Crystallogr 66:12-21. 686

11. Chen W, Sun S, and Li Z. 2012. Two glycosylation sites in H5N1 influenza 687

virus hemagglutinin that affect binding preference by computer-based analysis. 688

PLoS One 7:e38794. 689

12. Chen Y, Qin K, Wu WL, Li G, Zhang J, Du H, Ng MH, Shih JW, Peiris JS, 690

Guan Y, Chen H, and Xia N. 2009. Broad cross-protection against H5N1 avian 691

influenza virus infection by means of monoclonal antibodies that map to 692

conserved viral epitopes. J Infect Dis 199:49-58. 693

13. Corti D, Voss J, Gamblin SJ, Codoni G, Macagno A, Jarrossay D, Vachieri 694

SG, Pinna D, Minola A, Vanzetta F, Silacci C, Fernandez-Rodriguez BM, 695

Agatic G, Bianchi S, Giacchetto-Sasselli I, Calder L, Sallusto F, Collins P, 696

Haire LF, Temperton N, Langedijk JP, Skehel JJ, and Lanzavecchia A. 2011. 697

A neutralizing antibody selected from plasma cells that binds to group 1 and 698

group 2 influenza A hemagglutinins. Science 333:850-856. 699

14. Dreyfus C, Laursen NS, Kwaks T, Zuijdgeest D, Khayat R, Ekiert DC, Lee 700

JH, Metlagel Z, Bujny MV, Jongeneelen M, van der Vlugt R, Lamrani M, 701

Korse HJ, Geelen E, Sahin O, Sieuwerts M, Brakenhoff JP, Vogels R, Li OT, 702

Poon LL, Peiris M, Koudstaal W, Ward AB, Wilson IA, Goudsmit J, and 703

Friesen RH. 2012. Highly conserved protective epitopes on influenza B viruses. 704

Science 337:1343-1348. 705

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

33

15. Ekiert DC, Bhabha G, Elsliger MA, Friesen RH, Jongeneelen M, Throsby M, 706

Goudsmit J, and Wilson IA. 2009. Antibody recognition of a highly conserved 707

influenza virus epitope. Science 324:246-251. 708

16. Ekiert DC, Friesen RH, Bhabha G, Kwaks T, Jongeneelen M, Yu W, 709

Ophorst C, Cox F, Korse HJ, Brandenburg B, Vogels R, Brakenhoff JP, 710

Kompier R, Koldijk MH, Cornelissen LA, Poon LL, Peiris M, Koudstaal W, 711

Wilson IA, and Goudsmit J. 2011. A highly conserved neutralizing epitope on 712

group 2 influenza A viruses. Science 333:843-850. 713

17. Feng E, Ye D, Li J, Zhang D, Wang J, Zhao F, Hilgenfeld R, Zheng M, Jiang 714

H, and Liu H. 2012. Recent advances in neuraminidase inhibitor development as 715

anti-influenza drugs. ChemMedChem 7:1527-1536. 716

18. Ferreira HL, Lambrecht B, van Borm S, Torrieri-Dramard L, Klatzmann D, 717

Bellier B, and van den Berg T. 2010. Identification of a dominant epitope in the 718

hemagglutinin of an Asian highly pathogenic avian influenza H5N1 clade 1 virus 719

by selection of escape mutants. Avian Dis 54:565-571. 720

19. Fleury D, Barrere B, Bizebard T, Daniels RS, Skehel JJ, and Knossow M. 721

1999. A complex of influenza hemagglutinin with a neutralizing antibody that 722

binds outside the virus receptor binding site. Nat Struct Biol 6:530-534. 723

20. Fleury D, Daniels RS, Skehel JJ, Knossow M, and Bizebard T. 2000. 724

Structural evidence for recognition of a single epitope by two distinct antibodies. 725

Proteins 40:572-578. 726

21. Fleury D, Wharton SA, Skehel JJ, Knossow M, and Bizebard T. 1998. 727

Antigen distortion allows influenza virus to escape neutralization. Nat Struct Biol 728

5:119-123. 729

22. Ha Y, Stevens DJ, Skehel JJ, and Wiley DC. 2002. H5 avian and H9 swine 730

influenza virus haemagglutinin structures: possible origin of influenza subtypes. 731

EMBO J 21:865-875. 732

23. Ha Y, Stevens DJ, Skehel JJ, and Wiley DC. 2001. X-ray structures of H5 733

avian and H9 swine influenza virus hemagglutinins bound to avian and human 734

receptor analogs. Proc Natl Acad Sci U. S. A. 98:11181-11186. 735

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

34

24. Han T, Sui J, Bennett AS, Liddington RC, Donis RO, Zhu Q, and Marasco 736

WA. 2011. Fine epitope mapping of monoclonal antibodies against hemagglutinin 737

of a highly pathogenic H5N1 influenza virus using yeast surface display. Biochem 738

Biophys Res Commun 409:253-259. 739

25. Harlow KU, and Lane D. 1998. Antibodies: A Laboratory Manual. Cold Spring 740

Harbor Press, Cold Spring Harbor, NY. 741

26. Herfst S, Schrauwen EJ, Linster M, Chutinimitkul S, de Wit E, Munster VJ, 742

Sorrell EM, Bestebroer TM, Burke DF, Smith DJ, Rimmelzwaan GF, 743

Osterhaus AD, and Fouchier RA. 2012. Airborne transmission of influenza 744

A/H5N1 virus between ferrets. Science 336:1534-1541. 745

27. Hu H, Voss J, Zhang G, Buchy P, Zuo T, Wang L, Wang F, Zhou F, Wang 746

G, Tsai C, Calder L, Gamblin SJ, Zhang L, Deubel V, Zhou B, Skehel JJ, 747

and Zhou P. 2012. A human antibody recognizing a conserved epitope of H5 748

hemagglutinin broadly neutralizes highly pathogenic avian influenza H5N1 749

viruses. J Virol 86:2978-2989. 750

28. Imai M, Watanabe T, Hatta M, Das SC, Ozawa M, Shinya K, Zhong G, 751

Hanson A, Katsura H, Watanabe S, Li C, Kawakami E, Yamada S, Kiso M, 752

Suzuki Y, Maher EA, Neumann G, and Kawaoka Y. 2012. Experimental 753

adaptation of an influenza H5 HA confers respiratory droplet transmission to a 754

reassortant H5 HA/H1N1 virus in ferrets. Nature 486:420-428. 755

29. Jackson S, Van Hoeven N, Chen LM, Maines TR, Cox NJ, Katz JM, and 756

Donis RO. 2009. Reassortment between avian H5N1 and human H3N2 influenza 757

viruses in ferrets: a public health risk assessment. J Virol 83:8131-8140. 758

30. Kaverin NV, Rudneva IA, Govorkova EA, Timofeeva TA, Shilov AA, 759

Kochergin-Nikitsky KS, Krylov PS, and Webster RG. 2007. Epitope mapping 760

of the hemagglutinin molecule of a highly pathogenic H5N1 influenza virus by 761

using monoclonal antibodies. J Virol 81:12911-12917. 762

31. Kaverin NV, Rudneva IA, Ilyushina NA, Varich NL, Lipatov AS, Smirnov 763

YA, Govorkova EA, Gitelman AK, Lvov DK, and Webster RG. 2002. 764

Structure of antigenic sites on the haemagglutinin molecule of H5 avian influenza 765

virus and phenotypic variation of escape mutants. J Gen Virol 83:2497-2505. 766

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

35

32. Khurana S, Suguitan AL, Jr., Rivera Y, Simmons CP, Lanzavecchia A, 767

Sallusto F, Manischewitz J, King LR, Subbarao K, and Golding H. 2009. 768

Antigenic fingerprinting of H5N1 avian influenza using convalescent sera and 769

monoclonal antibodies reveals potential vaccine and diagnostic targets. PLoS Med 770

6:e1000049. 771

33. Le QM, Kiso M, Someya K, Sakai YT, Nguyen TH, Nguyen KH, Pham ND, 772

Ngyen HH, Yamada S, Muramoto Y, Horimoto T, Takada A, Goto H, Suzuki 773

T, Suzuki Y, and Kawaoka Y. 2005. Avian flu: isolation of drug-resistant H5N1 774

virus. Nature 437:1108. 775

34. Li J, Wang Y, Liang Y, Ni B, Wan Y, Liao Z, Chan KH, Yuen KY, Fu X, 776

Shang X, Wang S, Yi D, Guo B, Di B, Wang M, Che X, and Wu Y. 2009. Fine 777

antigenic variation within H5N1 influenza virus hemagglutinin's antigenic sites 778

defined by yeast cell surface display. Eur J Immunol 39:3498-3510. 779

35. Li Y, Shi J, Zhong G, Deng G, Tian G, Ge J, Zeng X, Song J, Zhao D, Liu L, 780

Jiang Y, Guan Y, Bu Z, and Chen H. 2010. Continued evolution of H5N1 781

influenza viruses in wild birds, domestic poultry, and humans in China from 2004 782

to 2009. J Virol 84:8389-8397. 783

36. Lim AP, Wong SK, Chan AH, Chan CE, Ooi EE, and Hanson BJ. 2008. 784

Epitope characterization of the protective monoclonal antibody VN04-2 shows 785

broadly neutralizing activity against highly pathogenic H5N1. Virol J 5:80. 786

37. Maines TR, Lu XH, Erb SM, Edwards L, Guarner J, Greer PW, Nguyen DC, 787

Szretter KJ, Chen LM, Thawatsupha P, Chittaganpitch M, Waicharoen S, 788

Nguyen DT, Nguyen T, Nguyen HH, Kim JH, Hoang LT, Kang C, Phuong 789

LS, Lim W, Zaki S, Donis RO, Cox NJ, Katz JM, and Tumpey TM. 2005. 790

Avian influenza (H5N1) viruses isolated from humans in Asia in 2004 exhibit 791

increased virulence in mammals. J Virol 79:11788-11800. 792

38. Martin V, Sims L, Lubroth J, Kahn S, Domenech J, and Begnino C. 2006. 793

History and evolution of HPAI viruses in southeast Asia. Ann N Y Acad Sci 794

1081:153-162. 795

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

36

39. McCoy AJ, Grosse-Kunstleve RW, Storoni LC, and Read RJ. 2005. 796

Likelihood-enhanced fast translation functions. Acta Crystallogr D Biol 797

Crystallogr 61:458-464. 798

40. McDonald IK, and Thornton JM. 1994. Satisfying hydrogen bonding potential 799

in proteins. J Mol Biol 238:777-793. 800

41. Murshudov GN, Vagin AA, and Dodson EJ. 1997. Refinement of 801

macromolecular structures by the maximum-likelihood method. Acta Crystallogr 802

D Biol Crystallogr 53:240-255. 803

42. Octaviani CP, Ozawa M, Yamada S, Goto H, and Kawaoka Y. 2010. High 804

genetic compatibility between swine-origin H1N1 and highly pathogenic avian 805

H5N1 influenza viruses. J Virol:10918-10922. 806

43. Oh HL, Akerstrom S, Shen S, Bereczky S, Karlberg H, Klingstrom J, Lal 807

SK, Mirazimi A, and Tan YJ. 2010. An antibody against a novel and conserved 808

epitope in the hemagglutinin 1 subunit neutralizes numerous H5N1 influenza 809

viruses. J Virol 84:8275-8286. 810

44. Ohkura T, Kikuchi Y, Kono N, Itamura S, Komase K, Momose F, and 811

Morikawa Y. 2012. Epitope mapping of neutralizing monoclonal antibody in 812

avian influenza A H5N1 virus hemagglutinin. Biochem Biophys Res Commun 813

418:38-43. 814

45. Paulson JC, and de Vries RP. 2013. H5N1 receptor specificity as a factor in 815

pandemic risk. Virus Res:Epub ahead of print, doi: 10.1016/j.virusres. 816

46. Philpott M, Easterday BC, and Hinshaw VS. 1989. Neutralizing epitopes of the 817

H5 hemagglutinin from a virulent avian influenza virus and their relationship to 818

pathogenicity. J Virol 63:3453-3458. 819

47. Prabakaran M, Ho HT, Prabhu N, Velumani S, Szyporta M, He F, Chan KP, 820

Chen LM, Matsuoka Y, Donis RO, and Kwang J. 2009. Development of 821

epitope-blocking ELISA for universal detection of antibodies to human H5N1 822

influenza viruses. PLoS One 4:e4566. 823

48. Qian M, Hu H, Zuo T, Wang G, Zhang L, and Zhou P. 2012. Unravel a 824

neutralization mechanism by two human antibodies against conserved epitopes in 825

the globular head of H5 hemagglutinin. J Virol 87:3571-3577. 826

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

37

49. Rudneva IA, Kushch AA, Masalova OV, Timofeeva TA, Klimova RR, Shilov 827

AA, Ignatieva AV, Krylov PS, and Kaverin NV. 2010. Antigenic epitopes in 828

the hemagglutinin of Qinghai-type influenza H5N1 virus. Viral Immunol 23:181-829

187. 830

50. Sambhara S, and Poland GA. 2010. H5N1 Avian influenza: preventive and 831

therapeutic strategies against a pandemic. Annu Rev Med 61:187-198. 832

51. Stevens J, Blixt O, Chen LM, Donis RO, Paulson JC, and Wilson IA. 2008. 833

Recent avian H5N1 viruses exhibit increased propensity for acquiring human 834

receptor specificity. J Mol Biol 381:1382-1394. 835

52. Stevens J, Blixt O, Tumpey TM, Taubenberger JK, Paulson JC, and Wilson 836

IA. 2006. Structure and receptor specificity of the hemagglutinin from an H5N1 837

influenza virus. Science 312:404-410. 838

53. Sui J, Hwang WC, Perez S, Wei G, Aird D, Chen LM, Santelli E, Stec B, 839

Cadwell G, Ali M, Wan H, Murakami A, Yammanuru A, Han T, Cox NJ, 840

Bankston LA, Donis RO, Liddington RC, and Marasco WA. 2009. Structural 841

and functional bases for broad-spectrum neutralization of avian and human 842

influenza A viruses. Nat Struct Mol Biol 16:265-273. 843

54. Sun L, Lu X, Li C, Wang M, Liu Q, Li Z, Hu X, Li J, Liu F, Li Q, Belser JA, 844

Hancock K, Shu Y, Katz JM, Liang M, and Li D. 2009. Generation, 845

characterization and epitope mapping of two neutralizing and protective human 846

recombinant antibodies against influenza A H5N1 viruses. PLoS One 4:e5476. 847

55. Ungchusak K, Auewarakul P, Dowell SF, Kitphati R, Auwanit W, 848

Puthavathana P, Uiprasertkul M, Boonnak K, Pittayawonganon C, Cox NJ, 849

Zaki SR, Thawatsupha P, Chittaganpitch M, Khontong R, Simmerman JM, 850

and Chunsutthiwat S. 2005. Probable person-to-person transmission of avian 851

influenza A (H5N1). N Engl J Med 352:333-340. 852

56. Wang SF, Chen KH, Thitithanyanont A, Yao L, Lee YM, Chan YJ, Liu SJ, 853

Chong P, Liu WT, Huang JC, and Chen YM. 2009. Generating and 854

characterizing monoclonal and polyclonal antibodies against avian H5N1 855

hemagglutinin protein. Biochem Biophys Res Commun 382:691-696. 856

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

38

57. Wang W, Lu B, Zhou H, Suguitan AL, Jr., Cheng X, Subbarao K, Kemble G, 857

and Jin H. 2010. Glycosylation at 158N of the hemagglutinin protein and 858

receptor binding specificity synergistically affect the antigenicity and 859

immunogenicity of a live attenuated H5N1 A/Vietnam/1203/2004 vaccine virus in 860

ferrets. J Virol 84:6570-6577. 861

58. Whittle JR, Zhang R, Khurana S, King LR, Manischewitz J, Golding H, 862

Dormitzer PR, Haynes BF, Walter EB, Moody MA, Kepler TB, Liao HX, 863

and Harrison SC. 2011. Broadly neutralizing human antibody that recognizes the 864

receptor-binding pocket of influenza virus hemagglutinin. Proc Natl Acad Sci U. 865

S. A. 108:14216-14221. 866

59. Wiley DC, Wilson IA, and Skehel JJ. 1981. Structural identification of the 867

antibody-binding sites of Hong Kong influenza haemagglutinin and their 868

involvement in antigenic variation. Nature 289:373-378. 869

60. Wu WL, Chen Y, Wang P, Song W, Lau SY, Rayner JM, Smith GJ, Webster 870

RG, Peiris JS, Lin T, Xia N, Guan Y, and Chen H. 2008. Antigenic profile of 871

avian H5N1 viruses in Asia from 2002 to 2007. J Virol 82:1798-1807. 872

61. Xu R, Ekiert DC, Krause JC, Hai R, Crowe JE, Jr., and Wilson IA. 2010. 873

Structural basis of preexisting immunity to the 2009 H1N1 pandemic influenza 874

virus. Science 328:357-360. 875

62. Xu X, Subbarao K, Cox NJ, and Guo Y. 1999. Genetic characterization of the 876

pathogenic influenza A/Goose/Guangdong/1/96 (H5N1) virus: similarity of its 877

hemagglutinin gene to those of H5N1 viruses from the 1997 outbreaks in Hong 878

Kong. Virology 261:15-19. 879

63. Zhu X, Yu W, McBride R, Li Y, Chen LM, Donis RO, Tong S, Paulson JC, 880

and Wilson IA. 2013. Hemagglutinin homologue from H17N10 bat influenza 881

virus exhibits divergent receptor-binding and pH-dependent fusion activities. Proc 882

Natl Acad Sci U. S. A. 110:1458-1463. 883

884 885

886

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

39

Table 1. Neutralizing activities of antibody H5M9 against H5N1 viruses* 887

H5N1 strain Clade H5M9 NT titer A/HongKong/156/97 0 >1280 A/Beijing/01/2003 7 640 A/Chicken/HK/8005.2/08 2.3.4 320

888

*We reported previously (34) that H5M9 (IgG1) showed strong neutralizing activity (neutralization titer 889

(NT) >1280) against H5N1 viruses isolated from 1997 to 2006, A/HongKong/482/97 (clade 0) and 890

A/HongKong/483/97 (clade 0), A/HK/213/03 (clade 1) and A/Vietnam/1194/2004 (clade 1), as well as 891

A/Shengzhen/406H/2006 (clade 2.3.4). 892

893

894

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

40

Table 2. Data collection and refinement statistics of VN1203 HA and GD1 HA crystals. 895

Data set VN1203 HA –

H5M9 Fab´

GD1 HA GD1 HA –

H5M9 Fab´

Space group C2 P21 P3121

Unit cell (Å)

Unit cell (deg.)

a = 142.6,

b = 251.3,

c = 230.5

β = 107.1

a = 72.5,

b = 225.7,

c = 211.6

β = 99.0

a = b = 199.6,

c = 466.9

Resolution (Å) a 50.0-3.60

(3.83-3.60)

50.0-2.60

(2.69-2.60)

50.0-7.0

(7.44-7.0)

X-ray source APS 23ID-B SSRL 9-2 SSRL 11-1

Unique refs 85,101 196,526 16,538

Redundancy a 2.8 (2.2) 3.4 (2.2) 5.8 (6.2)

Average I/σ(I) a 11.5 (1.2) 22.8 (1.9) 12.9 (1.2)

Completeness a 94.9 (84.3) 95.3 (75.6) 93.3 (64.0)

Rsyma,b 0.10 (0.94) 0.10 (0.70) 0.14 (0.92)

Rpima,b 0.08 (0.72) 0.06 (0.49) 0.06 (0.39)

HA monomers in a.u. 3 9 6

Vm (Å3/Da) 6.1 3.4 4.3

Refs used in refinement

Refined residues

84,244

2,820

195,939

4,464

15,673

5,569

Refined waters 0 117 0

Rcrystc 0.201 0.189 0.377

Rfreed 0.238 0.243 0.387

B-values (Å2)

Protein

Waters

142

-

95

75

-

-

Wilson B-values (Å2) 100 79 -

Ramachandran plot (%)e 92.4, 0.7 97.1, 0.1 -

rmsd bond (Å) 0.012 0.009 -

rmsd angle (deg.) 1.6 1.3 -

896 a Parenthesis denote outer-shell statistics. 897 b Rsym = hkli |Ihkl,i - <Ihkl>| /hkli Ihkl,i and Rpim = hkl[1/(N-1)]1/2i |Ihkl,i - <Ihkl>| /hkli Ihkl,i, where Ihkl,i is 898 the scaled intensity of the ith measurement of reflection h, k, l, < Ihkl> is the average intensity for that 899 reflection, and N is the redundancy.. 900 c Rcryst = hkl |Fo - Fc| / hkl |Fo|, where Fo and Fc are the observed and calculated structure factors. 901 d Rfree was calculated as for Rcryst, but on 5% of data excluded before refinement. 902 e The values are percentage of residues in the favored and outliers regions analyzed by MolProbity (10). 903

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

41

Table 3. Binding of H5 HAs by antibody H5M9 904

H5N1 strain Fab´ Kd (nM) IgG Kd (nM) A/Goose/Guangdong/1/96 1.0 0.008 A/Vietnam/1203/2004 0.9 0.04

905 906

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

42

907 Table 4. Sequence comparison of HA1 domain of H5N1 HAs and H1, H3 HAsa. 908 _________________________________________________________ 909 H5 numbering: 1 10 20 30 40 910 H3 numbering: 10 20 30 34 911 A/Goose/Guangdong/1/96(H5N1) MEKIVLLLAIVSLVK----------SDQICIGYHANNSTEQVDTIMEKNV 912 A/Vietnam/1203/2004(H5N1) MEKIVLLFAIVSLVK----------SDQICIGYHANNSTEQVDTIMEKNV 913 A/Vietnam/1194/2004(H5N1) MEKIVLLFAIVSLVK----------SDQICIGYHANNSTEQVDTIMEKNV 914 A/New Caledonia/20/1999(H1N1) MKAKLLVLLCTFTATY---------ADTICIGYHANNSTDTVDTVLEKNV 915 A/Panama/2007/1999(H3N2) MKTIIALSYILCLVFAQKLPGNDNSTATLCLGHHAVSNGTLVKTITNDQI 916 917 H5 numbering: 41 50 60 70 80 90 918 H3 numbering: 35 40 50 60 70 80 83 919 A/Goose/Guangdong/1/96(H5N1) TVTHAQDILEKTHNGKLCDLNGVKPLILRDCSVAGWLLGNPMCDEFINVP 920 A/Vietnam/1203/2004(H5N1) TVTHAQDILEKKHNGKLCDLDGVKPLILRDCSVAGWLLGNPMCDEFINVP 921 A/Vietnam/1194/2004(H5N1) TVTHAQDILEKTHNGKLCDLDGVKPLILRDCSVAGWLLGNPMCDEFINVP 922 A/New Caledonia/20/1999(H1N1) TVTHSVNLLEDSHNGKLCLLKGIAPLQLGNCSVAGWILGNPECELLISKE 923 A/Panama/2007/1999(H3N2) EVTNATELVQSSSTGRICDSP-HQILDGENCTLIDALLGDPHCDGFQNKE 924 925 H5 numbering: 91 100 110 120 130 140 926 H3 numbering: 83A 90 100 110 120 129 927 A/Goose/Guangdong/1/96(H5N1) EWSYIVEKASPANDLCYPGDFNDYEELKHLLSRTNHFEKIQIIPKS-SWSN 928 A/Vietnam/1203/2004(H5N1) EWSYIVEKANPVNDLCYPGDFNDYEELKHLLSRINHFEKIQIIPKS-SWSS 929 A/Vietnam/1194/2004(H5N1) EWSYIVEKANPVNDLCYPGDFNDYEELKHLLSRINHFEKIQIIPKS-SWSS 930 A/New Caledonia/20/1999(H1N1) SWSYIVETPNPENGTCYPGYFADYEELREQLSSVSSFERFEIFPKESSWPN 931 A/Panama/2007/1999(H3N2) -WDLFVERSKAYSN-CYPYDVPDYASLRSLVASSGTLEFNNESF---NWTG 932 933 H5 numbering: 141 150 160 170 180 190 934 H3 numbering: 130 140 150 160 170 178 935 A/Goose/Guangdong/1/96(H5N1) HDASSGVSSACPYHGRSSFFRNVVWLIKKNSAYPTIKRSYNNTNQEDLLV 936 A/Vietnam/1203/2004(H5N1) HEASLGVSSACPYQGKSSFFRNVVWLIKKNSTYPTIKRSYNNTNQEDLLV 937 A/Vietnam/1194/2004(H5N1) HEASLGVSSACPYQGKSSFFRNVVWLIKKNSTYPTIKRSYNNTNQEDLLV 938 A/New Caledonia/20/1999(H1N1) HTVT-GVSASCSHNGKSSFYRNLLWLTGKNGLYPNLSKSYVNNKEKEVLV 939 A/Panama/2007/1999(H3N2) VAQN-GTSSACKRRSNKSFFSRLNWLHQLKYKYPALNVTMPNNEKFDKLY 940 941 H5 numbering: 191 200 210 220 230 240 942 H3 numbering: 180 190 200 210 220 228 943 A/Goose/Guangdong/1/96(H5N1) LWGIHHPNDAAEQTKLYQNPTTYISVGTSTLNQRLVPEIATRPKVNGQSG 944 A/Vietnam/1203/2004(H5N1) LWGIHHPNDAAEQTKLYQNPTTYISVGTSTLNQRLVPRIATRSKVNGQSG 945 A/Vietnam/1194/2004(H5N1) LWGIHHPNDAAEQTKLYQNPTTYISVGTSTLNQRLVPRIATRSKVNGQSG 946 A/New Caledonia/20/1999(H1N1) LWGVHHPPNIGDQRALYHTENAYVSVVSSHYSRRFTPEIAKRPKVRDQEG 947 A/Panama/2007/1999(H3N2) IWGVHHPSTDSDQISIYAQASGRVTVSTKRSQQTVIPNIGSSPWVRGVSS 948 949 H5 numbering: 241 250 260 270 280 290 950 H3 numbering: 230 240 250 260 270 277 951 A/Goose/Guangdong/1/96(H5N1) RMEFFWTILKPNDAINFESNGNFIAPEYAYKIVKKGDSAIMKSELEYGNC 952 A/Vietnam/1203/2004(H5N1) RMEFFWTILKPNDAINFESNGNFIAPEYAYKIVKKGDSTIMKSELEYGNC 953 A/Vietnam/1194/2004(H5N1) RMEFFWTILKPNDAINFESNGNFIAPEYAYKIVKKGDSTIMKSELEYGNC 954 A/New Caledonia/20/1999(H1N1) RINYYWTLLEPGDTIIFEANGNLIAPWYAFALSRGFGSGIITSNAPMDEC 955 A/Panama/2007/1999(H3N2) RISIYWTIVKPGDILLINSTGNLIAPRGYFKI-RSGKSSIMRSDAPIGKC 956 957 H5 numbering: 291 300 310 320 330 340 958 H3 numbering: 280 290 300 310 320 959 A/Goose/Guangdong/1/96(H5N1) NTKCQTPMGAINSSMPFHNIHPLTIGECPKYVKSNRLVLATGLRNTPQRE 960 A/Vietnam/1203/2004(H5N1) NTKCQTPMGAINSSMPFHNIHPLTIGECPKYVKSNRLVLATGLRNSPQRE 961 A/Vietnam/1194/2004(H5N1) NTKCQTPMGAINSSMPFHNIHPLTIGECPKYVKSNRLVLATGLRNSPQRE 962 A/New Caledonia/20/1999(H1N1) DAKCQTPQGAINSSLPFQNVHPVTIGECPKYVRSAKLRMVTGLRNIPS-- 963 A/Panama/2007/1999(H3N2) NSECITPNGSIPNDKPFQNVNRITYGACPRYVKQNTLKLATGMRNVPE-- 964 _________________________________________________________ 965 a Red indicates H5M9 epitope residues on A/Vietnam/1203/2004 (VN1203) HA. The only amino 966 acid difference between VN1203 HA and A/Vietnam/1194/2004 (VN1194) HAs is highlighted in 967 green. The amino acid difference between VN1203 and GD1 HAs are highlighted in cyan. The 968 H1 and H3 residues not conserved in H5M9 epitope are highlighted in yellow. H3 numbering 969 system is used throughout in this paper and H5 numbering is shown here for comparison. 970 971

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

43

972 Table 5. Antibody H5M9 epitope residues in VN1203 HA and comparison with other 973

HAs from H5N1 viruses neutralized by H5M9 in this studya. 974

Amino acid position

Viral strain Clade 53 55 56 57 62 78 81 83 83a 117 119 273 274 276 278

A/Vietnam/1203/2004 1 D D V K R E N P E H E E Y N N

A/goose/Guangdong/1/96 0 · N · · · · · · · · · · · · ·

A/HongKong/156/97 0 · N · · · · · · · · · · · · ·

A/HongKong/483/97 0 · N · · · · · · · · · · · · ·

A/HongKong/213/03 1 · · · · · · · · · · · · · · ·

A/Vietnam/1194/2004 1 · · · · · · · · · · · · · · ·

A/Shenzhen/406H/2006 2.3.4 · · · · · · · · · · · · · · ·

A/Beijing/01/2003 7 · · · · · · · · · · · · · · ·

975 a The identified key epitope residues are shown in boldface type. 976 977

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

44

Table 6. Conservation of H5M9 epitope residues in influenza H5N1 HAsa. 978 979

Amino Residue number

acid 53 55 56 57 62 78 81 83 83a 117 119 273 274 276 278

Ala 3 0 0 0 0 6 0 0 0 0 0 2 0 0 0

Cys 0 0 0 0 0 0 0 0 0 0 0 0 1 0 0

Asp 1,326 1,314 0 0 0 0 26 0 0 0 1 1 0 25 35

Glu 1 3 0 1 0 1,565 0 0 1,572 0 1,560 1,557 0 0 0

Phe 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0

Gly 0 1 1 0 4 0 0 0 1 0 0 11 0 0 0

His 0 0 0 0 0 0 2 0 0 1,484 0 0 1 0 0

Ile 0 0 7 0 0 0 0 0 0 0 0 0 1 0 1

Lys 0 37 0 1,503 260 7 12 0 3 0 18 6 0 0 1

Leu 0 0 0 0 0 0 0 1 0 0 0 0 0 0 0

Met 0 0 2 0 0 0 0 0 0 0 0 0 0 0 0

Asn 197 221 0 0 0 0 1,537 0 0 0 0 0 0 1,545 1,480

Pro 0 0 0 0 0 0 0 1,443 0 0 0 0 0 0 0

Gln 0 0 0 0 0 0 0 7 0 0 0 0 0 0 1

Arg 0 0 0 75 1,315 0 0 0 0 95 0 0 0 0 0

Ser 52 0 0 0 0 0 1 127 0 0 0 0 0 5 56

Thr 0 0 0 0 0 0 0 0 0 0 0 0 0 0 2

Val 0 0 1,569 0 0 0 0 0 1 0 0 0 0 0 0

Trp 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0

Tyr 0 3 0 0 0 0 0 0 0 0 0 0 1,574 2 1

980 a The incidence of an amino acid occurring at certain position is shown. A total of 1,579 full-length, non-981

redundant HA sequences from all influenza H5N1 viruses were available in the Influenza A Virus Resource 982

at the NCBI in January 22, 2013. 983

984

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

45

Table 7. Conservation of H5M9 epitope residues of VN1203 H5 HA in different clades 985

and subclades of H5N1 virusesa. 986

Amino acid position

Viral strain Clade 53 55 56 57 62 78 81 83 83a 117 119 273 274 276 278

A/Vietnam/1203/2004 1 D D V K R E N P E H E E Y N N

A/Goose/Huadong/1/2000 0 · · · · · · · · · · · · · · ·

A/Cambodia/S1211394/2008 1.1 · · · R · · · · · · · · · · ·

A/Chicken/Indonesia/11/2003 2.1.1 · · · · · · · · · · · · · · ·

A/Indonesia/542H/2006 2.1.2 · · · · · · · · · · · · · · ·

A/Indonesia/5/2005 2.1.3.2 · · · · · · · · · · · · · · ·

A/Chicken/Medan/BPPVRI_15/2008 2.1.3.3 · · · · · · · · · · · · · · ·

A/bar-headed goose/Qinghai/12/05 2.2 · · · · · · · · · · · · · · ·

A/turkey/Turkey/1/2005 2.2.1 · · · · · · · · · · · · · · ·

A/Bangladesh/207095/2008 2.2.2 · · · · · · · · · · · · · · ·

A/Duck/Hunan/127/2005 2.3.1 · · · · · · · · · · · · · · ·

A/chicken/Guangdong/1/2005 2.3.2 · · · · · · · · · · · · · · ·

A/Hubei/1/2010 2.3.2.1 · N · · K · · · · · · · · · ·

A/chicken/Guiyang/3055/2005 2.3.3 · · · · · · · · · · · · · · ·

A/Anhui/1/2005 2.3.4 · · · · · · · · · · · · · · ·

A/chicken/Sichuan/81/2005 2.3.4.1 · · · · · · · · · · · · · · ·

A/Vietnam/HN36250/2010 2.3.4.2 · · · · · · · · · · · · · · · A/chicken/Vietnam/NCVD-188/2008 2.3.4.3 · · · · · · · · · · · · · · ·

A/chicken/jiyuan/1/03 2.4 · · · · · · · · · · · · · · ·

A/chicken/Korea/ES/03 2.5 · · · · · · · · · · · · · · ·

A/chicken/HongKong/YU562/01 3 · · · · · · · · · · · · · · ·

A/goose/Fujian/bb/2003 4 · · · · · · · · · · · · · · ·

A/goose/Guangxi/1097/2004 5 · · · · · · · · · · · · · · ·

A/swine/Anhui/ca/2004 6 · · · · · · · · · · · · · · ·

A/duck/Yunnan/5133/2005 7 · · · · · · · · · · · · · · ·

A/Ck/HK/YU777/02 8 · · · · · · · · · · · · · · ·

A/chicken/Fujian/1042/2005 9 · · · · · · · · · · · · · · ·

A/chicken/Egypt/1029/2010 2.2.1 N · · · · · · · · · · · · · ·

A/chicken/Shanxi/10/2006 7.1 N · · · K · · S · · K · · · ·

A/chicken/Henan/A-7/2006 7.2 N · · · K · · S · · K · · · ·

987 a The identified key epitope residues are shown in boldface type. 988 989

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

46

Table 8. Conservation of H5M9 HA epitope in HAs of influenza virus subtypes H5N1 to 990 H5N9. 991 992

Residue

Percent Conservation from H5N1 to H5N9 Consensus a

H5N1

(1679)b H5N2 (379)

H5N3 (68)

H5N4 (4)

H5N5 (4)

H5N6 (2)

H5N7 (7)

H5N8 (5)

H5N9 (21)

53 D 85.9 c 1.8 (S)d 0.0 (S) 0.0 (S) 75.0 0.0 (S) 0.0 (S) 0.0 (S) 0.0 (S)

55 D 86.1 0.8 (K) 0.0 (N) 0.0 (K) 50.0 0.0 (N) 0.0 (K) 0.0 (N) 0.0 (N)

56 V 99.4 99.7 100.0 100.0 100.0 100.0 100.0 100.0 100.0

57 K 95.9 46.7 (R) 69.1 0.0 (R) 75.0 100.0 28.6 (R) 80.0 47.6 (R)

62 R 86.1 32.2 (K) 64.7 0.0 (K) 75.0 100.0 28.6 (K) 80.0 42.9 (K)

78 E 99.4 97.1 98.5 100.0 100.0 100.0 100.0 100.0 95.2

81 N 98.0 92.3 94.1 100.0 50.0 100.0 100.0 100.0 100.0

83 P 93.6 99.2 100.0 100.0 100.0 100.0 100.0 100.0 90.5

83a E 99.6 99.7 100.0 100.0 100.0 100.0 100.0 100.0 100.0

117 H 95.4 95.0 100.0 100.0 100.0 100.0 100.0 100.0 100.0

119 E 99.0 97.9 98.5 100.0 100.0 100.0 100.0 100.0 100.0

273 E 98.9 87.6 91.2 100.0 100.0 100.0 71.4 100.0 95.2

274 Y 99.7 100.0 100.0 100.0 100.0 100.0 100.0 100.0 100.0

276 N 98.4 98.1 100.0 100.0 100.0 100.0 100.0 100.0 100.0

278 N 95.2 40.6 (D) 72.1 0.0 (D) 75.0 100.0 28.6 (D) 80.0 47.6 (D)

993 a Most common residue at position by simple majority in H5N1 virus HA sequences (full-length, non-994

redundant) that were available in the Influenza A Virus Resource at the NCBI in January 22, 2013. 995 b Number of sequences available for this subtype in the Influenza A Virus Resource at the NCBI in January 996

22, 2013. 997 c Percent of all H5 sequences that are identical to the consensus. 998 d Most common residue in this subtype. 999

1000

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

47

Figure Legends 1001 1002 FIG 1 Crystal structure of antibody H5M9 Fab´ in complex with VN1203 HA. (A) 1003

Overall structure of the H5M9 Fab´ - VN1203 HA complex. The H5M9 Fab´ binds in the 1004

vestigial esterase subdomain of the HA. One HA/ Fab´ protomer of the trimeric complex 1005

is colored with HA1 in pink and HA2 in cyan, with the corresponding bound Fab´ heavy 1006

chain in orange and Fab´ light chain in yellow. N-linked glycans are depicted in colored 1007

balls representing their atom types, and asparagines that code for potential N-1008

glycosylation sites are also shown in sticks. The other two protomers are in grey, but the 1009

third Fab molecule is hidden behind the HA trimer. (B) Surface presentation illustrating 1010

the H5M9 binding site (in green) on one VN1203 HA protomer, which is colored with 1011

HA1 in pink and HA2 in cyan. The H5M9 footprint on VN1203 HA is far from the 1012

receptor binding site (RBS, in red). (C) Closer view of the H5M9 epitope with selected 1013

epitope residues shown as sticks. The four identified key residues are labeled in red. (D) 1014

Footprint of HA on H5M9 combining site (i.e. paratope), highlighting contacting 1015

antibody residues from the heavy chain (colored with orange carbon atom) and light 1016

chain (with yellow carbon atoms). 1017

1018

FIG 2 Overall structures of GD1 H5 HA and its complex with antibody H5M9. (A) GD1 1019

H5 HA in apo form. One HA protomer of the trimeric complex is colored with HA1 in 1020

pink and HA2 in cyan, with the other two protomers in green. N-linked glycans are 1021

depicted in colored balls representing their atom types, and asparagines that code for 1022

potential N-glycosylation sites are also shown in sticks. (B) GD1 H5 HA in complex with 1023

H5M9 Fab´. The H5M9 Fab´ binds in the vestigial esterase subdomain of the HA, which 1024

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

48

is the same epitope as that in VN1203 H5 HA (Fig. 1). One HA/ Fab´ protomer of the 1025

trimeric complex is colored with HA1 in pink and HA2 in cyan, with the corresponding 1026

Fab´ heavy chain in orange and Fab´ light chain in yellow. The other two protomers are 1027

in green and the third Fab´ molecule is hidden behind the HA trimer. 1028

1029

FIG 3 Structural comparison of GD1 HA apo-form (in yellow side chains and pink CĮ 1030

atoms) and VN1203 HA (PDB code 3GBM, in grey side chains and CĮ atoms). (A) 1031

Comparison of the receptor binding sites. The overall structures as well as key binding 1032

residues are virtually identical for except one amino acid substitution of residue 221 from 1033

Pro (GD1 HA) to Ser (VN1203 HA) near the receptor binding site. (B) Comparison of 1034

the H5M9 epitopes in GD1 and VN1203 HAs. The H5 epitope residues are 1035

superimposable with only one amino acid substitution of Asn55 (GD1 HA) to Asp55 1036

(VN1203 HA). 1037

1038

FIG 4 Receptor binding was investigated against printed glycans on a microarray (Fig. 1039

S2) for GD1 HA (A) and control VN1203 HA (B). Different categories of glycans on the 1040

array are highlighted in colors, and the bars denote the fluorescent signal intensity, and 1041

error bars indicate the standard errors. The normal starting concentration of 15 µg/ml was 1042

applied to both HAs. Both GD1 HA and VN1203 HA showed similar binding affinities to 1043

most Į2-3 linked glycans, but no obvious binding to any Į2-6 linked glycan. 1044

Interestingly, VN1203 HA also showed strong binding to fucosylated Į2-3 glycans (in 1045

yellow box), in contrast to GD1 HA, which did not bind these glycans. 1046

1047

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

49

FIG 5 Escape mutant D53N of VN1994 HA to antibody H5M9. (A) Neutralization 1048

activity of H5M9 to H5N1 viruses with wild-type VN1194 HA and its escape mutant 1049

D53N. Neutralization activities were evaluated by plaque reduction neutralization assays 1050

using MDCK cells, which showed that D53N mutation confers resistance to H5M9. The 1051

data are representative of triple independent experiments. 1052

(B) Electrostatic potential surfaces around H5M9 paratope of antibody H5M9 1053

combining site with the interacting Asp53 from VN1203 HA labeled. Electrostatic 1054

potential surfaces around wild-type VN1203 HA with Asp53 (C) and VN1203 HA with 1055

the Asp53Asn mutant model (D). Electrostatic surface potentials were calculated using 1056

the APBS program (3). Negatively charged regions are red, positively charged regions are 1057

blue, and neutral regions are white (-10 to 10 KbT/ec potential range). The Asp53Asn 1058

mutation alters the electrostatic potential from weakly acidic (C) to near neutral (D) 1059

around the HA1 53 position, rendering it unfavorable to bind H5M9, which is weakly 1060

basic around the interaction site with HA1 Asp53 (B). 1061

1062

FIG 6 Flow cytometry histograms of antibody H5M9 binding to wild-type or mutant of 1063

VN1194 HA proteins from yeast surface display. Representative histograms are shown 1064

for anti-Xpress antibody, antibody H5M9, and antibody H5M11 with wild-type HA and 1065

each of the HA mutants. The H5 proteins expressed well on the yeast cell surface and can 1066

be detected by the reactivity of the Xpress epitope tag in the HA protein with the anti-1067

Xpress antibody, as well as the binding activity by a control antibody H5M11 (IgG2a) 1068

elicited from the same mouse immunization as H5M9 (IgG1), which appears to bind to a 1069

different HA epitope (34). Arrows indicate mutations that result in significant or almost 1070

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

50

complete loss of antibody binding. Data shown are representative of at least three 1071

independent experiments. 1072

1073

FIG 7 Reducing SDS-PAGE of trypsin digestion of VN1203 HA in the absence (lanes 1 1074

and 2) and presence (lanes 3 and 4) of H5M9 Fab´. The HA or HA- Fab´ mixture was 1075

exposed to pH 4.9 (lanes 1 and 3) or pH 8.0 (lanes 2 and 4) at 37 ºC for one hour before 1076

trypsin digestion at pH 8.4. As expected, the HA1 could be completely digested at pH 4.9 1077

without H5M9 Fab´ (lane 1), while in the control at pH 8.0, the HA1 remains contact. 1078

After precomplexing the HA with H5M9 Fab´, the HA appeared to be resistant to trypsin 1079

digestion even at pH 4.9 in addition to pH 8.0. These results suggest H5M9 may prevent 1080

low pH-induced conformational changes of the HA. 1081

1082

FIG 8 Prophylactic and therapeutic efficacy of H5M9 against lethal H5N1 infection in 1083

mice. (A) Survival curves (left) and body weight loss (right) of BALB/c mice (n=6) that 1084

received 2 or 20 mg/kg of H5M9 on 24 hours before intranasal infection with 10 MLD50 1085

(50% mouse lethal dose) of VN1194 virus. (B) Survival curves (left) and body weight 1086

loss (right) of mice (n=6) that received 2 or 20 mg/kg of H5M9 on 24 hours post 1087

infection with 10 MLD50 of VN11904 virus. 1088

1089

FIG 9 Comparison of antibody epitopes mapped onto the surface of corresponding HAs. 1090

(A) VN1203 H5 HA with the H5M9 footprint shown in green. (B) A/Aichi/2/68 (H3N2) 1091

with the HC45 footprint shown in red (PDB ID code 1QFU). The H3 HA1 domain was 1092

superimposed on the HA1 domain of VN1203 H5 for this comparison. 1093

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

on April 8, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from