JVI Accepted Manuscript Posted Online 11 February 2015 J. Virol. doi:10.1128/JVI.00078-15 Copyright © 2015, American Society for Microbiology. All Rights Reserved.

1

An anti-H5N1 influenza virus FcDART antibody is an highly efficacious therapeutic and

2

prophylactic against H5N1 influenza virus infection.

3 4

Mark Zanin,a Zhen-Yong Keck,b G. Jonah Rainey,c+ Chia-Ying Kao Lam,c Adrianus C.

5

M. Boon,a* Adam Rubrum,a Daniel Darnell,a Sook-San Wong,a Yolanda Griffin,a Jinming

6

Xia,b Robert G. Webster,a Richard Webby,a# Syd Johnson,c Steven Foungb

7 8

Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis,

9

Tennessee, USAa; Department of Pathology, Stanford University School of Medicine,

10

Stanford, California, USAb; MacroGenics Inc., Rockville, Maryland, USAc.

11

12

Running head: FcDART is effective against H5N1 influenza viruses

13

#Address correspondence to Richard Webby, [email protected]

14

15

*Present address: Division of Biology and Biomedical Sciences, Washington University,

16

St Louis, Missouri, USA.

17

+

Present address: MedImmune, LLC. Gaithersburg, MD, USA

18

Abstract word count: 237

19

Importance word count: 128

20

Text word count: 4844

1

21

Abstract:

22

Highly pathogenic H5N1 avian influenza viruses are associated with severe disease in

23

humans and continue to be a pandemic threat. Whilst vaccines are available, other

24

approaches are required for patients that typically respond poorly to vaccination, such

25

as the elderly and the immunocompromised. To produce a therapeutic that is highly

26

efficacious at low doses and is broadly specific against antigenically-drifted H5N1

27

influenza viruses, we developed two neutralizing monoclonal antibodies and combined

28

them into a single bispecific-Fc fusion protein (FcDART). In mice, a single 2.5mg/kg

29

therapeutic or prophylactic dose of either monoclonal antibody provided 100%

30

protection against challenge with A/Vietnam/1203/04 (H5N1) or the antigenically-drifted

31

strain A/Whooper swan/Mongolia/244/05 (H5N1). In ferrets, a single 1mg/kg

32

prophylactic dose provided 100% protection against A/Vietnam/1203/04 challenge.

33

FcDART was also effective, as a single 2.5mg/kg therapeutic or prophylactic dose in

34

mice provided 100% protection against A/Vietnam/1203/04 challenge. Antibodies bound

35

to conformational epitopes in antigenic sites on the globular head of the hemagglutinin

36

protein, based on antibody escape mutations. Whilst it was possible to generate escape

37

mutants in vitro, they were neutralized in vivo by the antibodies, as mice infected with

38

escape mutants were 100% protected after only a single therapeutic dose of the

39

antibody used to generate the escape mutant in vitro. In summary, we have combined

40

the antigen specificities of two highly efficacious anti-H5N1 influenza virus antibodies

41

into a bispecific FcDART, which represents a strategy to produce broadly neutralizing

42

antibodies that are effective against antigenically diverse influenza viruses.

43

2

44

Importance:

45

Highly pathogenic H5N1 avian influenza viruses are associated with severe disease in

46

humans and are a pandemic threat. A vaccine is available but other approaches are

47

required for patients that typically respond poorly to vaccination, such as the elderly and

48

the immunocompromised. The variability of the virus means that such an approach

49

must be broad spectrum. To achieve this, we developed two antibodies that neutralize

50

H5N1 influenza viruses. These antibodies provided complete protection in mice against

51

a spectrum of H5N1 influenza viruses at a single low dose. We then combined the two

52

antibodies into a single molecule, FcDART, which combined the broad spectrum activity

53

and protective efficacy of both antibodies. This treatment strategy represents a novel

54

and effective therapeutic or prophylactic against highly pathogenic H5N1 avian

55

influenza viruses.

56 57

Introduction:

58

Since 2003, a series of events has occurred in Asia that many believe to be the

59

harbinger of an incipient influenza pandemic. The virus responsible for these events, an

60

H5N1 influenza virus, has infected and killed millions of chickens and ducks and

61

infected in excess of 650 people (www.who.int; reviewed in (1)). Fortunately this virus

62

has yet to gain the ability to transmit from human to human, however, mammalian

63

transmissibility can be conferred experimentally with relatively few mutations (2, 3). The

64

virulence of this virus (4, 5), its wide geographic distribution (6) and potential for human

65

aerosol transmission (2, 3) urge the development of targeted therapeutic and

66

prophylactic measures. Currently two H5N1 vaccines have been licensed, one of which

67

is available worldwide, and these offer the best means of mass protection (reviewed in 3

68

(7)). However, it is likely the first line of defense against an emerging pandemic will

69

require additional options, including antivirals. Currently, the two options for treating

70

influenza are the M2 ion channel blockers (e.g., amantadine) and the neuraminidase

71

inhibitors (e.g., oseltamivir). Unfortunately, many of the current H5N1 viruses are

72

resistant to amantadine (6) and a higher dose and longer treatment schedule of

73

oseltamivir is needed for protection in murine models than used previously (8), proof

74

that new therapeutic options are very much needed.

75 76

Monoclonal antibodies against the influenza virus hemagglutinin (HA) protein have

77

shown efficacy as therapeutics and prophylactics in murine challenge models against

78

H5N1 influenza virus infection (9-13) and other antibodies have also shown efficacy in

79

mice against other strains of influenza viruses, including H1N1, H2N2 and H3N2 (14-

80

19). Dosages at which these antibodies provided 100% protection against challenge in

81

mice ranged from 5 to 50mg/kg when delivered therapeutically and 1 to 30mg/kg when

82

delivered prophylactically. In ferrets, an anti-H5 antibody provided 100% protection

83

against challenge with an H5N1 influenza virus at 10mg/kg delivered prophylactically

84

and at 30mg/kg delivered therapeutically (20).

85 86

The variability of the HA protein, which contains the main antigenic sites of the virus,

87

necessitates that any broadly utilized antibody must be effective against antigenically

88

diverse influenza viruses. The most conserved regions of HA are the stalk and the

89

receptor-binding pocket, and antibodies directed at these sites are efficacious against

90

antigenically diverse strains of influenza virus (14, 21, 22). However, the most exposed

91

and antibody-accessible epitopes on HA are located on the globular head, although 4

92

these regions are highly variable. Due to the variability of HA, it is likely that more than

93

one neutralizing antibody will be required to provide broad protection against different

94

lineages of H5N1 and to protect against antibody escape mutants. However, this

95

presents certain problems for clinical testing and product development as cocktails of

96

antibodies are difficult and expensive to produce and test. Therefore, in this study we

97

have developed a bi-specific Fc fusion protein, Fc Dual-Affinity Re-Targeting (FcDART),

98

that combines the neutralizing capacity and specificities of two different neutralizing

99

monoclonal antibodies from human and murine sources that target the globular head of

100

HA. Individually, the two monoclonal antibodies provided 100% protection against

101

A/Vietnam/1203/04 (H5N1) (VN1203) challenge in mice and ferrets as therapeutics and

102

prophylactics at low doses (2.5mg/kg in mice and 1mg/kg in ferrets). They also provided

103

100% protection in mice against challenge with A/Whooper swan/Mongolia/244/05

104

(H5N1) (Mon244), which is an antigenically drifted strain. Further, antibody escape

105

mutants generated in vitro were not lethal in mice treated with a single therapeutic dose

106

of antibody. The FcDART molecule that combined the antigen specificities of these two

107

antibodies also provided 100% protection against challenge in mice as a therapeutic or

108

prophylactic and represents a strategy to produce antibody-based therapeutics that are

109

effective against antigenically diverse influenza viruses.

110 111

Materials and Methods:

112

Cells Lines and Culture Conditions. 293T, Chinese Hamster Ovary (CHO) and Madin

113

Darby canine kidney (MDCK) cell lines were obtained from ATCC. 293T cells were

114

grown in Opti-MEM (Invitrogen, CA) supplemented with 5% fetal calf serum (FCS,

115

Gemini Bioproducts Inc, CA), CHO cells were grown in F12K medium (Invitrogen, CA) 5

116

supplemented with 10% FCS and MDCK cells were grown in Dulbecco’s modified

117

minimal essential medium (DMEM, Invitrogen, CA) supplemented with 10% FCS and 2

118

mM glutamine. For MDCK cell infections, viruses were diluted in infection medium

119

(MEM supplemented with 5% (v/v) bovine serum albumin (BSA) and 2 mM glutamine

120

(Sigma, MO)).

121 122

Immunofluorescence Assay (IFA). MDCK cells were infected with VN1203 or Mon244

123

overnight. The cells were harvested by trypsinization and resuspended in PBS with 2%

124

FCS. Aliquots containing 3x104 cells were spotted onto HTC Super Cured 24-spot

125

slides (Erie Scientific Company, NH), dried, and fixed with 100% acetone for 10 min at

126

room temperature. Fixed cells were incubated with hybridoma supernatants for 30 min

127

at 37C and washed for 5 min with PBS. Slides were then incubated for 30 min at 37C

128

with 50 ng/mL propidium iodide and fluorescein isothiocyanate (FITC)-conjugated goat

129

anti-human IgG (Jackson ImmunoResearch Laboratories, West Grove, PA). Bound

130

antibody was revealed by fluorescence microscopy.

131 132

Hemagglutination inhibition (HI) assay. HI assays were conducted using standard

133

methodologies. In brief, 25 l of diluted antigen at four agglutination doses in PBS was

134

added to wells of 96 well plates containing a two-fold dilution series of the test antibody.

135

After 30 minutes incubation at room temperature, 50l of 0.5% (v/v) chicken or horse

136

red blood cells was added to each well and incubated at room temperature for another

137

30 min. Titers were recorded as the lowest dilution of antibody able to inhibit

138

hemagglutination.

139

6

140

Microneutralization (MN) assay. MN assays were conducted using MDCK cells

141

according to standard methodologies. In brief, a two-fold dilution series of each antibody

142

was incubated with virus at 100TCID50/50µL for one hour at 37oC. The antibody/virus

143

solutions were then added to MDCK cells for an additional hour at 37oC and were then

144

washed off and the cells incubated at 37oC for 72hrs with 200µL infection medium

145

containing 1µg/mL TPCK trypsin. Neutralizing titers were read by incubating 50µL of cell

146

culture medium with 0.5% (v/v) chicken or horse red blood cells followed by incubation

147

at room temperature for another 30 min and were expressed as the reciprocal of the

148

serum dilution that inhibited 50% of viral growth of 100TCID50 of virus.

149 150

Human Monoclonal Antibody Generation. Plasma samples were obtained from

151

individuals immunized with a recombinant, baculovirus-expressed HA protein from the

152

A/Hong Kong/156/97 (H5N1) virus (23) followed by the inactivated, subvirion vaccine

153

against VN1203 (24). Samples were screened by HI or by IFA using H5N1 (VN1203)

154

HA protein or H5N1-infected MDCK cells, respectively. In this study, a donor with serum

155

HI titer of 320 was used as the source of peripheral B-cells for human hybridoma

156

production as previously described (25). Three hybridomas, designated as BF1-1, BF1-

157

10 and BF1-19, were identified by HI assay secreting neutralizing H5N1 human

158

monoclonal antibodies (HMAbs). Against a panel of Southeast Asian H5N1 isolates,

159

these three donors showed reactivity to the antigenically distinct H5N1 virus HK156 but

160

not to the clade 2.3.4 virus A/Common magpie/Hong Kong/645/06. BF1-32 was

161

identified by IFA. Monoclonality was confirmed by sequencing IgG genes isolated from

162

10 individual cell clones derived from each hybridoma cell line. Cloning and analyzing

163

the VL and VH domains of these clones were performed as previously described (26). 7

164

HMAb production and purification were performed as described (25) and biotinylation of

165

the antibodies was carried out according to manufacturer’s instructions (Pierce

166

Biotechnology, Inc, Rockford, IL). The cell lines produced IgG1 antibody with λ light

167

chain for BF1-1 and κ light chains for BF1-10, BF1-19 and BF1-32 and secreted 10-40

168

µg human IgG per mL in spent cultured supernatant.

169 170

Murine Monoclonal Antibody Generation. Generation of murine monoclonal antibodies

171

(mMAbs) was achieved via a vaccination regimen consisting of an initial dose of 0.5

172

mouse lethal dose 50% (MLD50) of a reverse-genetics virus bearing the HA and

173

neuraminidase genes of VN1203 and the six internal genes of A/Puerto Rico/8/1934

174

(H1N1) (rgH5N1) delivered intranasally (IN) followed by two booster doses delivered

175

intraperitoneally (IP)

176

hemagglutination units (HAU) of Mon244 delivered IP followed by 1000 egg infectious

177

dose 50% (EID50) of rgH5N1 delivered intravenously (IV). Post vaccination, hybridomas

178

were produced by Rockland Immunochemicals and were screened by HI and IFA as

179

described above.

four

weeks apart. Booster

doses consisted

of

10,000

180 181

Humanization of Murine Monoclonal Antibodies. mMAbs were humanized by first

182

obtaining cDNA of the heavy (VH) and light (VL) variable segments from total RNA via

183

3’ RACE followed by cloning into the pCR2.1-topo vector (Life Technologies), which

184

was then transformed into E.coli, recovered and sequenced. The VH and VL segments

185

were also combined with human Cg1 and K constant region cDNA segments in the

186

mammalian expression vectors pEE13.4 (LC) and pEE6.4 (HC) (Lonza), then combined

187

into a single expression vector and the resulting chimeric HC and LC were then 8

188

transfected into CHO-S cells (Life Technologies). Conditioned media from transfected

189

cells was assayed by ELISA for human IgG production and anti-H5N1 activity. Human

190

germLine genes with the highest homology to the VH and VL segments were identified

191

using Ig BLAST and humanized VH and VL segments were then designed using CDR

192

grafting, VH1-18/VH1-69 was used for VH frameworks, and VK-O12 used for VL for

193

humanization of 10C3 and humanized VH and VL segments were then synthesized by

194

gene synthesis (GenScript). The resulting fragments were then cloned into the

195

mammalian expression vector pCIneo (Promega) with the appropriate C region and

196

clones were sequenced.

197 198

Generation of the FcDART. The antigen specificities of BF1-19 and h10C3 were

199

combined into a bispecific Fc fusion protein, termed FcDART. This was achieved by

200

joining the two antibody chains such that each chain of the FcDART comprised of the

201

VL from one antibody and the VH of the second antibody and vice-versa, with a short

202

linker (GGGSGGG) between the VL and VH. One of the two chains was then fused to a

203

human IgG1 hinge-Fc region (figure 1A). The two chains were transfected into CHO

204

cells and a stable pool of producing cells was selected. The material was purified over a

205

protein A column followed by size exclusion chromatography (figure 1B).

206 207

Affinity Measurement. The binding affinity of each monoclonal antibody (MAb) to H5N1

208

HA was characterized by surface plasmon resonance (SPR) analysis. The experiments

209

described below were carried out using the BIAcore 3000 system (Biacore). Affinity

210

purified anti-human IgG, Fcγ (Jackson Immuno Research) was amine coupled (10,000

211

RU) to all four flow cells of a CM5 sensor chip (Biacore). MAb samples at 0.5-1 µg/mL in 9

212

running buffer were injected into flow cells 2-4 while keeping flow cell 1 as a reference,

213

which led to roughly 150 RU of MAb being captured in each flow cell. Association

214

kinetics were observed by injecting purified H5N1 HA (Protein Sciences) in running

215

buffer through all four flow cells at a flow rate of 30 µl/min for a duration of 90 seconds.

216

Disassociation kinetics were then measured with running buffer flowing at a rate of 30

217

µl/min for a duration of 10 minutes. Flow cells were regenerated using a 15 µl injection

218

of 100mM H3PO4. Kinetics were tested at HA concentrations of 800, 400, 200, 100, 50,

219

25, 12.5, 6.25, 3.125, 1.56 nM. After each binding cycle, the same protocol was run with

220

running buffer substituted for the analyte to serve as a double reference. Data were

221

analyzed using BIAevaluation software version 4.1 (Biacore). The data were processed

222

by subtracting the double reference and trimming out MAb loading and H3PO4

223

regeneration information. Kinetic values were obtained by applying the 1:1 (Langmuir)

224

binding, simultaneous ka/kd, fit method.

225 226

Antibody Binding Profiles. Direct (antigen down) or capture (antibody down) ELISA

227

formats were used to measure antibody binding to HA from different clades of H5N1

228

influenza viruses. In antigen down format, HA protein was coated on an ELISA plate at

229

a concentration of 2µg/mL. HA protein was obtained from eEnzyme and Protein

230

Sciences and was produced in 293 cells or an insect cell line, respectively. HA was

231

uncleaved or cleaved at the furin cleavage site (furin). The indicated concentration of

232

antibody was then added to the plate and detection was performed using an

233

horseradish

234

chemiluminescent detection (SuperSignal, Pierce). For the MAb-Down assays, an

235

antibody specific for CH1 (4A11, Abcam) was used to coat an ELISA plate at 2 µg/mL.

peroxidase

(HRP)-conjugated

anti-human

antibody,

followed

by

10

236

The indicated concentration of antibody was then captured. His-tagged antigen

237

(100ng/mL) was then applied to the plate, followed by an HRP-conjugated anti-His

238

antibody, followed by chemiluminescent detection (SuperSignal, Pierce). To detect

239

binding of h10C3, this assay was performed in an antibody down format to maximize

240

sensitivity using antibody captured with polyclonal anti-human IgG and antigen binding

241

was detected with streptavidin-HRP and developed colorimetrically with 3,3’,5,5’-

242

Tetramethylbenzidine (TMB, Sigma).

243 244

Prevention and Therapeutic Treatment in animals. All animal studies were conducted

245

under applicable laws and guidelines of and after approval from the St. Jude Children’s

246

Research Hospital Animal Care and Use Committee. Female 6-8-week old BALB/c mice

247

were housed in cages with 5 mice per cage. Mice received the indicated amount of

248

antibody in a total volume of 300µL sterile PBS by intraperitoneal (IP) injection. At

249

various times pre and post antibody administration mice were lightly anesthetized with

250

isoflourane and inoculated intranasally with 100 MLD50 of VN1203 (1000 EID50) or 100

251

MLD50 (5x105 EID50) of Mon244 in 30µL of PBS.

252 253

Male 4 to 5-month-old ferrets were housed in cages with three animals per cage and

254

were administered 1mg/kg of indicated antibodies in 1mL PBS intramuscularly and on

255

the next day ferrets were lightly anaesthetized with isoflurane and administered 3×104

256

EID50 of VN1203 in 1mL of PBS IN. For all animals, weight change and survival were

257

monitored for 14 days following virus challenge.

258

11

259

Isolation and characterization of antibody escape mutants. Antibody escape mutants

260

were selected by inoculating 10-day-old specific pathogen free (SPF) embryonated

261

chicken eggs with rgH5N1 incubated with neutralizing antibody essentially as described

262

previously (27, 28). Eggs were screened for the presence of virus by hemagglutination

263

assay on allantoic fluid, which was harvested from infected eggs and viral RNA was

264

extracted and used to generate cDNA by RT-PCR. The HA gene was then amplified

265

and sequenced.

266 267

Data analysis and statistics. Data collected were inputted and graphed using Graphpad

268

Prism version 5.03. Statistical analysis was performed using the log-rank (Mantel-Cox)

269

test.

270 271

Results:

272

Characterization of H5N1 HMAbs. To determine the genetic diversity of the antibodies

273

identified, we sequenced the CDR 1, 2 and 3 regions of the antibody heavy chains.

274

Sequence analysis of the 11 HI-positive hybridomas showed that they clustered into two

275

different groups. Two HMAbs clustered into group one while the others clustered into

276

the second group. One antibody representing group one (BF1-10), and two representing

277

group two (BF1-1 and BF1-19) were chosen for further analysis. HI assays were

278

performed with spent supernatants from human hybridomas to assess the cross

279

reactivity of the generated antibodies against a panel of H5N1 viruses isolated in Asia.

280

All antibodies were reactive against VN1203, showing titers of 640 to 2560 (table 1).

281

BF1-19 also showed reactivity against the antigenically drifted strain Mon244 (HI=40),

282

whilst BF1-1 and BF1-10 did not (table 1). No antibodies showed reactivity against 12

283

A/Japanese white eye/Hong Kong/1038/06 (HK1038), which is antigenically drifted from

284

VN1203 to a greater degree than Mon244 (table 1). BF1-19 showed strong reactivity

285

against A/Hong Kong/156/97 (HK156) (HI=1280) whilst BF1-1 and BF1-10 showed only

286

weak reactivity (HI=10 and 20, respectively) (table 1). All antibodies also showed strong

287

neutralization activity in microneutralization assay against VN1203, with titers of 640 to

288

10240, however no neutralization activity was observed to Mon244 or HK1038 (table 2).

289

BF1-19 showed strong microneutralization activity against HK156 (titer>20,480), whilst

290

BF1-1 and BF1-10 showed only weak activity (titer=10 and 40, respectively) (table 2).

291 292

To determine the nature of the epitopes recognized by each HI-positive HMAb, Western

293

Blot analysis was performed using rgH5N1-infected MDCK cell lysate. All neutralizing

294

HMAbs showed no binding to denatured H5N1 antigens, suggesting that they recognize

295

conformational epitopes. The one HI-negative HMAb that was included in this study,

296

BF1-32, was Western blot-positive and was identified by IFA with VN1203-infected

297

MDCK cells. This antibody appears to target HA1, as the most prominent bands

298

corresponded to HA0 (82 Kda) and HA1 (58 Kda), with a less intense band at 78 Kda

299

(figure 2). To assess the affinities of each of the HMAbs for the H5 HA protein, surface

300

plasmon resonance studies were conducted using a BIAcore 3000 instrument. The KD

301

values for the binding of HMAbs BF1-1, BF1-10, and BF1-19 to the VN1203 HA were

302

20.3nM, 10.8nM, and 48.2nM, respectively, showing that each had similar affinities for

303

their antigens. Following these studies, we selected BF1-19 for subsequent

304

experiments.

305

13

306

We next assessed the breadth of reactivity of BF1-19 to a panel of HAs from diverse

307

H5N1 viruses via ELISA. We used antigens obtained from two commercial sources,

308

eEnzyme and Protein Sciences, to investigate whether the antigen production system

309

had an effect on the ability of the antibodies to bind, as the eEnzyme antigens were

310

produced in 293T cells whilst the Protein Sciences antigens were produced in insect

311

cells. Further, we used both wild type (wt) HA antigens and antigens where the furin

312

cleavage site were deleted (furin), to determine if cleavage had an impact on the

313

exposure of the epitope recognized by these antibodies. All eEnzyme antigens with the

314

exception of A/Indonesia/5/2005 (H5N1) were engineered to remove the furin cleavage

315

site, resulting in a more homogeneous product (figure 3). The Protein Sciences antigens

316

and the eEnzyme A/Indonesia/5/2005 (H5N1) antigen retained the furin cleavage site,

317

resulting in oligomers that consisted of both cleaved and uncleaved antigens. Antigens

318

obtained from Protein Sciences were tested in ‘antibody down’ format, where the

319

antibody was bound to the solid phase via a capture antibody and the antigen was the

320

soluble binding partner (figure 3). eEnzyme antigens contained a C-terminal HIS tag,

321

which allowed them to be used as a soluble binding partner or to be immobilized on the

322

solid-phase using a HIS binding partner, termed ‘antigen down’ (figure 3).

323 324

BF1-19 bound to Clade 1 antigens from both suppliers in an antigen down format (figure

325

3A) and also bound to Clades 2.2.1 and 2.3.4 antigens in antibody down format (figure

326

3B). Some binding to Clade 2.1.3.2 antigen was also detected at high concentrations

327

(figure 3B). Given the oligomeric structure of the antigens, the ability to detect binding in

328

antibody down format may be the result of more avid binding compared to the antigen

329

down format. BF1-19 also bound to Clade 1 antigens with or without the furin cleavage 14

330

site (figure 3A). Whilst these data are not able to definitively demonstrate a quantitative

331

difference between cleaved and uncleaved HA, they do clearly demonstrate that the

332

recognized epitope is available on oligomers derived from both wt and furin constructs.

333 334

Generation of mMAbs. We used a three dose vaccination regimen in mice, consisting of

335

VN1203 delivered IN then IP followed by Mon244 delivered IV, to generate antibodies

336

broadly specific to different clades of H5 influenza viruses. One of these antibodies,

337

10C3, bound to Clades 1 and 2.2.1 antigens from both suppliers in antigen down format

338

and, like BF1-19, bound to both wt and furin constructs (figure 3C). Similar results

339

were also observed in antibody down format (figure 3D).

340 341

10C3 was then humanized to reduce potential immunogenicity by grafting the

342

complementarity determining regions of the antibody onto the framework regions from

343

closely related human VH and VL gene sequences. Interestingly, there were indications

344

of h10C3 binding to Clade 2.1.3.2 and 2.3.4 antigens, which was not observed in 10C3

345

(figure 3E). Whilst this data is not conclusive, it is indicative that the humanization

346

process broadened the coverage of this antibody (figure 3D and E). By HI, h10C3 was

347

strongly reactive to VN1203 (HI=2560) and showed relatively weak reactivity against

348

Mon244 (HI=40) and no reactivity against HK1038. Unlike BF1-19, h10C3 did not show

349

reactivity against HK156 (table 1). In microneutralization assays, h10C3 only showed

350

neutralization activity against VN1203 (titer=2560), no neutralization activity was

351

observed against HK156, Mon244 or HK1038 (table 2).

352

15

353

Design and Development of the FcDART. The FcDART molecule consisted of two

354

polypeptide chains that heterodimerized to form a bi-specific molecule. Each chain

355

contained the VL of one specificity and the VH of the other, such that heterodimerization

356

of the two chains results in assembly of matching VL/VH domains. In order to favor

357

heterodimeric assembly, a strong heterodimerization domain has been appended to the

358

C-terminus of each chain. These domains consisted of coiled coil-forming sequences

359

where the hydrophobic core of the coiled coil interface was flanked by opposite charges

360

(E- on one coil and K+ on the other). These charges are believed to favor

361

heterodimerization by repelling homotypic association and preventing the initiation of

362

“zippering” of the hydrophobic core (figure 1A). These DART molecules were modified

363

further by appending an Fc domain to the C-terminus of the E-coil, causing two DART

364

units to dimerize and form a tetravalent bispecific structure (figure 1A). The addition of

365

the Fc confers long serum half-life on the molecules and restores the avidity associated

366

with the bivalent parental mAbs. Further, the presence of the Fc confers protein A

367

binding and makes these molecules amenable to platform purification processes

368

routinely used for mAbs in large scale cGMP production.

369 370

Following expression in CHO cells and purification, analytical size exclusion

371

chromatography showed that purified material consisted of predominantly one species

372

that eluted at a volume consistent with the predicted molecular mass of FcDART. Only a

373

small amount of higher order aggregate was present, as indicated by the slight left

374

shoulder on the peak. Increasing the concentration from 2 to 24.5mg/mL did not cause

375

a significant increase in the amount of aggregated material (figure 1B). These

376

characteristics indicate that the molecule was properly assembled, homogeneously 16

377

purified and not prone to aggregation. We then tested the FcDART against a panel of

378

antigens in antibody down format and found that, with the exception of Clade 2.1.3.2,

379

the FcDART was able to bind to the antigens in the panel and was generally consistent

380

with the binding profile of BF1-19 and h10C3, indicating that FcDART indeed combined

381

the binding profile of BF1-19 and h10C3 (figure 3F). By HI, FcDART showed reactivity

382

against VN1203 and HK156 (HI=640), relatively weak reactivity against Mon244 (HI=40)

383

and no reactivity against HK1038, which is again consistent with a binding profile that is

384

a combination of BF1-19 and h10C3 (table 1). The neutralization activity of FcDART

385

was also consistent with an antibody with the combined specificities of h10C3 and BF1-

386

19, as strong neutralization activity was observed against both VN1203 (titer=1280) and

387

HK156 (titer=10240) (table 2). Interestingly, FcDART also showed neutralization activity

388

against Mon244 (titer=80), which was not observed with any other antibody tested,

389

suggesting an increased breadth of specificity (table 2).

390 391

Escape mutant characterization. To further characterize the epitopes recognized by

392

these antibodies, we inoculated embryonated chicken eggs with rgH5N1 and antibody

393

and sequenced viruses present after incubation for 48hrs. Based on the mutations in

394

these viruses, MAbs primarily bound to antigenic sites on the globular head of HA (29).

395

L128S in antigenic site C and M265I in antigenic site A were present in escape mutants

396

to h10C3, K139E in antigenic site A was in escape mutants to 10C3, S140Y/F in

397

antigenic site A were in escape mutants to BF1-19, K188N/E in antigenic site B were in

398

escape mutants to BF1-19 and FcDART, T213A in antigenic site D was in escape

399

mutants to FcDART (figure 4). With the exception of h10C3, where L128S and M265I

400

were present in one mutant, each escape mutant did not harbor more that one of the 17

401

mutations indicated in figure 4. Further, escape mutants harboring the same mutation

402

were seen in all eggs (n=3) at the dilution of antibody that was permissive to the

403

generation of escape mutants.

404 405

We next performed HI assays to determine if binding of the antibodies to these escape

406

mutants was diminished or abrogated. BF1-19 showed little activity against the BF1-19

407

escape mutants K188N and S140F (HI titers

An anti-H5N1 influenza virus FcDART antibody is a highly efficacious therapeutic agent and prophylactic against H5N1 influenza virus infection.

Highly pathogenic H5N1 avian influenza viruses are associated with severe disease in humans and continue to be a pandemic threat. While vaccines are a...
7MB Sizes 16 Downloads 11 Views