• Aucun résultat trouvé

Naturally-occurring capsid protein variants of human papillomavirus genotype 31 represent a single L1 serotype

N/A
N/A
Protected

Academic year: 2022

Partager "Naturally-occurring capsid protein variants of human papillomavirus genotype 31 represent a single L1 serotype"

Copied!
35
0
0

Texte intégral

(1)

Naturally-occurring capsid protein variants of human papillomavirus genotype 31 1

represent a single L1 serotype.

2 3

Sara L. Bissett,1 Anna Godi,1 Maxime J. J. Fleury,2 Antoine Touze,3 Clementina 4

Cocuzza,4 and Simon Beddows1#

5

6

1 Virus Reference Department, Public Health England, London, UK 7

2 GEIHP, UPRES EA 3142, Université d’ Angers, Angers, France 8

3 UMR 1282, Université François Rabelais/INRA, Tours, France 9

4 Department of Surgery and Translational Medicine, University of Milano-Bicocca, 10

Monza, Italy 11

12

# Corresponding author: Simon Beddows, Public Health England, 61 Colindale Avenue, 13

London NW9 5EQ, UK. Tel: +44 (0) 20 8327 6169. simon.beddows@phe.gov.uk 14

15

Running Title: HPV31 capsid protein variants (Max 54 characters including spaces) 16

Key words: Human Papillomavirus, L1, L2, Variant, Antibody, Vaccine, Neutralization, 17

Serotype 18

Abstract: 126 19

Main text: 3,956 20

JVI Accepted Manuscript Posted Online 20 May 2015 J. Virol. doi:10.1128/JVI.00842-15

Copyright © 2015, American Society for Microbiology. All Rights Reserved.

(2)

Abstract 21

We investigated naturally-occurring variation within the major (L1) and minor (L2) capsid 22

proteins of oncogenic human papillomavirus (HPV) genotype HPV31 to determine the 23

impact on capsid antigenicity. L1L2 pseudoviruses (PsV) representing the three HPV31 24

variant lineages A, B and C exhibited comparable particle to infectivity ratios and 25

morphology. Lineage-specific L1L2 PsV demonstrated subtle differences in 26

susceptibility to neutralization by antibodies elicited following vaccination, pre-clinical L1 27

VLP immunization or by monoclonal antibodies; however, these differences were 28

generally of a low magnitude. These data indicate diagnostic lineage-specific single 29

nucleotide polymorphisms within the HPV31 capsid genes have a limited effect on L1 30

antibody mediated neutralization and that the three HPV31 variant lineages belong to a 31

single L1 serotype. These data contribute to our understanding of HPV L1 variant 32

antigenicity.

33 34

Importance 35

The virus coat (capsid) of the human papillomavirus contains major (L1) and minor (L2) 36

capsid proteins. These proteins facilitate host cell attachment and viral infectivity and 37

are the targets for antibodies which interfere with these events. In this study we 38

investigated the impact of naturally-occurring variation within these proteins upon 39

susceptibility to viral neutralization by antibodies induced by L1 VLP immunization. We 40

demonstrate that HPV31 L1 and L2 variants exhibit similar susceptibility to antibody- 41

mediated neutralization and that for the purposes of L1 VLP-based vaccines these 42

variant lineages represent a single serotype.

43

(3)

Introduction 44

Human papillomaviruses (HPV) comprise a double-stranded DNA genome of 45

approximately 8 kb which is replicated via host cell polymerases with an error rate of ca.

46

2x10-8 base substitutions, per site, per year (1), substantially lower than that found in the 47

majority of single-stranded RNA viruses (ca. 1x10-3 base substitutions/site/year) (2).

48

Despite the low evolutionary rate of the HPV genome, variants have arisen over time 49

leading to the generation of distinct intra-genotype lineages classified by a sequence 50

difference of 1-10% across the whole genome (3). The single nucleotide polymorphisms 51

(SNP) that allow segregation of these variants into distinct lineages can be found in 52

each gene/region, with the highest number accumulating in the noncoding regions 53

(NCR1, NCR2 and URR) and the lowest in structural genes (L1 and L2) (4).

54 55

The HPV structural genes encode the major (L1) and minor (L2) proteins which form the 56

non-enveloped icosahedral viral capsid that comprises 72 pentameric L1 capsomers 57

with upper estimates of one L2 protein per capsomer (5). The L1 protein mediates 58

attachment to host cells (6) whilst the L2 protein is essential for subsequent viral 59

infectivity (7).

60 61

The humoral immune response following natural HPV infection predominately targets 62

conformational epitopes on the surface-exposed loop regions of the L1 protein (8, 9).

63

Seroconversion generally occurs 6-18 months after infection with low levels of L1 64

antibodies detected in 50-70% of individuals (10, 11). It is not clear whether antibodies 65

(4)

induced by natural infection protect against subsequent reinfection by the same HPV 66

genotype but increasing evidence indicates that high antibody titers can be associated 67

with a reduced risk of reinfection (12-15).

68 69

The L1 protein can self-assemble into virus-like particles (VLP) which are the basis of 70

the current prophylactic HPV vaccines, Cervarix® and Gardasil® (16). Clinical trials have 71

demonstrated the high efficacy of both vaccines against infection and cervical disease 72

associated with vaccine genotypes HPV16 and HPV18. A degree of vaccine-induced 73

cross-protection has also been demonstrated against closely-related genotypes, 74

particular HPV31, HPV33 and HPV45 (16-18). HPV vaccine type-specific protection is 75

assumed to be mediated by L1 neutralizing antibodies which can be detected in the 76

serum and cervicovaginal secretions of vaccinees (16, 19-22). The role of L1 77

neutralizing antibodies in mediating cross-protection is less clear although a recent 78

study reported an association between the presence of HPV31 cross-neutralizing 79

antibodies and a reduced risk of HPV31 infection (23). Next generation L1 VLP-based 80

vaccines aim to extend the breadth of coverage by incorporating an increased number 81

of L1 VLP (24, 25).

82 83

Intra-genotype variation within the L1 protein is generally localized to the surface- 84

exposed loop domains (26), akin to the majority of inter-genotype variation (27, 28).

85

Data informing the potential impact of such variation on L1 antigenicity are limited to 86

HPV16 where L1L2 pseudoviruses (PsV) representing lineage-specific L1 variants were 87

neutralized to a similar extent by antibodies elicited against a single L1 VLP (29).

88

(5)

However, the impact of a common Thr to Ala switch at amino acid residue position 266 89

within the FG loop of HPV16 was not evaluated, nor was variation within the 90

corresponding L2. Another study found that an FG loop specific neutralizing monoclonal 91

antibody (MAb), H16.E70, had reduced recognition for HPV16 L1 VLP bearing a Thr at 92

amino acid residue 266 indicating that variation within this region can impact upon 93

antigenicity (30).

94 95

HPV31 is closely related to HPV16 within the Alpha-9 species group and is associated 96

with ca. 3.8% of cervical cancer cases worldwide (31). The full genome sequence 97

analysis of HPV31 has led to the delineation of three distinct variant lineages: A, B and 98

C (4). Infections due to HPV31 lineage variants A or B have been associated with an 99

increased risk of developing cervical intraepithelial neoplasia grades 2–3 (CIN2/3) yet 100

somewhat paradoxically infections with lineage variant C appear to persist for longer 101

(32, 33). Differences in the natural history between variants of other HPV genotypes 102

have also been observed (34, 35). The relative infectivity of variants or the ability of 103

variants to differentially disrupt cellular differentiation are possible virological factors 104

which can contribute to the disparities in variant pathology (3). One study suggested 105

that the genetic background of the host may also play a role since African-American 106

women were found to be less likely to clear a HPV31 lineage variant C infection than a 107

lineage variant A infection, yet there was no difference in the likelihood of clearing 108

lineage variant A over C in Caucasian women (33).

109 110

(6)

Two non-synonymous, lineage-specific SNP within the L1 of HPV31 are located within 111

the FG loop at positions 267 and 274 (26). The FG loop of HPV31 has been shown to 112

be an important antigenic domain targeted by both type-specific and cross-reactive L1 113

MAbs (36, 37). In the present study we generated additional HPV31 L1 and L2 114

sequences and synthesized representative antigens in order to investigate the potential 115

impact of this variation. Such data should improve our understanding of the potential 116

biological impact of naturally-occurring HPV31 variation.

117

(7)

Materials and Methods 118

119

Study samples 120

Residual vulva-vaginal samples collected from 16-24 year old females undergoing 121

chlamydia testing in England which had previously been confirmed as HPV31 DNA 122

positive using the Hybrid Capture 2 HPV DNA test (Qiagen) and the Linear Array HPV 123

Genotyping test (Roche) (38) were selected for L1 and L2 sequencing. Cervical cell 124

samples from HPV31 DNA positive women (aged 19 – 76 years) attending 125

gynaecological care at the San Gerardo hospital (Milan, Italy: Ethical approval study 126

code: 08/UNIMIB-HPA/HPV1) following a cytological diagnosis of ASCUS (atypical 127

squamous cells of undetermined significance) or LSIL (Low grade squamous 128

intraepithelial lesion) were available for L1 and L2 sequencing. Serum samples were 129

available from HPV31 DNA positive women within this cohort. Serum samples were 130

available from 12-15 year old girls one month after receiving three doses of Cervarix® or 131

Gardasil® HPV vaccine (20). A panel of HPV31 MAbs were available as either ascites 132

fluid or tissue culture supernatant (39).

133 134

Sequencing of HPV31 capsid genes 135

The L1 (base pair 5443-7119 (40) or FG loop region 6141-6476; numbered according to 136

the HPV31 reference, J04353) and L2 (3921-5725) genes were amplified with Platinum 137

Taq® High Fidelity DNA polymerase (Life Technologies) and sequenced using the ABI 138

Genetic Analyzer 3730. Sequence data were collated using DNASTAR Lasergene® 9.0 139

(8)

software (DNASTAR, Inc). The HPV31 L1 and L2 sequences generated in this study 140

were assigned the following GenBank accession numbers: KJ754561 – KJ754580.

141

Additional HPV31 L1 and L2 sequences were downloaded from the National Center for 142

Biotechnology Information [NCBI: http://www.ncbi.nlm.nih.gov/, accession numbers:

143

HQ537666 - HQ537687 (4), U37410 (41) and J04353 (42)] and analyzed using the 144

Neighbour-Joining tree algorithm with bootstrap values (n=500 iterations) generated 145

using MEGA v6 (43). HPV31 L1 variant residues were mapped to the surface of the 146

HPV16 capsomer crystal structure (PDB code: 2R5H) and analyzed using Swiss-PDP 147

viewer v4.0 (Deep View) (44).

148 149

L1 VLP 150

HPV31 L1 VLP were expressed using the Bac-to-Bac® Baculovirus System (Life 151

Technologies) and purified on a iodixanol (Sigma-Aldrich) gradient as previously 152

described (45). The L1 protein was visualized by SDS-PAGE stained with SimplyBlue™

153

SafeStain (Life Technologies) alongside a standard curve derived from known input 154

concentrations of bovine serum albumin. Gel analysis was carried out using ImageJ 155

software (U. S. National Institutes of Health, http://imagej.nih.gov/ij) to determine the L1 156

concentration of the gradient fractions. VLP formation was confirmed by electron 157

microscopic analysis of negatively stained particles. The HPV31 L1 VLP shared a 100%

158

amino acid sequence identity with the L1 gene of the HPV31 reference sequence 159

(J04353) of lineage variant A. QuikChange® Site-Direct Mutagenesis (Stratagene) was 160

employed to generate L1 sequences representing lineage variants HPV31 B and 161

HPV31 C. The L1 VLP were used as target antigens in an ELISA, as previously 162

(9)

described (20, 45). The panel of HPV31 MAbs were tested at a standardised input 163

concentration of 250µg/mL of mouse IgG, except 31.D24 (starting input 20µg/mL). The 164

MAbs were subjected to serial dilutions and the IgG concentration which resulted in 165

50% maximal binding optimal density (OD) was estimated by interpolation and the 166

results presented as a 50% binding concentration.

167 168

Mouse immunizations 169

VLP were adsorbed onto aluminium hydroxide (Alhydrogel, Brenntag Biosector) before 170

addition of the Monophosphoryl Lipid A (MPL) based Sigma Adjuvant System (Sigma- 171

Aldrich). BALB/c mice were injected intramuscularly with 2μg of VLP on Day 0 and Day 172

14, before a terminal bleed was taken at Day 21. Pre-bleeds were taken from all mice 173

prior to initial immunization. A total of ten mice were immunized with either HPV31 A 174

VLP, HPV31 B VLP or HPV31 C VLP over three separate immunization schedules. All 175

animal husbandry and procedures were carried out in strict accordance with UK Home 176

Office guidelines and governed by the Animals (Scientific Procedures) Act 1986 and 177

performed under licences PPL 70/7412 & 70/7414.

178 179

L1L2 pseudoviruses 180

A bicistronic psheLL vector (46) containing codon-optimized HPV31 L1 and L2 genes 181

representing the HPV31 reference (J04353) of lineage variant A was expressed and 182

purified on a iodixanol (Sigma-Aldrich) gradient as previously described (47). The L1 183

and L2 gene representing lineage variants HPV31 B and HPV31 C were either 184

(10)

synthesisedby GeneArt® (Life Technologies) or generated by QuikChange® Site-Direct 185

Mutagenesis (Stratagene). Particle formation and size were determined by electron 186

microscopic analysis of negatively stained particles. The L1 concentrations of PsV 187

stocks were estimated by semi-quantitative L1 western blot using CamVir-1 (abcam, 188

U.K.) and the Tissue Culture Infectious Dose 50% (TCID50) was estimated using the 189

Spearman-Karber equation as previously described (47). Particle to infectivity (PI) ratios 190

were determined based on an estimated 3x107 particles per ng L1 protein 191

(http://home.ccr.cancer.gov/lco/production.asp) with the ratio normalized for input 192

volume and TCID50. The presence of L2 protein and reporter gene (Luciferase) in 193

purified PsV stocks was confirmed by qualitative L2 western blot using HPV16 L2 anti- 194

peptide sera (amino acids 17-36) and qualitative PCR (base pair 1222-1641; pGL4.51, 195

Promega) following DNA extraction (QIAamp DNA Blood Mini Kit, Qiagen), respectively.

196

The PsV neutralization assay was performed as previously described (48) with minor 197

modifications (47). A standardized input of 100 TCID50 was used for all PsV and 198

samples were subjected to serial dilutions with the antibody titer or concentration 199

resulting in a 80% reduction of the luciferase signal (RLU) produced by the control wells 200

containing PsV only estimated by interpolation. HPV antibody control reagents were 201

included in each assay run (49) alongside Heparin (H-4784, Sigma-Aldrich) which was 202

used as a positive inhibitor control. The median neutralization titer and inter-quartile 203

range for the positive antibody control reagent (High HPV16/18) were as follows: HPV31 204

A PsV 231 (173 – 337; n=14), HPV31 B PsV 462 (387 – 671; n=10) and HPV31 C PsV 205

500 (354 – 589; n=12). The negative antibody control reagent (HPV negative) had a titer 206

of <40 in all assays (n=42).

207

(11)

208

Statistical analysis 209

The Wilcoxon paired signed rank test was used to compare neutralization titers using 210

Stata 12.1(Statacorp, College Station, TX).

211

(12)

Results 212

213

HPV31 L1 and L2 amino acid variation 214

Full length HPV31 L1 and L2 sequences were represented by contemporary English 215

(n=17) and Italian (n=3) sequences, in addition to available NCBI database sequences 216

(n=24). Analysis of the aligned, concatenated L1 and L2 sequences demonstrated three 217

distinct clusters consistent with the lineages HPV31 A, HPV31 B and HPV31 C (Figure 218

1A). Lineage A contained 14 sequences, including the HPV31 reference sequence 219

(J04353), 7 of which demonstrated variation from the reference in the L1 (T432S) and a 220

single sequence demonstrated variation within the L2 (I270M) (Figure 1B). The 13 221

sequences in lineage B all demonstrated variation from the reference sequence at L1 222

amino acid position T274N within the FG loop whilst 4 sequences also varied at position 223

S194T and 1 varied at position T432A. Five sequences exhibit variation from the 224

reference at L2 position T362A. Lineage C contained 17 sequences all of which 225

demonstrate variation from the reference at L1 amino acid position T267A and T274N 226

within the FG loop and L2 positions V115I, I270M, V377L, 5 sequences also varied from 227

the reference at L2 amino acid position N269D.

228 229

Sensitivity of variant HPV31 L1L2 PsV to antibody-mediated neutralization 230

L1L2 PsV representing lineage variants HPV31 A (Ref: J04353), HPV31 B and HPV31 231

C were generated from the consensus L1 and L2 sequences representing each lineage 232

and bear the major L1 (267 and 274) and L2 (115, 270 and 377) variant residues 233

(13)

(Figure 2A). All three lineage variant PsV, herein referred to as HPV31 A PsV, HPV31 234

B PsV and HPV31 C PsV, generated similar sized PsV particles of around 50nm and 235

produced comparable PI ratios of ca. 102 (Figure 2B). The PsV preparations also 236

contained L2 protein and luciferase reporter plasmid (data not shown).

237

238

The HPV31 variant PsV were tested against sera from girls who received either the 239

Cervarix® or Gardasil® (n=46) HPV vaccine (Table 1). Both HPV31 B and C PsV were 240

more sensitive to neutralization when compared to HPV31 A PsV. HPV31 B PsV 241

displayed a median 1.7-fold (inter-quartile range, IQR, 1.1 – 2.4) (Wilcoxon paired 242

signed rank test p <0.001) increased sensitivity to vaccine-induced cross-neutralizing 243

antibodies whilst HPV31 C PsV displayed a 1.4-fold (1.1 – 1.6) (p <0.001) increase in 244

sensitivity compared to HPV31 A PsV. The increased sensitivity of HPV31 B and C PsV 245

to cross-neutralizing antibodies was independent of the HPV vaccine received (Table 246

247 1).

248

All three HPV31 variant PsV were susceptible to neutralization by a small panel of 249

longitudinal (month 0, 6, 12 and 18) serum samples from women naturally infected with 250

HPV31 (Table 2). Median antibody neutralization titers were 576 (391 – 1,144), 839 251

(587 – 1,899) and 882 (337 – 1,895) against HPV31 A, B and C PsV, respectively.

252

253

Immunogenicity of variant HPV31 L1 VLP 254

HPV31 L1 VLP bearing the L1 variant residues T267A and T274N (Figure 2A), herein 255

referred to as HPV31 A, HPV31 B and HPV31 C VLP were expressed and used to 256

(14)

immunize BALB/c mice. All three preparations contained VLPs of various sizes, ranging 257

from ca. 20 nm up to 60 nm in diameter with VLPs of >40 nm in diameter constituting 258

30% of the HPV31 B VLP, 37% of the HPV31 A VLP and 57% of HPV31 C VLP 259

preparations (Figure 2C).

260 261

The three variant PsV demonstrated differential susceptibility to neutralization by HPV31 262

VLP mouse polyclonal serum (Figure 3). Both HPV31 B PsV (median log10

263

neutralization titer, 4.27; IQR, 3.75 – 4.55) (Wilcoxon paired signed rank test p = 0.008) 264

and HPV31 C PsV (4.18; 3.86 – 4.49) (p = 0.007) were more sensitive to neutralization 265

by HPV31 A VLP sera compared to the homologous HPV31 A PsV (4.09; 3.54 – 4.26).

266

HPV31 A PsV was less sensitive to neutralization by HPV31 B VLP sera (3.74; 3.53 – 267

4.07) (p = 0.031) whilst HPV31 C PsV demonstrated increased sensitivity (4.12; 3.96 – 268

4.53) (p = 0.028) compared to HPV31 B PsV (3.98; 3.69 – 4.22). Both HPV31 B and C 269

PsV had similar sensitivity to neutralization by HPV31 C VLP sera (B PsV: 4.42 [4.04 – 270

4.35]; C PsV: 4.34 [4.08 – 4.55]) whilst HPV31 A PsV demonstrated a reduced 271

sensitivity (4.01; 3.84 – 4.14) (p = 0.021). There were also differences in the magnitude 272

of the antibody response based upon reactivity against homologous PsV with HPV31 C 273

VLP (4.34; 4.08 – 4.55) being slightly more immunogenic than HPV31 A VLP (4.09; 3.54 274

– 4.26) (p = 0.018) and HPV31 B VLP (3.98; 3.69 – 4.22) (p = 0.006).

275

276

Antigenicity of variant HPV31 L1 VLP and L1L2 PsV 277

(15)

HPV31 L1 MAbs were previously (39) generated against immunogens representing the 278

HPV31 reference sequence (J04353). The majority were raised against L1L2 VLP apart 279

from 31.D24 and 31.A19 where L1 VLP immunogens were used. Generally all the type- 280

specific MAbs bound L1 VLP and L1L2 PsV representing the lineages variants A, B and 281

C at similar 50% binding concentrations (µg/mL) by ELISA; the exceptions to this were 282

31.F16 which demonstrated a higher binding concentration against the HPV31 B PsV in 283

comparison the HPV31 A PsV whilst both 31.H12 and 31.H17 bound L1 VLP 284

representing HPV31 lineage variants B and C at lower concentrations compared to A.

285

Although the cross-reactive MAbs (31.D24, 31.B5, 31.C19 and 31.E22) bound the L1 286

VLP they did not bind or neutralize the L1L2 PsV (Table 3). All type-specific MAbs were 287

able to neutralize the three variant PsV to similar orders of magnitude (Table 3). HPV31 288

C PsV demonstrated an increased susceptibility, ca. 3.5-fold, to neutralization by FG 289

loop MAb 31.F16 and a ca. 4.0-fold increase in sensitivity to MAb 31.H17 (epitope 290

unknown) compared to HPV31 A and B PsV (Table 3).

291 292

The three neutralizing FG loop MAbs (31.B1, 31.F16 and 31.H12) recognize 293

conformational epitopes which encompass variant amino acid position 267 and are 294

adjacent to amino acid 274 (Figure 4A). The FG loop of monomer 1 (FG1) is adjacent to 295

the BC5, DE1, DE5, EF1, HI4 andHI5 loops within the capsomer (Figure 4B) and residues 296

267 and 274 are within close proximity (within 10Å) to residue positions predominantly 297

within the adjacent BC5, FG1,and HI5 loops (Figure 4C and 4D). These include Lysine 298

residues at position 279 within the FG loop and 362 within the HI loop.

299

(16)

Discussion 300

This study attempted to evaluate the potential impact of non-synonymous SNP within 301

the HPV31 L1 and L2 genes on capsid protein antigenicity and immunogenicity. We 302

generated additional HPV31 L1 and L2 sequences to supplement those already 303

available and created L1L2 PsV and L1 VLP representing lineage variants A, B and C to 304

evaluate lineage-specific immunogenicity and antigenicity, including susceptibility to 305

antibody-mediated neutralization.

306

307

All three variant L1L2 PsV were susceptible to neutralization by vaccine-induced cross- 308

neutralizing antibodies. HPV31 B and C PsV demonstrated an increased sensitivity to 309

neutralization compared to HPV31 A PsV but the difference was of a low magnitude.

310

The cross-protection afforded by the current prophylactic vaccines is an unexpected 311

additional benefit, although no correlate of protection has been defined (10). If cross- 312

neutralizing antibodies are determined to be the immune effectors of vaccine-induced 313

cross-protection it is important to demonstrate that the contemporary circulating HPV31 314

variants, represented by L1L2 PsV, do not exhibit resistance to cross-neutralization by 315

such antibody specificities.

316 317

The increased sensitivity of HPV31 B and C PsV to cross-neutralization suggests that 318

the Asn residue at position 274, which is common to both variants, enhances the 319

recognition of HPV31 L1 epitopes by cross-neutralizing antibodies produced against 320

vaccine HPV16 L1 VLPs. It is unlikely that the Asn residue in itself has a critical role 321

(17)

within a cross-neutralizing epitope since the switch from Thr and Asn is a relative subtle 322

change as both amino acids have polar uncharged side chains. However, the change of 323

residue at position 274, near the tip of the FG loop, may result in local structural 324

changes which increase recognition of more distal epitope residues.

325 326

Subtle differences in variant antigenicity were identified when a panel of HPV31 MAbs 327

were tested in a neutralization assay against the HPV31 lineage variants. HPV31 C PsV 328

demonstrated increased sensitivity to neutralization by the FG loop MAb 31.F16 in 329

comparison to both HPV31 A and B PsV indicating that the double residue switch at 330

positions T267A and T274N impacts upon 31.F16 epitope recognition. It has previously 331

been demonstrated by comparison of L1 pentamer crystal structures from different 332

genotypes (HPV11, HPV16, HPV18 and HPV35) that L1 antigenic determinants can be 333

altered by a shift of a few angstroms within the loop as a result of a single residue 334

substitution (28).

335 336

In contrast the other two FG loop MAbs (31.B1 and 31.H12) neutralized all variants to a 337

similar extent. These data corroborate previous data from a bacterial cell surface display 338

model which demonstrated that these three MAbs recognise overlapping, yet distinct FG 339

loop epitopes (36). Residues 267 and 274 are in close proximity to (Lys279 & Lys362), 340

or near (Lys54, Asn57, Lys60 & Lys367) residues, the corresponding residues of 341

HPV16 are involved in HPV binding to heparin sulfate (50) which is an essential step for 342

a successful HPV infection and the FG loop MAbs may neutralize by abrogating this 343

(18)

345

The panel of HPV31 MAbs bound all three variant L1 VLP, when used as target 346

antigens in an ELISA format, indicating that the residues at positions 267 and 274 were 347

not critical in the epitope footprints recognized by this panel of MAbs. However, when 348

variant L1L2 PsV were used as target antigens, the 50% binding concentration of the 349

four cross-reactive MAbs (31.D24, 31.B5, 31.C19 and 31.E22) could not be determined 350

due to the reduction in epitope recognition. This observation implies that inclusion of the 351

L2 protein within the PsV capsid alters L1 epitope exposure and therefore impacts upon 352

L1 protein antigenicity. It has been reported that a subset of HPV16 MAbs demonstrated 353

reduced binding to L1L2 PsV compared to L1 VLP with the differential binding thought 354

to be as a result of L2 altering the conformation or availability of L1 epitopes (51). These 355

findings imply that the capsids of native HPV virions, represented by L1L2 PsV, and the 356

L1-only VLP within the prophylactic vaccine preparations differ in their L1 antigenicity.

357 358

All three variant L1L2 PsV were susceptible to neutralization by HPV31 VLP antibodies 359

generated in mice; however, differences in neutralization sensitivity were evident. Both 360

HPV31 B and C PsV, in comparison to HPV31 A PsV, demonstrated increased 361

sensitivity to antibody-mediated neutralization irrespective of the variant L1 VLP used as 362

the immunogen. These findings are in line with a previous study of HPV16 variants 363

which demonstrated that HPV16 VLP sera raised against a European variant was able 364

to neutralize pseudoviruses representing a range of geographical variants of HPV16 365

with ≤4-fold difference in neutralization titer between the homologous and heterologous 366

types, leading to the conclusion that HPV16 variants belong to a single serotype (29).

367

(19)

368

The criterion used to designate serotypes is generally based upon an fold difference in 369

antibody-mediated neutralization titers between viral types, which differ in magnitude 370

and range between virus families: Adenovirus 8- to 16-fold (52), Rotavirus ≥20-fold (53), 371

Polyomavirus 4- to 100-fold (54). For HPV there are no currently defined criteria with 372

which to designate L1 serotypes. It is reasonably clear that HPV genotypes induce high 373

titer, type-specific neutralizing antibody responses which represent different serotypes 374

(55-57). However, for lineage variants the relationship between L1 sequence and 375

antigenicity is less clear (29, 58).

376 377

Although HPV31 lineage variants demonstrated differences in susceptibility to antibody- 378

mediated neutralization by vaccine, pre-clinical and MAb panels the difference was <4- 379

fold and under this criterion, as defined for HPV16 (29), HPV31 variants should be 380

considered as belonging to a single serotype. This implies that the choice of a 381

representative HPV31 L1 sequence for VLP based vaccines is not critical.

382

383

Given the relatively low prevalence of HPV31 (59), only a small panel of serum samples 384

from HPV31 DNA positive women were available. These data suggest that all three 385

HPV31 lineage variants were susceptible to neutralization by antibodies derived from 386

natural infection. However, further work will be required to address this issue 387

appropriately by utilizing a larger panel of samples with an equal representation of 388

variant lineages.

389

(20)

There are potential shortcomings to this work. L1 VLP immunizations were carried out 391

using a relative small number of animals and while all three constructs induced 392

neutralizing antibodies against all three variant PsV, the variability inherent in using 393

small groups of animals may have concealed subtle differences in immunogenicity.

394

Although HPV L1L2 PsV have been used widely to monitor antibody responses to 395

vaccine and natural infection (22, 47, 48, 60), as well as elucidate steps in the entry 396

process (61-64), there are likely to be some differences between how these behave in 397

vitro and how authentic HPV31 lineage variants behave in vivo, although this is a 398

limitation of most PsV-based systems.

399 400

Despite these caveats, these data suggest that HPV31 lineage variant PsV display 401

similar sensitivity to recognition by antibodies elicited following vaccination with the 402

current HPV vaccines and after pre-clinical HPV31 L1 VLP immunization indicating that 403

HPV31 variants belong to a single L1 serotype. Such data may be useful to guide 404

impact modelling of the current L1 VLP vaccines and informing post-vaccine 405

surveillance programmes. These data also inform our understanding of the antigenicity 406

of the HPV structural proteins.

407

(21)

Acknowledgments 408

The sera from young girls who received Gardasil® or Cervarix® came from a study 409

funded by the Department of Health Policy Research Programme (National Vaccine 410

Evaluation Consortium, 039/0031). The views expressed in this publication are those of 411

the author(s) and not necessarily those of the Department of Health.

412 413

We are indebted to Prof. John T. Schiller and Dr. Chris Buck (National Cancer Institute, 414

Bethesda, U.S.A.) for access to the p31sheLL clone. We thank Ilaria Epifano from the 415

University of Milano-Bicocca, Monza, Italy for the natural infection HPV31 DNA and 416

antibody status data.

417 418

The authors of this paper declare no conflicts of interest.

419

(22)

References 420

421

1. Rector A, Lemey P, Tachezy R, Mostmans S, Ghim SJ, Van Doorslaer K, Roelke M, 422

Bush M, Montali RJ, Joslin J, Burk RD, Jenson AB, Sundberg JP, Shapiro B, Van 423

Ranst M. 2007. Ancient papillomavirus-host co-speciation in Felidae. Genome Biol 424

8:R57.

425

2. Duffy S, Shackelton LA, Holmes EC. 2008. Rates of evolutionary change in viruses:

426

patterns and determinants. Nat Rev Genet 9:267-276.

427

3. Burk RD, Harari A, Chen Z. 2013. Human papillomavirus genome variants. Virology 428 445:232-243.

429 4. Chen Z, Schiffman M, Herrero R, Desalle R, Anastos K, Segondy M, 430

Sahasrabuddhe VV, Gravitt PE, Hsing AW, Burk RD. 2011. Evolution and taxonomic 431

classification of human papillomavirus 16 (HPV16)-related variant genomes: HPV31, 432

HPV33, HPV35, HPV52, HPV58 and HPV67. PLoS One 6:e20183.

433

5. Buck CB, Cheng N, Thompson CD, Lowy DR, Steven AC, Schiller JT, Trus BL.

434

2008. Arrangement of L2 within the papillomavirus capsid. Journal of virology 82:5190-

435 5197.

436 6. Buck CB, Day PM, Trus BL. 2013. The papillomavirus major capsid protein L1. Virology 437

445:169-174.

438

7. Wang JW, Roden RB. 2013. L2, the minor capsid protein of papillomavirus. Virology 439

445:175-186.

440

8. Carter JJ, Koutsky LA, Wipf GC, Christensen ND, Lee SK, Kuypers J, Kiviat N, 441

Galloway DA. 1996. The natural history of human papillomavirus type 16 capsid 442

antibodies among a cohort of university women. The Journal of infectious diseases 443 174:927-936.

444 9. Carter JJ, Wipf GC, Madeleine MM, Schwartz SM, Koutsky LA, Galloway DA. 2006.

445

Identification of human papillomavirus type 16 L1 surface loops required for 446

neutralization by human sera. Journal of virology 80:4664-4672.

447

10. Stanley M, Pinto LA, Trimble C. 2012. Human papillomavirus vaccines--immune 448

responses. Vaccine 30 Suppl 5:F83-87.

449

11. Carter JJ, Koutsky LA, Hughes JP, Lee SK, Kuypers J, Kiviat N, Galloway DA.

450

2000. Comparison of human papillomavirus types 16, 18, and 6 capsid antibody 451 responses following incident infection. The Journal of infectious diseases 181:1911-

452 1919.

453

12. Safaeian M, Porras C, Schiffman M, Rodriguez AC, Wacholder S, Gonzalez P, Quint 454

W, van Doorn LJ, Sherman ME, Xhenseval V, Herrero R, Hildesheim A, Costa 455

Rican Vaccine Trial G. 2010. Epidemiological study of anti-HPV16/18 seropositivity and 456

subsequent risk of HPV16 and -18 infections. Journal of the National Cancer Institute 457

102:1653-1662.

458

13. Ho GY, Studentsov Y, Hall CB, Bierman R, Beardsley L, Lempa M, Burk RD. 2002.

459 Risk factors for subsequent cervicovaginal human papillomavirus (HPV) infection and 460 the protective role of antibodies to HPV-16 virus-like particles. The Journal of infectious 461

diseases 186:737-742.

462

14. Castellsague X, Naud P, Chow SN, Wheeler CM, Germar MJ, Lehtinen M, Paavonen 463

J, Jaisamrarn U, Garland SM, Salmeron J, Apter D, Kitchener H, Teixeira JC, 464

Skinner SR, Limson G, Szarewski A, Romanowski B, Aoki FY, Schwarz TF, Poppe 465

WA, Bosch FX, de Carvalho NS, Peters K, Tjalma WA, Safaeian M, Raillard A, 466

Descamps D, Struyf F, Dubin G, Rosillon D, Baril L. 2014. Risk of Newly Detected 467 Infections and Cervical Abnormalities in Women Seropositive for Naturally Acquired 468 Human Papillomavirus Type 16/18 Antibodies: Analysis of the Control Arm of PATRICIA.

469

The Journal of infectious diseases 210:517-534.

470

(23)

15. Wilson L, Pawlita M, Castle PE, Waterboer T, Sahasrabuddhe V, Gravitt PE, 471

Schiffman M, Wentzensen N. 2014. Seroprevalence of 8 oncogenic human 472

papillomavirus genotypes and acquired immunity against reinfection. The Journal of 473 infectious diseases 210:448-455.

474

16. Schiller JT, Castellsague X, Garland SM. 2012. A review of clinical trials of human 475

papillomavirus prophylactic vaccines. Vaccine 30 Suppl 5:F123-138.

476

17. Lehtinen M, Dillner J. 2013. Clinical trials of human papillomavirus vaccines and 477

beyond. Nat Rev Clin Oncol 10:400-410.

478

18. Malagon T, Drolet M, Boily MC, Franco EL, Jit M, Brisson J, Brisson M. 2012.

479

Cross-protective efficacy of two human papillomavirus vaccines: a systematic review and 480 meta-analysis. The Lancet. Infectious diseases 12:781-789.

481 19. Einstein MH, Baron M, Levin MJ, Chatterjee A, Fox B, Scholar S, Rosen J, 482

Chakhtoura N, Meric D, Dessy FJ, Datta SK, Descamps D, Dubin G. 2011.

483

Comparative immunogenicity and safety of human papillomavirus (HPV)-16/18 vaccine 484

and HPV-6/11/16/18 vaccine: follow-up from months 12-24 in a Phase III randomized 485

study of healthy women aged 18-45 years. Hum Vaccin 7:1343-1358.

486

20. Draper E, Bissett SL, Howell-Jones R, Waight P, Soldan K, Jit M, Andrews N, Miller 487

E, Beddows S. 2013. A randomized, observer-blinded immunogenicity trial of

488 Cervarix((R)) and Gardasil((R)) Human Papillomavirus vaccines in 12-15 year old girls.

489 PLoS One 8:e61825.

490

21. Kemp TJ, Hildesheim A, Safaeian M, Dauner JG, Pan Y, Porras C, Schiller JT, Lowy 491

DR, Herrero R, Pinto LA. 2011. HPV16/18 L1 VLP vaccine induces cross-neutralizing 492

antibodies that may mediate cross-protection. Vaccine 29:2011-2014.

493

22. Krajden M, Cook D, Yu A, Chow R, Mei W, McNeil S, Money D, Dionne M, 494

Karunakaran KP, Palefsky JM, Dobson S, Ogilvie G, Petric M. 2011. Human 495

papillomavirus 16 (HPV 16) and HPV 18 antibody responses measured by pseudovirus 496 neutralization and competitive Luminex assays in a two- versus three-dose HPV vaccine 497 trial. Clin Vaccine Immunol 18:418-423.

498

23. Safaeian M, Kemp TJ, Pan DY, Porras C, Rodriguez AC, Schiffman M, Cortes B, 499

Katki H, Wacholder S, Schiller JT, Gonzalez P, Penrose K, Lowy DR, Quint W, van 500

Doorn LJ, Herrero R, Hildesheim A, Pinto LA. 2013. Cross-protective vaccine efficacy 501

of the bivalent HPV vaccine against HPV31 is associated with humoral immune 502

responses: results from the Costa Rica Vaccine Trial. Hum Vaccin Immunother 9:1399- 503

1406.

504 24. Van Damme P, Leroux-Roels G, Simon P, Foidart JM, Donders G, Hoppenbrouwers 505 K, Levin M, Tibaldi F, Poncelet S, Moris P, Dessy F, Giannini SL, Descamps D, 506 Dubin G. 2014. Effects of varying antigens and adjuvant systems on the immunogenicity 507

and safety of investigational tetravalent human oncogenic papillomavirus vaccines:

508

results from two randomized trials. Vaccine 32:3694-3705.

509

25. Joura EA, Giuliano AR, Iversen OE, Bouchard C, Mao C, Mehlsen J, Moreira ED, 510

Jr., Ngan Y, Petersen LK, Lazcano-Ponce E, Pitisuttithum P, Restrepo JA, Stuart G, 511

Woelber L, Yang YC, Cuzick J, Garland SM, Huh W, Kjaer SK, Bautista OM, Chan 512 IS, Chen J, Gesser R, Moeller E, Ritter M, Vuocolo S, Luxembourg A, Broad 513 Spectrum HPVVS. 2015. A 9-valent HPV vaccine against infection and intraepithelial 514

neoplasia in women. The New England journal of medicine 372:711-723.

515

26. Ahmed AI, Bissett SL, Beddows S. 2013. Amino acid sequence diversity of the major 516

human papillomavirus capsid protein: implications for current and next generation 517

vaccines. Infection, genetics and evolution : journal of molecular epidemiology and 518

evolutionary genetics in infectious diseases 18:151-159.

519

(24)

27. Chen XS, Garcea RL, Goldberg I, Casini G, Harrison SC. 2000. Structure of small 520

virus-like particles assembled from the L1 protein of human papillomavirus 16. Mol Cell 521

5:557-567.

522 28. Bishop B, Dasgupta J, Klein M, Garcea RL, Christensen ND, Zhao R, Chen XS.

523

2007. Crystal structures of four types of human papillomavirus L1 capsid proteins:

524

understanding the specificity of neutralizing monoclonal antibodies. J Biol Chem 525

282:31803-31811.

526

29. Pastrana DV, Vass WC, Lowy DR, Schiller JT. 2001. NHPV16 VLP vaccine induces 527

human antibodies that neutralize divergent variants of HPV16. Virology 279:361-369.

528

30. White WI, Wilson SD, Palmer-Hill FJ, Woods RM, Ghim SJ, Hewitt LA, Goldman 529 DM, Burke SJ, Jenson AB, Koenig S, Suzich JA. 1999. Characterization of a major 530 neutralizing epitope on human papillomavirus type 16 L1. Journal of virology 73:4882- 531

4889.

532

31. Li N, Franceschi S, Howell-Jones R, Snijders PJ, Clifford GM. 2010. Human 533

papillomavirus type distribution in 30,848 invasive cervical cancers worldwide: Variation 534

by geographical region, histological type and year of publication. International journal of 535

cancer. Journal international du cancer 128:927-935.

536

32. Xi LF, Schiffman M, Koutsky LA, Hulbert A, Lee SK, Defilippis V, Shen Z, Kiviat NB.

537 2012. Association of human papillomavirus type 31 variants with risk of cervical

538 intraepithelial neoplasia grades 2-3. International journal of cancer. Journal international 539

du cancer 131:2300-2307.

540

33. Xi LF, Schiffman M, Koutsky LA, He Z, Winer RL, Hulbert A, Lee SK, Ke Y, Kiviat 541

NB. 2013. Persistence of newly detected human papillomavirus type 31 infection, 542

stratified by variant lineage. International journal of cancer. Journal international du 543

cancer 132:549-555.

544

34. Xi LF, Schiffman M, Koutsky LA, Hughes JP, Winer RL, Mao C, Hulbert A, Lee SK, 545 Shen Z, Kiviat NB. 2014. Lineages of oncogenic human papillomavirus types other than 546 type 16 and 18 and risk for cervical intraepithelial neoplasia. Journal of the National 547

Cancer Institute 106.

548

35. Schiffman M, Rodriguez AC, Chen Z, Wacholder S, Herrero R, Hildesheim A, 549

Desalle R, Befano B, Yu K, Safaeian M, Sherman ME, Morales J, Guillen D, Alfaro 550

M, Hutchinson M, Solomon D, Castle PE, Burk RD. 2010. A population-based 551

prospective study of carcinogenic human papillomavirus variant lineages, viral 552

persistence, and cervical neoplasia. Cancer research 70:3159-3169.

553 36. Fleury MJ, Touze A, Maurel MC, Moreau T, Coursaget P. 2009. Identification of 554 neutralizing conformational epitopes on the human papillomavirus type 31 major capsid 555

protein and functional implications. Protein Sci 18:1425-1438.

556

37. Carpentier GS, Fleury MJ, Touze A, Sadeyen JR, Tourne S, Sizaret PY, Coursaget 557

P. 2005. Mutations on the FG surface loop of human papillomavirus type 16 major 558

capsid protein affect recognition by both type-specific neutralizing antibodies and cross- 559

reactive antibodies. J Med Virol 77:558-565.

560

38. Howell-Jones R, de Silva N, Akpan M, Oakeshott P, Carder C, Coupland L, Sillis M, 561 Mallinson H, Ellis V, Frodsham D, Robinson TI, Gill ON, Beddows S, Soldan K.

562 2012. Prevalence of human papillomavirus (HPV) infections in sexually active 563

adolescents and young women in England, prior to widespread HPV immunisation.

564

Vaccine 30:3867-3875.

565

39. Fleury MJ, Touze A, Alvarez E, Carpentier G, Clavel C, Vautherot JF, Coursaget P.

566

2006. Identification of type-specific and cross-reactive neutralizing conformational 567

epitopes on the major capsid protein of human papillomavirus type 31. Arch Virol 568

151:1511-1523.

569

(25)

40. Cornut G, Gagnon S, Hankins C, Money D, Pourreaux K, Franco EL, Coutlee F, 570

Canadian Women's HIVSG. 2010. Polymorphism of the capsid L1 gene of human 571

papillomavirus types 31, 33, and 35. J Med Virol 82:1168-1178.

572 41. Icenogle JP, Clancy KA, Lin SY. 1995. Sequence variation in the capsid protein genes 573

of human papillomavirus type 16 and type 31. Virology 214:664-669.

574

42. Goldsborough MD, DiSilvestre D, Temple GF, Lorincz AT. 1989. Nucleotide 575

sequence of human papillomavirus type 31: a cervical neoplasia-associated virus.

576

Virology 171:306-311.

577

43. Tamura K, Stecher G, Peterson D, Filipski A, Kumar S. 2013. MEGA6: Molecular 578

Evolutionary Genetics Analysis version 6.0. Mol Biol Evol 30:2725-2729.

579 44. Guex N, Peitsch MC. 1997. SWISS-MODEL and the Swiss-PdbViewer: an environment 580 for comparative protein modeling. Electrophoresis 18:2714-2723.

581

45. Huo Z, Bissett SL, Giemza R, Beddows S, Oeser C, Lewis DJ. 2012. Systemic and 582

mucosal immune responses to sublingual or intramuscular Human Papilloma Virus 583

antigens in healthy female volunteers. PLoS One 7:e33736.

584

46. Buck CB, Thompson CD. 2007. Production of papillomavirus-based gene transfer 585

vectors. Curr Protoc Cell Biol Chapter 26:Unit 26 21.

586

47. Draper E, Bissett SL, Howell-Jones R, Edwards D, Munslow G, Soldan K, Beddows 587 S. 2011. Neutralization of non-vaccine human papillomavirus pseudoviruses from the A7 588 and A9 species groups by bivalent HPV vaccine sera. Vaccine 29:8585-8590.

589

48. Pastrana DV, Buck CB, Pang YY, Thompson CD, Castle PE, FitzGerald PC, Kruger 590

Kjaer S, Lowy DR, Schiller JT. 2004. Reactivity of human sera in a sensitive, high- 591

throughput pseudovirus-based papillomavirus neutralization assay for HPV16 and 592

HPV18. Virology 321:205-216.

593

49. Bissett SL, Wilkinson D, Tettmar KI, Jones N, Stanford E, Panicker G, Faust H, 594

Borrow R, Soldan K, Unger ER, Dillner J, Minor P, Beddows S. 2011. Human 595 papillomavirus antibody reference reagents for use in postvaccination surveillance 596 serology. Clin Vaccine Immunol 19:449-451.

597

50. Richards KF, Bienkowska-Haba M, Dasgupta J, Chen XS, Sapp M. 2013. Multiple 598

heparan sulfate binding site engagements are required for the infectious entry of human 599

papillomavirus type 16. Journal of virology 87:11426-11437.

600

51. Culp TD, Spatz CM, Reed CA, Christensen ND. 2007. Binding and neutralization 601

efficiencies of monoclonal antibodies, Fab fragments, and scFv specific for L1 epitopes 602

on the capsid of infectious HPV particles. Virology 361:435-446.

603 52. Heemskerk B, Lankester AC, van Vreeswijk T, Beersma MF, Claas EC, Veltrop- 604 Duits LA, Kroes AC, Vossen JM, Schilham MW, van Tol MJ. 2005. Immune 605

reconstitution and clearance of human adenovirus viremia in pediatric stem-cell 606

recipients. The Journal of infectious diseases 191:520-530.

607

53. Wyatt RG, Greenberg HB, James WD, Pittman AL, Kalica AR, Flores J, Chanock 608

RM, Kapikian AZ. 1982. Definition of human rotavirus serotypes by plaque reduction 609

assay. Infection and immunity 37:110-115.

610

54. Pastrana DV, Ray U, Magaldi TG, Schowalter RM, Cuburu N, Buck CB. 2013. BK 611 polyomavirus genotypes represent distinct serotypes with distinct entry tropism. Journal 612 of virology 87:10105-10113.

613

55. Giroglou T, Sapp M, Lane C, Fligge C, Christensen ND, Streeck RE, Rose RC. 2001.

614

Immunological analyses of human papillomavirus capsids. Vaccine 19:1783-1793.

615

56. Combita AL, Touze A, Bousarghin L, Christensen ND, Coursaget P. 2002.

616

Identification of two cross-neutralizing linear epitopes within the L1 major capsid protein 617

of human papillomaviruses. Journal of virology 76:6480-6486.

618

(26)

57. Bissett SL, Mattiuzzo G, Draper E, Godi A, Wilkinson DE, Minor P, Page M, 619

Beddows S. 2014. Pre-clinical immunogenicity of human papillomavirus alpha-7 and 620

alpha-9 major capsid proteins. Vaccine 32:6548-6555.

621 58. Cheng G, Icenogle JP, Kirnbauer R, Hubbert NL, St Louis ME, Han C, Svare EI, 622

Kjaer SK, Lowy DR, Schiller JT. 1995. Divergent human papillomavirus type 16 623

variants are serologically cross-reactive. The Journal of infectious diseases 172:1584- 624

1587.

625

59. Bruni L, Diaz M, Castellsague X, Ferrer E, Bosch FX, de Sanjose S. 2010. Cervical 626

human papillomavirus prevalence in 5 continents: meta-analysis of 1 million women with 627

normal cytological findings. The Journal of infectious diseases 202:1789-1799.

628 60. Einstein MH, Baron M, Levin MJ, Chatterjee A, Edwards RP, Zepp F, Carletti I, 629 Dessy FJ, Trofa AF, Schuind A, Dubin G, Group HPVS. 2009. Comparison of the 630

immunogenicity and safety of Cervarix and Gardasil human papillomavirus (HPV) 631

cervical cancer vaccines in healthy women aged 18-45 years. Hum Vaccin 5:705-719.

632

61. Bousarghin L, Touze A, Sizaret PY, Coursaget P. 2003. Human papillomavirus types 633

16, 31, and 58 use different endocytosis pathways to enter cells. Journal of virology 634

77:3846-3850.

635

62. Day PM, Baker CC, Lowy DR, Schiller JT. 2004. Establishment of papillomavirus 636 infection is enhanced by promyelocytic leukemia protein (PML) expression. Proc Natl 637 Acad Sci U S A 101:14252-14257.

638

63. Day PM, Lowy DR, Schiller JT. 2008. Heparan sulfate-independent cell binding and 639

infection with furin-precleaved papillomavirus capsids. Journal of virology 82:12565- 640

12568.

641

64. Bienkowska-Haba M, Williams C, Kim SM, Garcea RL, Sapp M. 2012. Cyclophilins 642

facilitate dissociation of the human papillomavirus type 16 capsid protein L1 from the 643

L2/DNA complex following virus entry. Journal of virology 86:9875-9887.

644 645

(27)

Figure 1. HPV31 L1 and L2 variation. (A) Phylogenetic tree constructed from 646

concatenated L1 and L2 nucleotide sequences, labelled with designated lineages (A, B 647

& C) (51) and bootstrap values >95%. (B) Site-specific amino acid co-variation within 648

the L1 and L2 proteins. N indicates the number of sequences in the phylogentic tree 649

represented by the each L1 and L2 combination.

650 651

Figure 2. HPV31 L1 and L2 variants. (A) Graphical representation of L1 and L2 variant 652

protein combinations. (B) L1L2 pseudovirus preparation characterized for particle 653

dimension in nm (median, inter-quartile range [IQR]), infectivity and L1 concentration.

654

(C) L1 VLP dimensions in nm (median, IQR for particles >40 nm in diameter) and L1 655

concentration.

656

657

Figure 3. Heatmaps representing the neutralization potential of variant HPV31 L1 VLP 658

immunization serum against variant HPV31 L1L2 PsV. Log10 neutralization titers of sera 659

from BALB/c mice (n = 10) following variant HPV31 VLP immunization carried out over 660

three separate schedules and presented as the average of two data sets per sample.

661

Key indicates log10 heatmap gradient. p values represent difference in median 662

neutralization titers from homologous variant VLP and PsV pair: *p < 0.05; **p < 0.01;

663

***p < 0.001; NSp > 0.05 using the Wilcoxon paired signed rank test.

664

665

Figure 4. Crystal model surface highlighting HPV31 FG loop variant residue locations.

666

(A) Linear amino acid epitope footprint of HPV31 FG loop MAbs. (B) Side view 667

highlighting loops in close proximity to FG loop variant residues 267 and 274 (C and D).

668

(28)

Top view of loop ribbons, circled areas indicate regions within a 10Å radius of residues 669

267 (C) and 274 (D) as determined by Swiss-PDP viewer algorithm. Lysine residues at 670

positons 279 and 362 are highlighted in blue. The FG loop of monomer 1 (FG1) is 671

coloured orange and residues 267 and 274 are highlighted in black. Neighbouring loops 672

on the same (DE1 - dark pink; EF1 - red) or adjacent monomers (HI4 -dark greeen; BC5 - 673

yellow; DE5 - light pink; HI5 - light green) are indicated. The remaining surface exposed 674

regions of the capsomer are coloured in light grey and core regions are coloured in dark 675

grey.

676

(29)
(30)
(31)
(32)

Références

Documents relatifs

Human Papillomavirus Type 31 E5 Protein Supports Cell Cycle Progression and Activates Late Viral Functions upon Epithelial Differentiation.. Interactions between Papillomavirus L1

Immunoblot experiments with a dysferlin antibody on proteins extracted from the patient ’ s muscle identified a ~73-kD band, supporting the use of the cryptic downstream

Generally, all the type- specific MAbs bound L1 VLPs and L1L2 PsVs representing the lineage variants A, B, and C at similar 50% binding concentrations (in micrograms per milliliter)

Pre-vaccination Prevalence and Genotype Distribution of Human Papillomavirus Infection among Women from Urban Tunis: a Cross-sectional Study.... Prevalence and Genotype Distribution

Thus, some model compounds were the same as those used in the charge optimization, and some were fragments from modified bases, whose charges were either well predicted by the

* Ce taux de recours représente le nombre de personnes en recherche d’emploi dont le dossier de formation a été accepté (le nombre d’heures acquises pouvant être nul), rapporté

D’autres facteurs peuvent induire le choix du certificateur et des diplômes : les modalités d’organisation des épreuves jugées plus adaptées aux candidats concernés (mises

enllT ces pôles commerciaux de la Méditerranée occidentale.. - La rivalité avec les Perses va jouer le même rôle: expéditions et initiatives diplomatiques] pouvaicnt