2 types 7 years following immunization with either cervarix® or … · 2020. 3. 27. · 1 1...

21
1 Durability of the Neutralizing Antibody Response to Vaccine and Non-Vaccine HPV 1 Types 7 Years Following Immunization with either Cervarix® or Gardasil® Vaccine 2 3 Anna Godi, 1 Kavita Panwar, 1 Mahmoud Haque, 1 Clementina E. Cocuzza, 3 Nick Andrews, 4 Jo 4 Southern, 2 Paul Turner, 5 Elizabeth Miller, 2 Simon Beddows, 1 5 6 7 1 Virus Reference Department, Public Health England, London, UK. 8 2 Immunisation and Countermeasures Public Health England, London, UK. 9 3 Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy 10 4 Statistics, Modelling and Economics Department, Public Health England, London, UK. 11 5 Section of Paediatrics, Imperial College London, London, UK. 12 13 * Corresponding author: Dr. Simon Beddows, Virus Reference Department, Public Health England, 61 14 Colindale Avenue, London NW9 5EQ, UK. T: +44 (0) 20 8327 6169. E: [email protected] 15 16 Submission to: Vaccine 17 18 19 20 Running title: HPV vaccine antibody durability 21 22 23 *Manuscript (Unmarked) Click here to view linked References

Upload: others

Post on 19-Aug-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

1

Durability of the Neutralizing Antibody Response to Vaccine and Non-Vaccine HPV 1

Types 7 Years Following Immunization with either Cervarix® or Gardasil® Vaccine 2

3

Anna Godi,1 Kavita Panwar,1 Mahmoud Haque,1 Clementina E. Cocuzza,3 Nick Andrews,4 Jo 4

Southern,2 Paul Turner,5 Elizabeth Miller,2 Simon Beddows,1 5

6

7

1 Virus Reference Department, Public Health England, London, UK. 8

2 Immunisation and Countermeasures Public Health England, London, UK. 9

3 Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy 10

4 Statistics, Modelling and Economics Department, Public Health England, London, UK. 11

5 Section of Paediatrics, Imperial College London, London, UK. 12

13

* Corresponding author: Dr. Simon Beddows, Virus Reference Department, Public Health England, 61 14

Colindale Avenue, London NW9 5EQ, UK. T: +44 (0) 20 8327 6169. E: [email protected] 15

16

Submission to: Vaccine 17

18

19

20

Running title: HPV vaccine antibody durability 21

22

23

*Manuscript (Unmarked)Click here to view linked References

2

Abstract 24

Bivalent (Cervarix®) and quadrivalent (Gardasil®) Human Papillomavirus (HPV) vaccines 25

demonstrate remarkable efficacy against the targeted genotypes, HPV16 and HPV18, but also a 26

degree of cross-protection against non-vaccine incorporated genotypes, HPV31 and HPV45. These 27

outcomes seem to be supported by observations that the HPV vaccines induce high titer 28

neutralizing antibodies against vaccine types and lower responses against non-vaccine types. Few 29

data are available on the robustness of the immune response against non-vaccine types. We 30

examined the durability of vaccine and non-vaccine antibody responses in a follow up of a head-to-31

head study of 12-15 year old girls initially randomized to receive three doses of Cervarix® or 32

Gardasil® vaccine. Neutralizing antibodies against both vaccine and non-vaccine types remained 33

detectable up to 7 years following initial vaccination and a mixed effects model was used to predict 34

the decline in antibody titers over a 15 year period. The decline in vaccine and non-vaccine type 35

neutralizing antibody titers over the study period was estimated to be 30% every 5-7 years, with 36

Cervarix® antibody titers expected to remain 3 – 4 fold higher than Gardasil® antibody titers over 37

the long term. The antibody decline rates in those with an initial response to non-vaccine types were 38

similar to that of vaccine types and are predicted to remain detectable for many years. Empirical 39

data on the breadth, magnitude, specificity and durability of the immune response elicited by the 40

HPV vaccines contribute to improving the evidence base supporting this important public health 41

intervention. Original trial: ClinicalTrials.gov NCT00956553 42

43

Keywords 44

Human papillomavirus 45

Vaccine 46

Antibody 47

Neutralization 48

Durability 49

3

Introduction 50

The bivalent (Cervarix®) and quadrivalent (Gardasil®) Human Papillomavirus (HPV) vaccines target 51

the most prevalent oncogenic genotypes (HPV16 and HPV18), while the next generation 52

nonavalent HPV vaccine (Gardasil®9) targets an additional 5 oncogenic genotypes (HPV31, 53

HPV33, HPV45, HPV52, and HPV58) [1]. Both quadrivalent and nonavalent vaccines target HPV6 54

and HPV11, which are associated with the development of genital warts. Cervarix® and Gardasil® 55

vaccines demonstrate remarkable efficacy against vaccine types (HPV16 and HPV18) and some 56

degree of efficacy against related non-vaccine types (cross-protection) in clinical trials [2, 3]. 57

Vaccine effectiveness studies are beginning to confirm these experimental observations in target 58

populations following introduction of national immunization programmes [4, 5]. 59

60

Neutralizing antibodies against vaccine genotypes can be detected in the serum and genital 61

secretions of vaccinees and passive transfer of neutralizing antibodies can protect animals against 62

papillomavirus challenge, leading to the reasonable assumption that type-specific protection is 63

mediated by neutralizing antibodies [3]. The degree of cross-protection afforded by the HPV 64

vaccines appears to be consistent with the detection of cross-neutralizing antibodies [6] suggesting 65

that such antibodies may be effectors or their detection may be useful as a correlate or surrogate of 66

vaccine-induced cross-protection. 67

68

Empirical data on the breadth, magnitude, specificity and durability of the immune response elicited 69

by the HPV vaccines contribute to improving the evidence base supporting this important public 70

health intervention. We have previously demonstrated that HPV vaccines induce high titer 71

neutralizing antibodies against vaccine types, with lower responses against non-vaccine types, and 72

that Cervarix® responses are typically higher and broader than Gardasil® responses [7]. In this 73

study we examined the durability of vaccine and non-vaccine antibody responses by resampling 12-74

15 year old girls immunized with three doses of Cervarix® or Gardasil® 7 years post immunization. 75

76

Methods 77

Ethics statement 78

This follow up study was approved by the NHS Health Research Authority and the London - 79

Hampstead Research Ethics Committee (reference 16/LO/1108). The original study protocol was 80

approved by the UK Medicines and Healthcare products Regulation Agency (MHRA) and registered 81

on the ClinicalTrials.gov website (NCT00956553) [7]. 82

83

Study population 84

The original study protocol and recruitment criteria have been published [7]. Briefly, 12-15 year old 85

girls recruited from two sites in England were randomized to receive three doses of either Cervarix® 86

4

or Gardasil® vaccine over a six month period. Serum samples were collected at month (M) 0 (prior 87

to vaccination), M2 (one month post second dose), M7 (one month post third dose) and M12 (six 88

months post third dose). Participants from the original study were invited to enrol for this follow up 89

study. For simplicity, the follow up study sample was designated as M84 (84 months following first 90

immunization) (Figure 1). Following informed consent a single blood sample was collected and 91

shipped to the testing laboratory (Public Health England, London) for serum separation and 92

subsequent storage at -80°C. In the original study, lower genital swab samples were collected at M7 93

for the detection of vaccine and non-vaccine type antibodies. Vaccine-type responses were 94

detected in around 100% of samples but non-vaccine type responses in only 4-20% of samples. 95

Given the expected decline in antibody titers between the original study and the follow up study and 96

the expected low enrolment rate, we decided it was unrealistic to expect to be able to detect non-97

vaccine type antibodies in a sufficient number of genital samples within the present study to conduct 98

any meaningful analysis. 99

100

Laboratory methods 101

Serum samples (M84) were assessed for the presence of neutralizing and binding antibodies in 102

parallel with the archived individual M12 serum sample from the original study [7]. This paired 103

testing was carried out in order to evaluate and potentially adjust for any differences between the 104

original and repeat M12 data. The pseudovirus (PsV) neutralization assay was carried out as 105

previously described [7] using PsV representing both vaccine (HPV16. HPV18) and non-vaccine 106

(HPV31, HPV45) genotypes. Inter-assay reproducibility was demonstrated by including the High 107

HPV16/18 antibody plasma pool and HPV Negative plasma pool as internal quality controls in 108

every experiment [8]. The median (interquartile range, IQR) titers of the High HPV16/18 plasma pool 109

were as follows: HPV16 (44,351; 32,636 - 58,979; n=12); HPV18 (18,051; 15,345 - 25,435), HPV31 110

(320; 253 - 369) and HPV45 (42; 39 - 61). The HPV Negative plasma pool was negative (<40) in all 111

runs. 112

113

Binding antibodies were evaluated in a virus-like particle (VLP) ELISA as previously described [7], 114

except that non-reporter containing L1L2 PsV were used as the target antigens. The median (IQR) 115

titers of the High HPV16/18 plasma pool were as follows: HPV16 (24,798; 24,508 - 27,348), HPV18 116

(4,832; 4,690 - 5,473), HPV31 (634; 519 - 836) and HPV45 (170; 162 - 176). The HPV Negative 117

plasma pool was negative (<50) in all runs. 118

119

Serum samples were obtained from women (Gynaecology Outpatients Clinic, San Gerardo Hospital, 120

Monza, Italy; ethics committee reference 08/UNIMIB-HPA/HPV1; No. 1191) following a cytological 121

diagnosis of atypical squamous cells of undetermined significance or low-grade squamous 122

intraepithelial lesion [9]. Samples from this cohort of women were used to provide an estimate of the 123

5

typical type-specific antibody titers generated during natural infection. Samples were selected from 124

women who were HPV DNA negative but HPV seropositive to the genotype in question at the time 125

of sampling and therefore represent women with a cleared infection, though no inference is made 126

about the role of antibodies in infection clearance. 127

128

International Standards (IS) for HPV serology are limited to HPV16 (IS16; 05-134; National Institute 129

for Biological Standards and Control, UK) and HPV18 (IS18; 10/140). No IS for HPV31 and HPV45 130

exist. Although serum neutralization data are usually reported in titers we made use of the IS16 and 131

IS18 to calibrate the High HPV16/18 plasma pool [8] allowing us to also report relative antibody 132

levels (IU/mL) against these two types for study samples. HPV vaccine and natural infection 133

antibody titers against HPV16 or HPV18 were converted to IU/mL by reference to the titer of the 134

calibrated High HPV16/18 plasma pool carried out in the same run. Reporting study data in IU/mL is 135

made to facilitate easier comparisons between studies where such data are also reported in 136

standardized units. 137

138

Statistical analysis 139

The sample size was limited by the response rate. With an estimated return of 27 per arm (30% 140

return rate) and assuming a standard deviation of 0.68 Log10 scale for antibody responses (based 141

on the original M12 data for HPV16, HPV18 and HPV31) and of 0.42 fold changes in geometric 142

mean titers (GMT; based on M7 to M12 titer decline) the precision (95% CI width) was estimated to 143

be within 1.9 fold for GMT at follow up and within 1.5 fold for fold changes from M12 to M84. 144

Differences of about 3.4 fold between vaccines would be detectable with 80% power at 5% 145

significance. Empirical outcomes were HPV16, HPV18, HPV31 and HPV45 antibody titers from the 146

PsV neutralization assay (cut-off 40) or VLP ELISA (cut-off 50). Values below the indicated assay 147

cut-off were given a censored value of half this limit. The proportion of seropositive samples as well 148

as GMT and fold changes between time points (with 95% confidence intervals) were estimated for 149

each vaccine and non-vaccine type. Proportions were compared by Fisher’s exact test. Comparison 150

between study arms was made by the Kruskal-Wallis test. Significance was taken at the 5% level 151

and 95% confidence intervals used. Two-sided significance tests were used. The relationship 152

between time since vaccination and antibody titers was modelled using all post vaccination data 153

(M7, M12 and M84) using a log-time on log-titre mixed effects model with random slope and 154

intercept. For HPV31 and HPV45 this was done within the subset with a response at M7. These 155

models were used to compare decline between vaccines and to extrapolate long term predicted 156

titres to 15 years post vaccination. Stata version 15 (StataCorp, USA) was used for the analyses. 157

158

Results 159

6

Subject characteristics 160

A similar number of Cervarix® (28/96; 29%) and Gardasil® (30/102; 30%) vaccinees took part in the 161

follow up study (Figure 1). Serum samples were collected a median 7.0 (range 6.7 – 7.6) years 162

after first immunization. The median age of the vaccinees at initial study entry was 12.9 (12.0 – 163

15.9) years and 19.7 (18.2 – 23.6) years old for the follow up (M84) sample. The median age of 164

participants at initial study entry for those who did not enrol in the follow up study was similar at 12.6 165

years (p>0.05). The median HPV16, HPV18, HPV31 and HPV45 serum neutralizing antibody titers 166

at M7 for those enrolled in the follow up study were similar to those that did not enrol (p>0.05; 167

Kruskal-Wallis test). These data suggest that the subset of individuals enrolled in the follow up study 168

is representative of the study participants overall. 169

170

Serum neutralization titers 171

M12 samples (n=58) were retested against all four PsV targets and compared to the original study 172

data for this time point. When combining data across all PsV targets there was a high correlation 173

between original and retest results (Pearson’s r = 0.98), but there was evidence of a deviation from 174

the line of equivalence at the high end of the titre range with a slope of the regression line (95% CI) 175

of retest vs. original = 0.894 (0.881 – 0.918). The fitted regression line was therefore used to 176

transform M2 and M7 data for HPV16 and HPV18 to allow appropriate comparisons to be made 177

across the whole study period for the subset of vaccinees taking part in the follow up study. 178

179

Seropositivity at M84 was 100% (95%CI 88 – 100%) against HPV16 for both vaccine arms and 180

against HPV18 for Cervarix® vaccinees (Table 1). HPV18 seropositivity for Gardasil® vaccinees 181

was 97% (83 – 100%). HPV31 seropositivity at M84 was 75% (55 – 89%) for Cervarix® vaccinees 182

compared to 50% (31 – 69%) for Gardasil® vaccinees. HPV45 seropositivity at M84 was 25% (11 – 183

45%) for Cervarix® vaccinees compared to 3% (0 – 17%) for Gardasil® vaccinees. 184

185

GMTs for Cervarix® vaccinees were typically higher than Gardasil® vaccinees throughout the 186

study. For example, the GMT (95%CI) of HPV16 neutralizing antibodies at M84 was 11,661 (8,272 - 187

16,439) and 2,898 (1,854 – 4,530) for Cervarix® and Gardasil® vaccinees, respectively. HPV16 and 188

HPV18 antibody titers were maintained above the GMT typical of natural infection up to 7 years 189

following initial immunization (Figure 2). The overall fold change in GMT (95%CI) between M12 and 190

M84 combined across both vaccines was 0.30 (0.25 – 0.36) for HPV16 and 0.28 (0.24 – 0.34) for 191

HPV18. 192

193

7

The High HPV16/18 plasma pool control [8] was calibrated to the International Standards for HPV16 194

and HPV18 antibodies allowing the HPV16 and HPV18 titers to be reported in IU/mL. The geometric 195

mean concentration (GMC; 95%CI) of neutralizing antibodies against HPV16 declined from 2,433 196

(1,755 – 3,372) IU/mL to 884 (664 – 1,177) IU/mL between M12 and M84 for Cervarix® vaccinees 197

(n=28) and from 839 (551 – 1,277) IU/mL to 206 (128 – 331) IU/mL between M12 and M84 for 198

Gardasil® vaccinees (n=30). By comparison, the GMC of neutralizing antibodies following natural 199

infection in women who were HPV16 antibody positive but HPV16 DNA negative at the time of 200

sampling was 14 (9 – 22; n=29) IU/mL. The GMC of neutralizing antibodies against HPV18 declined 201

from 753 (471 – 1,206) IU/mL to 230 (152 – 347) IU/mL between M12 and M84 for Cervarix® 202

vaccinees (n=28) and from 214 (133 – 346) IU/mL to 57 (35 – 94) IU/mL between M12 and M84 for 203

Gardasil® vaccinees (n=30). By comparison, the GMC of neutralizing antibodies following natural 204

infection in women who were HPV18 antibody positive but HPV18 DNA negative at the time of 205

sampling was 12 (7 – 20; n=17) IU/mL. 206

207

Non-vaccine type titers were substantially lower than vaccine type titers (Table 1). For example, the 208

GMT (95%CI) for HPV31 neutralizing antibodies at M84 was 110 (65 – 185) and 56 (37 – 85) for 209

Cervarix® and Gardasil® vaccinees, respectively. The overall fold change in GMT (95%CI) between 210

M12 and M84 was 0.56 (0.46 – 0.69) for HPV31 and 0.91 (0.80 – 1.03) for HPV45. Both HPV31 and 211

HPV45 antibody responses were lower than the GMT typical of natural infection (Figure 2). There 212

are no International Standards for HPV31 or HPV45 antibodies so antibody levels could not be 213

formally benchmarked. 214

215

A VLP ELISA was employed to corroborate the seropositivity and antibody decline rates seen in the 216

neutralization assay. Seropositivity at M84 was 100% (95%CI 88 – 100%) against HPV16 for both 217

vaccine arms and against HPV18 for Cervarix® vaccinees. HPV18 seropositivity for Gardasil® 218

vaccinees was 90% (73 – 98%). HPV31 seropositivity at M84 was 75% (55 – 89%) for Cervarix® 219

vaccinees compared to 53% (34 – 72%) for Gardasil® vaccinees. HPV45 seropositivity at M84 was 220

25% (11 – 45%) for Cervarix® vaccinees compared to 3% (0 – 17%) for Gardasil® vaccinees. The 221

overall fold change in GMT (95%CI) between M12 and M84 was 0.32 (0.27 – 0.39) for HPV16, 0.28 222

(0.24 – 0.33) for HPV18, 0.37 (0.31 – 0.45) for HPV31 and 0.66 (0.53 – 0.82) for HPV45. 223

Seropositivity at M84 and the fold change in binding antibodies between M12 and M84 were 224

generally similar to those derived from the pseudovirus neutralization assay. 225

226

Model prediction 227

8

The decline models for each vaccine for HPV16, HPV18, HPV31 and HPV45 showed an 228

approximate 20-35% decline per doubling of time (Figure 3). The rate of decline for HPV16 titers 229

was 0.74 (95%CI 0.71 – 0.78) fold per doubling of time compared to 0.66 (0.63 – 0.70) and 0.79 230

(0.75 – 0.83) for HPV18 and HPV31, respectively. The rate of decline for HPV45 in the Cervarix® 231

arm was 0.82 (0.76 – 0.89). The decline rates were similar for both vaccines (p>0.1) but Cervarix® 232

antibody titers were predicted to remain 3 – 4 fold higher than Gardasil® over the long term. 233

Extrapolating these declines out to 15 years following the third vaccine dose gave estimated 234

geometric mean HPV16 titers of 7,080 (4,820 – 10,380) for Cervarix® and 2,260 (1,560 – 3,280) for 235

Gardasil®. For HPV18 the titers at 15 years post vaccination are estimated as 1,820 (1,180 – 2,820) 236

for Cervarix® and 460 (300 – 700) for Gardasil®. For HPV31 within those who initially responded to 237

vaccination, Cervarix® and Gardasil® titers are predicted to decline to an average of 110 (65 – 187) 238

and 67 (38 – 118) respectively. For HPV 45, Cervarix® titers are predicted to decline to 25 (14 – 45) 239

over the long term. 240

241

9

Discussion 242

This study assessed the durability of the functional antibody response against vaccine and non-243

vaccine HPV genotypes elicited by the Cervarix® and Gardasil® HPV vaccines when administered 244

to 12-15 year old girls in a three dose regimen. Although there was a decline over the examination 245

period, neutralizing antibodies against both vaccine and non-vaccine types remained detectable up 246

to 7 years following initial vaccination. 247

248

All participants remained seropositive to HPV16 following immunization with either vaccine and 249

against HPV18 following immunization with Cervarix®, but seropositivity to HPV18 in the recipients 250

who received the Gardasil® vaccine was lower than 100% after 7 years of follow up. These data are 251

consistent with antibody binding data from 8-9 years of follow up of adolescents [10] or 16 – 23 year 252

old women [11] immunized with three doses of Gardasil®, a 10 year follow up study of 15 – 25 year 253

old women immunized with three doses of Cervarix® [12] and long term follow up (ca. 12 year) of 254

women enrolled in the Finnish cohorts of the Phase 3 licensure trials of both Cervarix® and 255

Gardasil® vaccines [13]. A lower vaccine-type seropositivity rate for the neutralizing antibody 256

response in Gardasil® vaccinees was also reported at 5 years in a head to head study of both 257

vaccines in 18 – 26 year old women [14]. Lower HPV18 seropositivity rates in Gardasil® vaccinees 258

were evident in both the neutralizing and binding antibody responses reported here but these were 259

higher than the rates reported from other studies [11, 12, 14] likely due to the younger age of the 260

vaccine recipients in the present study. 261

262

Vaccine-type neutralizing antibody titers remained high and above the GMT observed in a study of 263

natural infection for both HPV16 and HPV18. Cervarix® titers continued to be higher than Gardasil® 264

titers. When calibrated against the International Standards for HPV16 and HPV18 antibodies the 265

GMC for HPV16 and HPV18 antibodies remained 1-2 orders of magnitude higher than the natural 266

infection antibody level. The decline in vaccine-type neutralizing antibody titers over the study 267

period was estimated to be 30% every 5-7 years and would be expected to remain above the level 268

of natural infection at least 15 years following three doses of HPV vaccine in keeping with studies 269

that have attempted to model the durability of the vaccine type neutralizing antibody response [14]. 270

The robust immune memory induced by the HPV vaccines, exemplified by the durability of the 271

antibody response, is supported by studies demonstrating a strong anamnestic response to a 272

booster dose 5-7 years following initial immunization [15, 16]. 273

274

Neutralizing antibody seropositivity to non-vaccine types was lower than for vaccine types. 275

Nevertheless, responses against both HPV31 and HPV45 were detectable after 7 years, with 276

Cervarix® vaccinees exhibiting higher rates of seropositivity than Gardasil® vaccinees. There are 277

few studies examining the durability of the neutralizing antibody response against non-vaccine 278

10

types. The rates seen here are higher than those found in a head to head study of 18 – 26 year old 279

women 2 years after immunization [17] and for HPV31 higher than that reported from a study of 18 280

– 25 year old women 4 years after initial immunization with Cervarix® [18]. Non-vaccine type titers 281

were substantially lower than vaccine type titers following initial immunization and remained so 282

during follow up. There are no International Standards for HPV31 or HPV45 antibodies so it was not 283

possible to benchmark the titers observed here although they were clearly lower than the typical 284

GMT of antibodies elicited following natural infection. The antibody decline rate within those with an 285

initial response was similar to that estimated for vaccine-type antibodies and it is estimated that low 286

levels of antibodies against non-vaccine types would be detectable for many years following 287

vaccination in some individuals. 288

289

Following recommendation by the World Health Organization [19], many countries have 290

implemented a two dose HPV vaccination schedule for young girls. Limited non-vaccine type 291

serological data derived from reduced dosing schedules are available but anecdotal data from a trial 292

of older women vaccinated with Cervarix® does suggest [18] a reduced seropositivity to the non-293

vaccine type, HPV31, although vaccine efficacy against this type remains high [20]. 294

295

There are no defined correlates of protection for HPV vaccination. Empirical studies have suggested 296

that the amount of vaccine-type antibody required to protect mice challenged with HPV PsV in vivo 297

is substantially lower than that required to neutralize the same PsV in vitro [21]. Although the level 298

of natural infection antibody has been used as a benchmark for gauging vaccine immunity in 299

vaccine efficacy studies, little is known about the specificity of antibodies elicited during natural 300

infection although there are suggestions that they can be modestly protective [22]. A study of B cell 301

clones suggested that the specificity of antibodies elicited by vaccination, while overlapping with 302

those derived from natural infection in some cases, were largely distinct in their specificity [23]. In 303

addition, cross-reactive HPV31 antibodies elicited by the HPV vaccines appear to target antigenic 304

domains distinct from those targeted by type-specific HPV31 antibodies [24]. Vaccine efficacy [2] 305

and impact surveillance [5] studies suggest that vaccine effectiveness against HPV31 and HPV45 306

genotypes is significant, despite having relatively lower immunity against these genotypes. Indeed, 307

in reduced dose settings vaccine efficacy against these genotypes remained robust [20]. Taken 308

together these data might suggest that, despite the low levels of non-vaccine type antibodies 309

elicited by the HPV vaccines, it is their specificity that distinguishes them from type-specific 310

antibodies elicited by natural infection and contributes to the vaccine effectiveness against non-311

vaccine types observed in vaccine trials and impact studies [2, 5]. A recent review [25] and post-hoc 312

analysis of vaccine efficacy data [26] have attempted to delineate possible mechanism(s) behind 313

the potency of the HPV vaccines, including the activation of additional immune pathways and 314

uncertainty about the levels and site of action of these immune effectors. The role that neutralizing 315

11

antibodies play in the long-term protection against infection and subsequent disease is uncertain 316

and the utility of such antibodies as a potential correlate or surrogate of such protection in unclear. 317

318

This study has several strengths including the evaluation of the durability of the immune response to 319

both vaccines in participants at the target age for vaccination in national immunization programmes; 320

evaluation in the context of a randomized head-to-head trial; the reporting of vaccine and natural 321

infection antibody levels in International Units for the vaccine types; and the reporting of data to 7 322

years derived from the neutralization assay which is considered to be the gold standard for in vitro 323

serology testing. Shortcomings in this study include the relatively low number of responders to the 324

follow up study which inevitably impacts on the precision of some of the estimates. Nevertheless, 325

these data provide additional empirical support for the observed effectiveness of the HPV vaccines, 326

particularly against non-vaccine genotypes. 327

328

The nonavalent Gardasil®9 vaccine has demonstrated broad efficacy in a three dose schedule [27] 329

and will likely be adopted by many national immunization programmes in time. However, tens of 330

millions of adolescent girls have been vaccinated with the bivalent or quadrivalent vaccine [28], and 331

a better understanding of vaccine immunity, particularly the breadth, magnitude and durability of the 332

antibody responses, is warranted. 333

334

12

Acknowledgments 335

Special thanks to the study participants without whom this study would not have been possible and 336

we are grateful to the NVEC vaccine research nurses and other staff for the execution of this study. 337

We are indebted to Prof. John T. Schiller and Dr. Chris Buck (National Cancer Institute, Bethesda, 338

MD., U.S.A.) for use of the pseudovirus clones. 339

340

Author Contributions 341

Study design: AG KP NA JS PT EM SB. Data generation: AG KP MH. Data handling and analysis: 342

KP NA SB. Material contribution: CEC EM SB. Manuscript preparation: AG KP MH CEC NA JS PT 343

EM SB. 344

345

Declarations of interest 346

None 347

348

Funding 349

This publication is independent research funded by the National Institute for Health Research Policy 350

Research Programme (“Vaccine Evaluation Consortium Phase II”, 039/0031). The views expressed 351

in this publication are those of the author(s) and not necessarily those of the NHS, the National 352

Institute for Health Research or the Department of Health and Social Care. 353

354

13

References 355

[1] Herrero R, Gonzalez P, Markowitz LE. Present status of human papillomavirus vaccine development and 356

implementation. Lancet Oncol. 2015;16:e206-e16. 357

[2] Lehtinen M, Dillner J. Clinical trials of human papillomavirus vaccines and beyond. Nature reviews Clinical 358

oncology. 2013;10:400-10. 359

[3] Schiller JT, Lowy DR. Understanding and learning from the success of prophylactic human papillomavirus 360

vaccines. Nat Rev Microbiol. 2012;10:681-92. 361

[4] Drolet M, Benard E, Boily MC, Ali H, Baandrup L, Bauer H, Beddows S, Brisson J, Brotherton JM, 362

Cummings T, Donovan B, Fairley CK, Flagg EW, Johnson AM, Kahn JA, Kavanagh K, Kjaer SK, Kliewer EV, 363

Lemieux-Mellouki P, Markowitz L, Mboup A, Mesher D, Niccolai L, Oliphant J, Pollock KG, Soldan K, 364

Sonnenberg P, Tabrizi SN, Tanton C, Brisson M. Population-level impact and herd effects following human 365

papillomavirus vaccination programmes: a systematic review and meta-analysis. Lancet Infect Dis. 366

2015;15:565-80. 367

[5] Mesher D, Soldan K, Lehtinen M, Beddows S, Brisson M, Brotherton JM, Chow EP, Cummings T, Drolet 368

M, Fairley CK, Garland SM, Kahn JA, Kavanagh K, Markowitz L, Pollock KG, Soderlund-Strand A, 369

Sonnenberg P, Tabrizi SN, Tanton C, Unger E, Thomas SL. Population-Level Effects of Human 370

Papillomavirus Vaccination Programs on Infections with Nonvaccine Genotypes. Emerg Infect Dis. 371

2016;22:1732-40. 372

[6] Bissett SL, Godi A, Jit M, Beddows S. Seropositivity to non-vaccine incorporated genotypes induced by the 373

bivalent and quadrivalent HPV vaccines: A systematic review and meta-analysis. Vaccine. 2017;35:3922-9. 374

[7] Draper E, Bissett SL, Howell-Jones R, Waight P, Soldan K, Jit M, Andrews N, Miller E, Beddows S. A 375

randomized, observer-blinded immunogenicity trial of Cervarix((R)) and Gardasil((R)) Human Papillomavirus 376

vaccines in 12-15 year old girls. PLoS One. 2013;8:e61825. 377

[8] Bissett SL, Wilkinson D, Tettmar KI, Jones N, Stanford E, Panicker G, Faust H, Borrow R, Soldan K, Unger 378

ER, Dillner J, Minor P, Beddows S. Human papillomavirus antibody reference reagents for use in 379

postvaccination surveillance serology. Clin Vaccine Immunol. 2012;19:449-51. 380

[9] Godi A, Epifano I, Bissett SL, Dell'Anna T, Piana A, Cocuzza C, Beddows S. Amino acid motifs in both the 381

major and minor capsid proteins of HPV51 impact antigenicity and infectivity. J Gen Virol. 2015;96:1842 - 9. 382

[10] Ferris D, Samakoses R, Block SL, Lazcano-Ponce E, Restrepo JA, Reisinger KS, Mehlsen J, Chatterjee 383

A, Iversen OE, Sings HL, Shou Q, Sausser TA, Saah A. Long-term study of a quadrivalent human 384

papillomavirus vaccine. Pediatrics. 2014;134:e657-65. 385

14

[11] Nygard M, Saah A, Munk C, Tryggvadottir L, Enerly E, Hortlund M, Sigurdardottir LG, Vuocolo S, Kjaer 386

SK, Dillner J. Evaluation of the Long-Term Anti-Human Papillomavirus 6 (HPV6), 11, 16, and 18 Immune 387

Responses Generated by the Quadrivalent HPV Vaccine. Clin Vaccine Immunol. 2015;22:943-8. 388

[12] Schwarz TF, Galaj A, Spaczynski M, Wysocki J, Kaufmann AM, Poncelet S, Suryakiran PV, Folschweiller 389

N, Thomas F, Lin L, Struyf F. Ten-year immune persistence and safety of the HPV-16/18 AS04-adjuvanted 390

vaccine in females vaccinated at 15-55 years of age. Cancer medicine. 2017;6:2723-31. 391

[13] Artemchuk H, Eriksson T, Poljak M, Surcel HM, Dillner J, Lehtinen M, Faust H. Long-term Antibody 392

Response to Human Papillomavirus Vaccines: Up to 12 Years of Follow-up in the Finnish Maternity Cohort. J 393

Infect Dis. 2019;219:582-9. 394

[14] Einstein MH, Takacs P, Chatterjee A, Sperling RS, Chakhtoura N, Blatter MM, Lalezari J, David MP, Lin 395

L, Struyf F, Dubin G. Comparison of long-term immunogenicity and safety of human papillomavirus (HPV)-396

16/18 AS04-adjuvanted vaccine and HPV-6/11/16/18 vaccine in healthy women aged 18-45 years: end-of-397

study analysis of a Phase III randomized trial. Hum Vaccin Immunother. 2014;10:3435-45. 398

[15] Moscicki AB, Wheeler CM, Romanowski B, Hedrick J, Gall S, Ferris D, Poncelet S, Zahaf T, Moris P, 399

Geeraerts B, Descamps D, Schuind A. Immune responses elicited by a fourth dose of the HPV-16/18 AS04-400

adjuvanted vaccine in previously vaccinated adult women. Vaccine. 2012;31:234-41. 401

[16] Olsson SE, Villa LL, Costa RL, Petta CA, Andrade RP, Malm C, Iversen OE, Hoye J, Steinwall M, Riis-402

Johannessen G, Andersson-Ellstrom A, Elfgren K, von Krogh G, Lehtinen M, Paavonen J, Tamms GM, 403

Giacoletti K, Lupinacci L, Esser MT, Vuocolo SC, Saah AJ, Barr E. Induction of immune memory following 404

administration of a prophylactic quadrivalent human papillomavirus (HPV) types 6/11/16/18 L1 virus-like 405

particle (VLP) vaccine. Vaccine. 2007;25:4931-9. 406

[17] Einstein MH, Baron M, Levin MJ, Chatterjee A, Fox B, Scholar S, Rosen J, Chakhtoura N, Lebacq M, van 407

der Most R, Moris P, Giannini SL, Schuind A, Datta SK, Descamps D. Comparison of the immunogenicity of 408

the human papillomavirus (HPV)-16/18 vaccine and the HPV-6/11/16/18 vaccine for oncogenic non-vaccine 409

types HPV-31 and HPV-45 in healthy women aged 18-45 years. Hum Vaccin. 2011;7:1359-73. 410

[18] Safaeian M, Porras C, Pan Y, Kreimer A, Schiller JT, Gonzalez P, Lowy DR, Wacholder S, Schiffman M, 411

Rodriguez AC, Herrero R, Kemp T, Shelton G, Quint W, van Doorn LJ, Hildesheim A, Pinto LA. Durable 412

antibody responses following one dose of the bivalent human papillomavirus L1 virus-like particle vaccine in 413

the Costa Rica Vaccine Trial. Cancer Prev Res (Phila). 2013;6:1242-50. 414

[19] WHO. Human papillomavirus vaccines: WHO position paper, October 2014-Recommendations. Vaccine. 415

2015;33:4383-4. 416

[20] Kreimer AR, Struyf F, Del Rosario-Raymundo MR, Hildesheim A, Skinner SR, Wacholder S, Garland SM, 417

Herrero R, David MP, Wheeler CM. Efficacy of fewer than three doses of an HPV-16/18 AS04-adjuvanted 418

15

vaccine: combined analysis of data from the Costa Rica Vaccine and PATRICIA trials. Lancet Oncol. 419

2015;16:775-86. 420

[21] Longet S, Schiller JT, Bobst M, Jichlinski P, Nardelli-Haefliger D. A murine genital-challenge model is a 421

sensitive measure of protective antibodies against human papillomavirus infection. J Virol. 2011;85:13253-9. 422

[22] Beachler DC, Jenkins G, Safaeian M, Kreimer AR, Wentzensen N. Natural Acquired Immunity Against 423

Subsequent Genital Human Papillomavirus Infection: A Systematic Review and Meta-analysis. J Infect Dis. 424

2016;213:1444-54. 425

[23] Scherer EM, Smith RA, Gallego DF, Carter JJ, Wipf GC, Hoyos M, Stern M, Thurston T, Trinklein ND, 426

Wald A, Galloway DA. A Single Human Papillomavirus Vaccine Dose Improves B Cell Memory in Previously 427

Infected Subjects. EBioMedicine. 2016;10:55-64. 428

[24] Bissett SL, Godi A, Beddows S. The DE and FG loops of the HPV major capsid protein contribute to the 429

epitopes of vaccine-induced cross-neutralising antibodies. Scientific reports. 2016;6:39730. 430

[25] Schiller J, Lowy D. Explanations for the high potency of HPV prophylactic vaccines. Vaccine. 431

2018;36:4768-73. 432

[26] Ryser M, Berlaimont V, Karkada N, Mihalyi A, Rappuoli R, van der Most R. Post-hoc analysis from phase 433

III trials of human papillomavirus vaccines: considerations on impact on non-vaccine types. Expert Rev 434

Vaccines. 2019:1-14. 435

[27] Joura EA, Giuliano AR, Iversen OE, Bouchard C, Mao C, Mehlsen J, Moreira ED, Jr., Ngan Y, Petersen 436

LK, Lazcano-Ponce E, Pitisuttithum P, Restrepo JA, Stuart G, Woelber L, Yang YC, Cuzick J, Garland SM, 437

Huh W, Kjaer SK, Bautista OM, Chan IS, Chen J, Gesser R, Moeller E, Ritter M, Vuocolo S, Luxembourg A. A 438

9-valent HPV vaccine against infection and intraepithelial neoplasia in women. N Engl J Med. 2015;372:711-439

23. 440

[28] Bruni L, Diaz M, Barrionuevo-Rosas L, Herrero R, Bray F, Bosch FX, de Sanjose S, Castellsague X. 441

Global estimates of human papillomavirus vaccination coverage by region and income level: a pooled 442

analysis. The Lancet Global health. 2016;4:e453-63. 443

444

16

Figure legends 445

446

Figure 1. CONSORT diagram. Allocations, interventions, follow up and analysis of subjects 447

recruited into the comparative HPV vaccine immunogenicity study with the current extension study 448

participants highlighted (red box). 449

450

Figure 2: Seropositivity and neutralizing antibody titers 451

Left panels: Percentage seropositive and 95% confidence intervals. Right panels: Error bars denote 452

95% confidence intervals of geometric mean titers (GMT). Antibody titers are presented on a Log10 453

scale. Black dashed line, limit of detection (LOD) of the assay. Titers <40 were assigned a censored 454

value of 20 for analysis purposes. Grey dashed line, GMT of neutralizing antibody titers in women 455

from a separate study who were, at the time of sampling, HPV seropositive and HPV DNA negative 456

for the genotype under study and represent typical antibody titers induced by natural infection for: 457

HPV16 (196; 125 – 309; n=29), HPV18 (225; 134 – 378; n=17), HPV31 (356; 213 – 594; n=20), and 458

HPV45 (93; 65 – 133; n=13). 459

460

Figure 3: Modelling antibody declines by time since vaccination using a log-titre log-time 461

relationship 462

The relationship between time since vaccination and antibody titers was modelled using all post 463

vaccination data (M7, M12 and M84) using a log-time on log-titre mixed effects model with random 464

slope and intercept. For HPV31 and HPV45 this was done within the subset with a response at M7. 465

Antibody titers are presented on a Log10 scale. Filled circles represent individual point data for 466

Cervarix® (Blue) and Gardasil® (Red) vaccinees. Titers <40 were assigned a censored value of 20 467

for analysis purposes. Solid lines represent model estimate for Cervarix® (Blue) and Gardasil® 468

(Red) vaccines with dashed lines showing 95% confidence intervals on the modelled declines. 469

Black dashed line, limit of detection (LOD) of the assay. Antibody declines by time since first 470

vaccination extrapolated to 15 years (184 months). For plotting, data were modelled by time since 471

3rd vaccination dose then shifted by 6 months to show as time since first vaccination. 472

Table 1. Seropositivity and geometric mean neutralizing antibody titers against vaccine and non-vaccine type HPV pseudoviruses

Percentage seropositive Geometric mean titers

HPV Timing Cervarix Gardasil p value Cervarix Gardasil p value

16 M0 0/28 0% (0-12%) 1/27 4% (0-19%) 1.000 20 (20-20) 21 (19-24) 0.308

M2 28/28 100% (88-100%) 28/28 100% (88-100%) 1.000 4589 (3357-6274) 1885 (1188-2992) 0.002

M7 26/26 100% (87-100%) 29/29 100% (88-100%) 1.000 78951 (57715-107999) 25153 (16566-38191) <0.001

M12 28/28 100% (88-100%) 30/30 100% (88-100%) 1.000 32089 (22522-45720) 11803 (8017-17376) <0.001

M84 28/28 100% (88-100%) 30/30 100% (88-100%) 1.000 11661 (8272-16439) 2898 (1854-4530) <0.001

Fold change 0.36 (0.28-0.48) 0.25 (0.19-0.31) 0.090

18 M0 0/28 0% (0-12%) 0/27 0% (0-13%) N/A 20 (20-20) 20 (20-20) 1.000

M2 28/28 100% (88-100%) 28/28 100% (88-100%) 1.000 2694 (1818-3993) 611 (400-932) 0.001

M7 26/26 100% (87-100%) 29/29 100% (88-100%) 1.000 45395 (27547-74808) 10660 (6618-17170) 0.002

M12 28/28 100% (88-100%) 30/30 100% (88-100%) 1.000 10489 (6617-16626) 3011 (1897-4778) 0.003

M84 28/28 100% (88-100%) 29/30 97% (83-100%) 1.000 3201 (2149-4768) 799 (494-1292) 0.002

Fold change 0.31 (0.23-0.41) 0.27 (0.22-0.33) 0.418

31 M0 1/28 4% (0-18%) 0/27 0% (0-13%) 1.000 22 (19-25) 20 (20-20) 0.326

M2 7/28 25% (11-45%) 3/28 11% (2-28%) 0.314 33 (22-49) 24 (19-31) 0.166

M7 23/26 88% (70-98%) 20/29 69% (49-85%) 0.684 396 (192-817) 121 (65-223) 0.019

M12 23/28 82% (63-94%) 21/30 70% (51-85%) 0.842 196 (105-368) 100 (58-172) 0.089

M84 21/28 75% (55-89%) 15/30 50% (31-69%) 0.399 110 (65-185) 56 (37-85) 0.069

Fold change 0.56 (0.4-0.79) 0.56 (0.44-0.72) 0.434

45 M0 0/28 0% (0-12%) 0/27 0% (0-13%) N/A 20 (20-20) 20 (20-20) 1.000

M2 0/28 0% (0-12%) 1/28 4% (0-18%) 1.000 20 (20-20) 21 (19-22) 0.317

M7 10/26 38% (20-59%) 1/29 3% (0-18%) 0.009 39 (26-59) 21 (19-23) 0.001

M12 7/28 25% (11-45%) 1/30 3% (0-17%) 0.058 32 (23-46) 21 (19-22) 0.015

M84 7/28 25% (11-45%) 1/30 3% (0-17%) 0.058 26 (21-32) 21 (19-22) 0.018

Fold change 0.82 (0.64-1.05) 1 (0.9-1.1) 0.306

p values<0.05 highlighted in bold type. N/A, not applicable. Fold change refers to the difference between M84 and M12 neutralizing antibody titers. Titers <40 were assigned a censored value of 20 for analysis purposes.

Table-1

Assessed for eligibility (n=198)

Excluded (n=0)

Analysed

• Month 0 (n=94)

• Month 2 (n=91)

• Month 7 (n=91)

• Month 12 (n=92) Excluded from analysis

• Month 0 (n=1) • Month 2 (n=2) • Month 7 (n=3) • Month 12 (n=1)

Lost to follow-up

• Month 0 (n=1)

• Month 2 (n=3)

• Month 7 (n=2)

• Month 12 (n=3) Discontinued intervention (n=0)

Allo

ca

tio

n

An

aly

sis

F

ollo

w-U

p

Randomized (n=198)

En

roll

me

nt

Allocated to Cervarix (n=96)

• Received all three doses (n=96)

Lost to follow-up

• Month 0 (n=9)

• Month 2 (n=3)

• Month 7 (n=3)

• Month 12 (n=5) Discontinued intervention (n=0)

Analysed

• Month 0 (n=93)

• Month 2 (n=98)

• Month 7 (n=97)

• Month 12 (n=96) Excluded from analysis

• Month 0 (n=0) • Month 2 (n=1) • Month 7 (n=2) • Month 12 (n=1)

Allocated to Gardasil (n=102)

• Received all three doses (n=102)

Figure 1

Analysed

• Month 84 (n=28) Excluded from analysis

• Month 84 (n=0)

Analysed

• Month 84 (n=30) Excluded from analysis

• Month 84 (n=0)

Ex

ten

sio

n

Figure-1

Figure 2

10

100

1,000

10,000

100,000

1,000,000

0 12 24 36 48 60 72 84

Tit

er

Months post 1st immunization

HPV16Cervarix

Gardasil

10

100

1,000

10,000

100,000

1,000,000

0 12 24 36 48 60 72 84

Tit

er

Months post 1st immunization

HPV18

10

100

1,000

10,000

100,000

1,000,000

0 12 24 36 48 60 72 84

Tit

er

Months post 1st immunization

HPV31

10

100

1,000

10,000

100,000

1,000,000

0 12 24 36 48 60 72 84

Tit

er

Months post 1st immunization

HPV45

0%

20%

40%

60%

80%

100%

0 2 7 12 84

Perc

en

tag

e p

osit

ive

Months post 1st immunization

HPV16

Cervarix

Gardasil

0%

20%

40%

60%

80%

100%

0 2 7 12 84

Perc

en

tag

e p

osit

ive

Months post 1st immunization

HPV18

0%

20%

40%

60%

80%

100%

0 2 7 12 84

Perc

en

tag

e p

osit

ive

Months post 1st immunization

HPV31

0%

20%

40%

60%

80%

100%

0 2 7 12 84

Perc

en

tag

e p

osit

ive

Months post 1st immunization

HPV45

Figure-2

Figure 3

10

100

1000

10000

100000

1000000

0 24 48 72 96 120 144 168 192

Tit

er

Time since first vaccination in months

HPV16

10

100

1000

10000

100000

1000000

0 24 48 72 96 120 144 168 192

Tit

er

Time since first vaccination in months

HPV18

10

100

1000

10000

100000

0 24 48 72 96 120 144 168 192

Tit

er

Time since first vaccination in months

HPV31

10

100

1000

10000

100000

0 24 48 72 96 120 144 168 192

Tit

er

Time since first vaccination in months

HPV45

Figure-3