A randomized, observer-blinded immunogenicity trial of Cervarix(®) and Gardasil(®) Human Papillomavirus vaccines in 12-15 year old girls

Eve Draper, Sara L Bissett, Rebecca Howell-Jones, Pauline Waight, Kate Soldan, Mark Jit, Nicholas Andrews, Elizabeth Miller, Simon Beddows, Eve Draper, Sara L Bissett, Rebecca Howell-Jones, Pauline Waight, Kate Soldan, Mark Jit, Nicholas Andrews, Elizabeth Miller, Simon Beddows

Abstract

Background: The current generation of Human Papillomavirus (HPV) vaccines, Cervarix® and Gardasil®, exhibit a high degree of efficacy in clinical trials against the two high-risk (HR) genotypes represented in the vaccines (HPV16 and HPV18). High levels of neutralizing antibodies are elicited against the vaccine types, consistent with preclinical data showing that neutralizing antibodies can mediate type-specific protection in the absence of other immune effectors. The vaccines also confer protection against some closely related non-vaccine HR HPV types, although the vaccines appear to differ in their degree of cross-protection. The mechanism of vaccine-induced cross-protection is unknown. This study sought to compare the breadth and magnitudes of neutralizing antibodies against non-vaccine types elicited by both vaccines and establish whether such antibodies could be detected in the genital secretions of vaccinated individuals.

Methods and findings: Serum and genital samples were collected from 12-15 year old girls following vaccination with either Cervarix® (n = 96) or Gardasil® (n = 102) HPV vaccine. Serum-neutralizing antibody responses against non-vaccine HPV types were broader and of higher magnitude in the Cervarix®, compared to the Gardasil®, vaccinated individuals. Levels of neutralizing and binding antibodies in genital secretions were closely associated with those found in the serum (r = 0.869), with Cervarix® having a median 2.5 (inter-quartile range, 1.7-3.5) fold higher geometric mean HPV-specific IgG ratio in serum and genital samples than Gardasil® (p = 0.0047). There was a strong positive association between cross-neutralizing antibody seropositivity and available HPV vaccine trial efficacy data against non-vaccine types.

Conclusions: These data demonstrate for the first time that cross-neutralizing antibodies can be detected at the genital site of infection and support the possibility that cross-neutralizing antibodies play a role in the cross-protection against HPV infection and disease that has been reported for the current HPV vaccines.

Trial registration: ClinicalTrials.gov NCT00956553.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. CONSORT diagram.
Figure 1. CONSORT diagram.
Allocations, interventions, follow up and analysis of 198 subjects recruited into the comparative HPV vaccine immunogenicity study.
Figure 2. Neutralizing antibody responses over time…
Figure 2. Neutralizing antibody responses over time course of study.
Percentage of vaccinees with neutralization titers of ≥20 (left panels) and GMT (right panels) against indicated vaccine (HPV16, HPV18) and non-vaccine (HPV31, HPV45) types. Error bars, ±95% CI. * p<0.05; ** p<0.01; *** p<0.001. Denominators for each measure were as follows: Cervarix® (blue) M0 (n = 94), M2 (n = 91), M7 (n = 91), M12 (n = 92); Gardasil® (red) M0 (n = 93), M2 (n = 98), M7 (n = 97), M12 (n = 96).
Figure 3. Breadth of neutralizing antibody responses…
Figure 3. Breadth of neutralizing antibody responses elicited by HPV vaccines.
Percentage of serum samples neutralizing indicated number of A9 (top panel) or A7 (bottom panel) non-vaccine types within each of the low (T1), middle (T2) or high (T3) vaccine-type tertiles (dark to light gradient shading) for indicated vaccine (blue, Cervarix®; red, Gardasil®).
Figure 4. Relationship between HPV-specific vaccine antibodies…
Figure 4. Relationship between HPV-specific vaccine antibodies in sera and genital samples.
(A) Neutralization titers were normalized to IgG levels in serum and genital samples for HPV16 (left panel; Cervarix® n = 21, Gardasil® n = 28) and HPV18 (right panel; Cervarix® n = 21, Gardasil® n = 26). (B) VLP binding titers were normalized to IgG levels in serum and genital samples for HPV16 (top left panel; Cervarix® n = 20, Gardasil® n = 21), HPV18 (top right panel; Cervarix® n = 19, Gardasil® n = 21), HPV31 (bottom left panel; Cervarix® n = 14, Gardasil® n = 21) and HPV45 (bottom right panel; Cervarix® n = 17, Gardasil® n = 24). Geometric mean ratios of the HPV specific IgG/total IgG for each sample type presented for Cervarix® (blue lines) and Gardasil® (red lines). (C) Relationship between serum and genital samples using normalized neutralizing and binding antibody for all tests. Overall Pearson's (r) coefficients are given. PsV, HPV pseudovirus. VLP, HPV virus-like particle.
Figure 5. Type-specific neutralizing antibody seroprevalence in…
Figure 5. Type-specific neutralizing antibody seroprevalence in study participants against reported vaccine efficacy.
Seroprevalence data are plotted against (A) efficacy against persistent infection and (B) efficacy against CIN2+ reported from trials of Cervarix® (blue points) and Gardasil® (red points) vaccinees for each non-vaccine HPV type (HPV31, HPV33, HPV45, HPV52 and HPV58). Vertical error bars represent exact 95% confidence intervals for the efficacy data, while horizontal error bars represent exact 95% confidence intervals for the neutralization seroprevalence data. The best fitting linear relationship between the two variables (black line) and the 95% range of bootstrap estimates for this relationship (dotted lines) is also shown. Exact 95% confidence intervals for data points were calculated using the Fisher's exact method for vaccine efficacy (1-odds ratio) and Clopper-Pearson for seroprevalence.

References

    1. Arbyn M, Castellsague X, de Sanjose S, Bruni L, Saraiya M, et al. (2011) Worldwide burden of cervical cancer in 2008. Ann Oncol 22: 2675–2686.
    1. Jemal A, Bray F (2011) Center MM, Ferlay J, Ward E, et al (2011) Global cancer statistics. CA Cancer J Clin 61: 69–90.
    1. Li N, Franceschi S, Howell-Jones R, Snijders PJ, Clifford GM (2010) Human papillomavirus type distribution in 30,848 invasive cervical cancers worldwide: Variation by geographical region, histological type and year of publication. Int J Cancer 128: 927–935.
    1. de Sanjose S, Quint WG, Alemany L, Geraets DT, Klaustermeier JE, et al. (2010) Human papillomavirus genotype attribution in invasive cervical cancer: a retrospective cross-sectional worldwide study. Lancet Oncol 11: 1048–1056.
    1. Guan P, Howell-Jones R, Li N, Bruni L, de Sanjose S, et al... (2012) Human papillomavirus types in 115,789 HPV-positive women: A meta-analysis from cervical infection to cancer. Int J Cancer.
    1. Romanowski B (2011) Long term protection against cervical infection with the human papillomavirus: review of currently available vaccines. Hum Vaccin 7: 161–169.
    1. Lu B, Kumar A, Castellsague X, Giuliano AR (2011) Efficacy and safety of prophylactic vaccines against cervical HPV infection and diseases among women: a systematic review & meta-analysis. BMC Infect Dis 11: 13.
    1. Draper E, Bissett SL, Howell-Jones R, Edwards D, Munslow G, et al. (2011) Neutralization of Non-Vaccine Human Papillomavirus Pseudoviruses from the A7 and A9 Species Groups by Bivalent HPV Vaccine Sera. Vaccine 29: 8585–8590.
    1. De Carvalho N, Teixeira J, Roteli-Martins CM, Naud P, De Borba P, et al. (2010) Sustained efficacy and immunogenicity of the HPV-16/18 AS04-adjuvanted vaccine up to 7.3 years in young adult women. Vaccine 28: 6247–6255.
    1. Kemp TJ, Hildesheim A, Safaeian M, Dauner JG, Pan Y, et al. (2011) HPV16/18 L1 VLP vaccine induces cross-neutralizing antibodies that may mediate cross-protection. Vaccine 29: 2011–2014.
    1. Brown DR, Garland SM, Ferris DG, Joura E, Steben M, et al. (2011) The humoral response to Gardasil over four years as defined by total IgG and competitive Luminex immunoassay. Hum Vaccin 7: 230–238.
    1. Einstein MH, Baron M, Levin MJ, Chatterjee A, Edwards RP, et al. (2009) Comparison of the immunogenicity and safety of Cervarix and Gardasil human papillomavirus (HPV) cervical cancer vaccines in healthy women aged 18–45 years. Hum Vaccin 5: 705–719.
    1. Giannini SL, Hanon E, Moris P, Van Mechelen M, Morel S, et al. (2006) Enhanced humoral and memory B cellular immunity using HPV16/18 L1 VLP vaccine formulated with the MPL/aluminium salt combination (AS04) compared to aluminium salt only. Vaccine 24: 5937–5949.
    1. Smith JF, Brownlow M, Brown M, Kowalski R, Esser MT, et al. (2007) Antibodies from Women Immunized with Gardasil ((R)) Cross-Neutralize HPV 45 Pseudovirions. Human Vaccines 3: 109–116.
    1. Suzich JA, Ghim SJ, Palmer-Hill FJ, White WI, Tamura JK, et al. (1995) Systemic immunization with papillomavirus L1 protein completely prevents the development of viral mucosal papillomas. Proc Natl Acad Sci U S A 92: 11553–11557.
    1. Breitburd F, Kirnbauer R, Hubbert NL, Nonnenmacher B, Trin-Dinh-Desmarquet C, et al. (1995) Immunization with viruslike particles from cottontail rabbit papillomavirus (CRPV) can protect against experimental CRPV infection. J Virol 69: 3959–3963.
    1. Cohen J (2005) Public health. High hopes and dilemmas for a cervical cancer vaccine. Science 308: 618–621.
    1. Stanley M (2010) Potential mechanisms for HPV vaccine-induced long-term protection. Gynecol Oncol 118: S2–7.
    1. Schiller JT, Lowy DR (2009) Immunogenicity testing in human papillomavirus virus-like-particle vaccine trials. J Infect Dis 200: 166–171.
    1. de Villiers EM, Fauquet C, Broker TR, Bernard HU, zur Hausen H (2004) Classification of papillomaviruses. Virology 324: 17–27.
    1. Bernard HU, Burk RD, Chen Z, van Doorslaer K, Hausen H, et al. (2010) Classification of papillomaviruses (PVs) based on 189 PV types and proposal of taxonomic amendments. Virology 401: 70–79.
    1. Bouvard V, Baan R, Straif K, Grosse Y, Secretan B, et al. (2009) A review of human carcinogens--Part B: biological agents. Lancet Oncol 10: 321–322.
    1. Keam SJ, Harper DM (2008) Human papillomavirus types 16 and 18 vaccine (recombinant, AS04 adjuvanted, adsorbed) [Cervarix]. Drugs 68: 359–372.
    1. Siddiqui MA, Perry CM (2006) Human papillomavirus quadrivalent (types 6, 11, 16, 18) recombinant vaccine (Gardasil). Drugs 66: 1263–1271; discussion 1272–1263.
    1. Pastrana DV, Buck CB, Pang YY, Thompson CD, Castle PE, et al. (2004) Reactivity of human sera in a sensitive, high-throughput pseudovirus-based papillomavirus neutralization assay for HPV16 and HPV18. Virology 321: 205–216.
    1. Huo Z, Bissett SL, Giemza R, Beddows S, Oeser C, et al. (2012) Systemic and mucosal immune responses to sublingual or intramuscular Human Papilloma Virus antigens in healthy female volunteers. PLoS One 7: e33736.
    1. Bissett SL, Wilkinson D, Tettmar KI, Jones N, Stanford E, et al. (2012) Human papillomavirus antibody reference reagents for use in postvaccination surveillance serology. Clin Vaccine Immunol 19: 449–451.
    1. Schwarz TF, Kocken M, Petaja T, Einstein MH, Spaczynski M, et al. (2010) Correlation between levels of human papillomavirus (HPV)-16 and 18 antibodies in serum and cervicovaginal secretions in girls and women vaccinated with the HPV-16/18 AS04-adjuvanted vaccine. Hum Vaccin 6: 1054–1061.
    1. Wheeler CM, Castellsague X, Garland SM, Szarewski A, Paavonen J, et al. (2012) Cross-protective efficacy of HPV-16/18 AS04-adjuvanted vaccine against cervical infection and precancer caused by non-vaccine oncogenic HPV types: 4-year end-of-study analysis of the randomised, double-blind PATRICIA trial. Lancet Oncol 13: 100–110.
    1. Brown DR, Kjaer SK, Sigurdsson K, Iversen OE, Hernandez-Avila M, et al. (2009) The impact of quadrivalent human papillomavirus (HPV; types 6, 11, 16, and 18) L1 virus-like particle vaccine on infection and disease due to oncogenic nonvaccine HPV types in generally HPV-naive women aged 16–26 years. J Infect Dis 199: 926–935.
    1. Efron B, Tibshirani RJ (1998) An introduction to the bootstrap: Chapman & Hall/CRC.
    1. Einstein MH, Baron M, Levin MJ, Chatterjee A, Fox B, et al. (2011) Comparative immunogenicity and safety of human papillomavirus (HPV)-16/18 vaccine and HPV-6/11/16/18 vaccine: follow-up from months 12–24 in a Phase III randomized study of healthy women aged 18–45 years. Hum Vaccin 7: 1343–1358.
    1. Jit M, Vyse A, Borrow R, Pebody R, Soldan K, et al. (2007) Prevalence of human papillomavirus antibodies in young females in England. British Journal of Cancer 97: 989–991.
    1. Desai S, Chapman R, Jit M, Nichols T, Borrow R, et al. (2011) Prevalence of human papillomavirus antibodies in males and females in England. Sex Transm Dis 38: 622–629.
    1. Howell-Jones R, de Silva N, Akpan M, Oakeshott P, Carder C, et al. (2012) Prevalence of human papillomavirus (HPV) infections in sexually active adolescents and young women in England, prior to widespread HPV immunisation. Vaccine 30: 3867–3875.
    1. Howell-Jones R, Bailey A, Beddows S, Sargent A, de Silva N, et al. (2010) Multi-site study of HPV type-specific prevalence in women with cervical cancer, intraepithelial neoplasia and normal cytology, in England. British Journal of Cancer 103: 209–216.
    1. Harper DM, Franco EL, Wheeler CM, Moscicki AB, Romanowski B, et al. (2006) Sustained efficacy up to 4.5 years of a bivalent L1 virus-like particle vaccine against human papillomavirus types 16 and 18: follow-up from a randomised control trial. Lancet 367: 1247–1255.
    1. Villa LL, Ault KA, Giuliano AR, Costa RL, Petta CA, et al. (2006) Immunologic responses following administration of a vaccine targeting human papillomavirus Types 6, 11, 16, and 18. Vaccine 24: 5571–5583.
    1. Einstein MH, Baron M, Levin MJ, Chatterjee A, Fox B, et al. (2011) Comparison of the immunogenicity of the human papillomavirus (HPV)-16/18 vaccine and the HPV-6/11/16/18 vaccine for oncogenic non-vaccine types HPV-31 and HPV-45 in healthy women aged 18–45 years. Hum Vaccin 7: 1359–1373.
    1. Petaja T, Keranen H, Karppa T, Kawa A, Lantela S, et al. (2009) Immunogenicity and safety of human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine in healthy boys aged 10–18 years. J Adolesc Health 44: 33–40.
    1. Romanowski B, Schwarz TF, Ferguson LM, Peters K, Dionne M, et al. (2011) Immunogenicity and safety of the HPV-16/18 AS04-adjuvanted vaccine administered as a 2-dose schedule compared with the licensed 3-dose schedule: results from a randomized study. Hum Vaccin 7: 1374–1386.
    1. Krajden M, Cook D, Yu A, Chow R, Mei W, et al. (2011) Human papillomavirus 16 (HPV 16) and HPV 18 antibody responses measured by pseudovirus neutralization and competitive Luminex assays in a two- versus three-dose HPV vaccine trial. Clin Vaccine Immunol 18: 418–423.
    1. Kemp TJ, Garcia-Pineres A, Falk RT, Poncelet S, Dessy F, et al. (2008) Evaluation of systemic and mucosal anti-HPV16 and anti-HPV18 antibody responses from vaccinated women. Vaccine 26: 3608–3616.
    1. Schiller JT, Lowy DR (2012) Understanding and learning from the success of prophylactic human papillomavirus vaccines. Nat Rev Microbiol 10: 681–692.
    1. Roberts JN, Buck CB, Thompson CD, Kines R, Bernardo M, et al... (2007) Genital transmission of HPV in a mouse model is potentiated by nonoxynol–9 and inhibited by carrageenan. Nat Med.
    1. Day PM, Kines RC, Thompson CD, Jagu S, Roden RB, et al. (2010) In vivo mechanisms of vaccine-induced protection against HPV infection. Cell Host Microbe 8: 260–270.
    1. Longet S, Schiller JT, Bobst M, Jichlinski P, Nardelli-Haefliger D (2011) A murine genital-challenge model is a sensitive measure of protective antibodies against human papillomavirus infection. J Virol 85: 13253–13259.
    1. Plotkin SA (2010) Correlates of protection induced by vaccination. Clin Vaccine Immunol 17: 1055–1065.

Source: PubMed

3
Sottoscrivi