Immunogenicity and HPV infection after one, two, and three doses of quadrivalent HPV vaccine in girls in India: a multicentre prospective cohort study

Rengaswamy Sankaranarayanan, Priya Ramesh Prabhu, Michael Pawlita, Tarik Gheit, Neerja Bhatla, Richard Muwonge, Bhagwan M Nene, Pulikottil Okuru Esmy, Smita Joshi, Usha Rani Reddy Poli, Parimal Jivarajani, Yogesh Verma, Eric Zomawia, Maqsood Siddiqi, Surendra S Shastri, Kasturi Jayant, Sylla G Malvi, Eric Lucas, Angelika Michel, Julia Butt, Janki Mohan Babu Vijayamma, Subha Sankaran, Thiraviam Pillai Rameshwari Ammal Kannan, Rintu Varghese, Uma Divate, Shila Thomas, Geeta Joshi, Martina Willhauck-Fleckenstein, Tim Waterboer, Martin Müller, Peter Sehr, Sanjay Hingmire, Alka Kriplani, Gauravi Mishra, Sharmila Pimple, Radhika Jadhav, Catherine Sauvaget, Massimo Tommasino, Madhavan Radhakrishna Pillai, Indian HPV Vaccine Study Group, Rengaswamy Sankaranarayanan, Priya Ramesh Prabhu, Michael Pawlita, Tarik Gheit, Neerja Bhatla, Richard Muwonge, Bhagwan M Nene, Pulikottil Okuru Esmy, Smita Joshi, Usha Rani Reddy Poli, Parimal Jivarajani, Yogesh Verma, Eric Zomawia, Maqsood Siddiqi, Surendra S Shastri, Kasturi Jayant, Sylla G Malvi, Eric Lucas, Angelika Michel, Julia Butt, Janki Mohan Babu Vijayamma, Subha Sankaran, Thiraviam Pillai Rameshwari Ammal Kannan, Rintu Varghese, Uma Divate, Shila Thomas, Geeta Joshi, Martina Willhauck-Fleckenstein, Tim Waterboer, Martin Müller, Peter Sehr, Sanjay Hingmire, Alka Kriplani, Gauravi Mishra, Sharmila Pimple, Radhika Jadhav, Catherine Sauvaget, Massimo Tommasino, Madhavan Radhakrishna Pillai, Indian HPV Vaccine Study Group

Abstract

Background: An increase in worldwide HPV vaccination could be facilitated if fewer than three doses of vaccine are as effective as three doses. We originally aimed to compare the immunogenicity and frequency of persistent infection and cervical precancerous lesions caused by vaccine-targeted HPV after vaccination with two doses of quadrivalent vaccine on days 1 and 180 or later, with three doses on days 1, 60, and 180 or later, in a cluster-randomised trial. Suspension of the recruitment and vaccination due to events unrelated to our study meant that some enrolled girls could not be vaccinated and some vaccinated girls received fewer than the planned number of vaccinations by default. As a result, we re-analysed our data as an observational cohort study.

Methods: Our study was designed to be done in nine locations (188 clusters) in India. Participants were unmarried girls aged 10-18 years vaccinated in four cohorts: girls who received three doses of vaccine on days 1, 60, and 180 or later, two doses on days 1 and 180 or later, two doses on days 1 and 60 by default, and one dose by default. The primary outcomes were immunogenicity in terms of L1 genotype-specific binding antibody titres, neutralising antibody titres, and antibody avidity after vaccination for the vaccine-targeted HPV types 16, 18, 6, and 11 and incident and persistent infections with these HPVs. Analysis was per actual number of vaccine doses received. This study is registered with ISRCTN, number ISRCTN98283094; and with ClinicalTrials.gov, number NCT00923702.

Findings: Vaccination of eligible girls was initiated on Sept 1, 2009, and continued until April 8, 2010. Of 21 258 eligible girls identified at 188 clusters, 17 729 girls were recruited from 178 clusters before suspension. 4348 (25%) girls received three doses, 4979 (28%) received two doses on days 1 and 180 or later, 3452 (19%) received two doses at days 1 and 60, and 4950 (28%) received one dose. Immune response in the two-dose HPV vaccine group was non-inferior to the three-dose group (median fluorescence intensity ratio for HPV 16 1·12 [95% CI 1·02-1·23] and for HPV 18 1·04 [0·92-1·19]) at 7 months, but was inferior in the two-dose default (0·33 [0·29-0·38] for HPV 16 and 0·51 [0·43-0·59] for HPV 18) and one-dose default (0·09 [0·08-0·11] for HPV 16 and 0·12 [0·10-0·14] for HPV 18) groups at 18 months. The geometric mean avidity indices after fewer than three doses by design or default were non-inferior to those after three doses of vaccine. Fewer than three doses by design and default induced detectable concentrations of neutralising antibodies to all four vaccine-targeted HPV types, but at much lower concentration after one dose. Cervical samples from 2649 participants were tested and the frequency of incident HPV 16, 18, 6, and 11 infections was similar irrespective of the number of vaccine doses received. The testing of at least two samples from 838 participants showed that there was no persistent HPV 16 or 18 infections in any study group at a median follow-up of 4·7 years (IQR 4·2-5·1).

Interpretation: Despite the limitations imposed by the suspension of the HPV vaccination, our findings lend support to the WHO recommendation of two doses, at least 6 months apart, for routine vaccination of young girls. The short-term protection afforded by one dose of HPV vaccine against persistent infection with HPV 16, 18, 6, and 11 is similar to that afforded by two or three doses of vaccine and merits further assessment.

Funding: Bill & Melinda Gates Foundation.

© 2015 International Agency for Research on Cancer. Published by Elsevier Ltd/Inc/BV. All rights reserved.

Figures

Figure 1
Figure 1
Study profile
Figure 2
Figure 2
Mean MFI values for HPV types 16, 18, 6, and 11 L1 antibodies Dashed lines show the threshold (cutoff) values for seroconversion. MFI=median fluorescence intensity. *MFI values for month 7 were used for the three-dose and two-dose vaccine groups, whereas MFI values for month 12 were used for the two-dose default and one-dose default groups.
Figure 3
Figure 3
Box plots of the avidity index of MFI for HPV types 16 (A), 18 (B), 6 (C), and 11 (D) L1 antibodies at 7 months and 18 months after the first dose MFI=median fluorescence intensity.
Figure 4
Figure 4
Box plots of neutralisation titres of HPV types 16 (A), 18 (B), and 6 (C) L1 antibodies at 18 months after the first dose Samples without neutralising activity were not included in the GMT analyses. GMT=geometric mean neutralisation titre.

References

    1. Schiller JT, Castellsague X, Garland SM. A review of clinical trials of human papillomavirus prophylactic vaccines. Vaccine. 2012;30(suppl 5):F123–F138.
    1. Bosch FX, Broker TR, Forman D. Comprehensive control of human papillomavirus infections and related diseases. Vaccine. 2013;31(suppl 5):F1–31.
    1. Erickson BK, Landers EE, Huh WK. Update on vaccination clinical trials for HPV-related disease. Clin Ther. 2014;36:8–16.
    1. WHO Summary of the SAGE April 2014 meeting. (accessed Sept 16, 2015).
    1. World Health Organization Evidence based recommendations on Human Papilloma Virus (HPV) Vaccines Schedules. Background paper for SAGE discussions. 2014. (accessed Sept 16, 2015).
    1. Block SL, Nolan T, Sattler C. Comparison of the immunogenicity and reactogenicity of a prophylactic quadrivalent human papillomavirus (types 6, 11, 16, and 18) L1 virus-like particle vaccine in male and female adolescents and young adult women. Pediatrics. 2006;118:2135–2145.
    1. Pedersen C, Petaja T, Strauss G. Immunization of early adolescent females with human papillomavirus type 16 and 18 L1 virus-like particle vaccine containing AS04 adjuvant. J Adolesc Health. 2007;40:564–571.
    1. Waterboer T, Sehr P, Michael KM. Multiplex human papillomavirus serology based on in situ-purified glutathione s-transferase fusion proteins. Clin Chem. 2005;51:1845–1853.
    1. Sehr P, Rubio I, Seitz H. High-throughput pseudovirion-based neutralization assay for analysis of natural and vaccine-induced antibodies against human papillomaviruses. PLoS One. 2013;8:e75677.
    1. Schmitt M, Dondog B, Waterboer T, Pawlita M, Tommasino M, Gheit T. Abundance of multiple high-risk human papillomavirus (HPV) infections found in cervical cells analyzed by use of an ultrasensitive HPV genotyping assay. J Clin Microbiol. 2010;48:143–149.
    1. Gheit T, Landi S, Gemignani F. Development of a sensitive and specific assay combining multiplex PCR and DNA microarray primer extension to detect high-risk mucosal human papillomavirus types. J Clin Microbiol. 2006;44:2025–2031.
    1. Reisinger KS, Block SL, Lazcano-Ponce E. Safety and persistent immunogenicity of a quadrivalent human papillomavirus types 6, 11, 16, 18 L1 virus-like particle vaccine in preadolescents and adolescents: a randomized controlled trial. Pediatr Infect Dis J. 2007;26:201–209.
    1. Neuzil KM, Canh DG, Thiem VD. Immunogenicity and reactogenicity of alternative schedules of HPV vaccine in Vietnam: a cluster randomized noninferiority trial. JAMA. 2011;305:1424–1431.
    1. Wang WW, Mehrotra DV, Chan IS, Heyse JF. Statistical considerations for noninferiority/equivalence trials in vaccine development. J Biopharm Stat. 2006;16:429–441.
    1. Sankaranarayanan R, Bhatla N, Gravitt PE. Human papillomavirus infection and cervical cancer prevention in India, Bangladesh, Sri Lanka and Nepal. Vaccine. 2008;26(suppl 12):M43–M52.
    1. Hussain S, Bharadwaj M, Nasare V. Human papillomavirus infection among young adolescents in India: impact of vaccination. J Med Virol. 2012;84:298–305.
    1. Santhya KG, Ram U, Acharya R. Pre-marital sexual relations among youth in India: findings from the Youth in India, Situations and Needs Study. XXVI International Union for the Scientific Study Population Conference; Marrakech, Morocco; Sept 27, to Oct 2, 2009. (accessed Sept 16, 2015).
    1. Kelkar DS, Patwardhan M, Joshi VD. Prevalence and causalities of tobacco consumption (TC) among adolescents: a cross sectional study at Pune. J Assoc Physicians India. 2013;61:174–178.
    1. Madaan M, Agrawal S, Puri M, Meena J, Kaur H, Trivedi SS. Health profile of urban adolescent girls from India. Int J Adolesc Med Health. 2014;26:233–237.
    1. Datta P, Bhatla N, Dar L. Prevalence of human papillomavirus infection among young women in North India. Cancer Epidemiol. 2010;34:157–161.
    1. Datta P, Bhatla N, Pandey RM. Type-specific incidence and persistence of HPV infection among young women: a prospective study in North India. Asian Pac J Cancer Prev. 2012;13:1019–1024.
    1. Dutta S, Begum R, Mazumder ID. Prevalence of human papillomavirus in women without cervical cancer: a population-based study in eastern India. Int J Gynecol Pathol. 2012;31:178–183.
    1. Sharma K, Kathait A, Jain A. Higher prevalence of human papillomavirus infection in adolescent and young adult girls belonging to different Indian tribes with varied socio-sexual lifestyle. PLoS One. 2015;10:e0125693.
    1. Joura EA, Kjaer SK, Wheeler CM. HPV antibody levels and clinical efficacy following administration of a prophylactic quadrivalent HPV vaccine. Vaccine. 2008;26:6844–6851.
    1. Dobson SR, McNeil S, Dionne M. Immunogenicity of 2 doses of HPV vaccine in younger adolescents vs 3 doses in young women: a randomized clinical trial. JAMA. 2013;309:1793–1802.
    1. Romanowski B, Schwarz TF, Ferguson LM. Immune response to the HPV-16/18 AS04-adjuvanted vaccine administered as a 2-dose or 3-dose schedule up to 4 years after vaccination: results from a randomized study. Hum Vaccin Immunother. 2014;10:1155–1165.
    1. Safaeian M, Porras C, Pan Y. Durable antibody responses following one dose of the bivalent human papillomavirus L1 virus-like particle vaccine in the Costa Rica Vaccine Trial. Cancer Prev Res (Phila) 2013;6:1242–1250.
    1. Lazcano-Ponce E, Stanley M, Munoz N. Overcoming barriers to HPV vaccination: non-inferiority of antibody response to human papillomavirus 16/18 vaccine in adolescents vaccinated with a two-dose vs. a three-dose schedule at 21 months. Vaccine. 2014;32:725–732.
    1. Kreimer AR, Rodriguez AC, Hildesheim A. Proof-of-principle evaluation of the efficacy of fewer than three doses of a bivalent HPV16/18 vaccine. J Natl Cancer Inst. 2011;103:1444–1451.
    1. Kreimer AR, Struyf F, Del Rosario-Raymundo MR, for the Costa Rica Vaccine Trial and the PATRICIA study groups Efficacy of fewer than three doses of an HPV-16/18 AS04-adjuvanted vaccine: combined analysis of data from the Costa Rica Vaccine and PATRICIA trials. Lancet Oncol. 2015;16:775–786.
    1. Breitburd F, Kirnbauer R, Hubbert NL. Immunization with viruslike particles from cottontail rabbit papillomavirus (CRPV) can protect against experimental CRPV infection. J Virol. 1995;69:3959–3963.
    1. Day PM, Kines RC, Thompson CD. In vivo mechanisms of vaccine-induced protection against HPV infection. Cell Host Microbe. 2010;8:260–270.
    1. Schiller JT, Lowy DR. Raising expectations for subunit vaccine. J Infect Dis. 2015;211:1373–1375.
    1. Bachmann MF, Jennings GT. Vaccine delivery: a matter of size, geometry, kinetics and molecular patterns. Nat Rev Immunol. 2010;10:787–796.
    1. Herweijer E, Leval A, Ploner A. Association of varying number of doses of quadrivalent human papillomavirus vaccine with incidence of condyloma. JAMA. 2014;311:597–603.
    1. Centers for Disease Control and Prevention National and state vaccination coverage among adolescents aged 13–17 years—United States, 2012. MMWR Morb Mortal Wkly Rep. 2013;62:685–693.
    1. Bertaut A, Chavanet P, Aho S, Astruc K, Douvier S, Fournel I. HPV vaccination coverage in French girls attending middle and high schools: a declarative cross sectional study in the department of Cote d'Or. Eur J Obstet Gynecol Reprod Biol. 2013;170:526–532.

Source: PubMed

3
Sottoscrivi