Vaccine efficacy against persistent human papillomavirus (HPV) 16/18 infection at 10 years after one, two, and three doses of quadrivalent HPV vaccine in girls in India: a multicentre, prospective, cohort study

Partha Basu, Sylla G Malvi, Smita Joshi, Neerja Bhatla, Richard Muwonge, Eric Lucas, Yogesh Verma, Pulikkottil O Esmy, Usha Rani Reddy Poli, Anand Shah, Eric Zomawia, Sharmila Pimple, Kasturi Jayant, Sanjay Hingmire, Aruna Chiwate, Uma Divate, Shachi Vashist, Gauravi Mishra, Radhika Jadhav, Maqsood Siddiqi, Subha Sankaran, Priya Ramesh Prabhu, Thiraviam Pillai Rameshwari Ammal Kannan, Rintu Varghese, Surendra S Shastri, Devasena Anantharaman, Tarik Gheit, Massimo Tommasino, Catherine Sauvaget, M Radhakrishna Pillai, Rengaswamy Sankaranarayanan, Partha Basu, Sylla G Malvi, Smita Joshi, Neerja Bhatla, Richard Muwonge, Eric Lucas, Yogesh Verma, Pulikkottil O Esmy, Usha Rani Reddy Poli, Anand Shah, Eric Zomawia, Sharmila Pimple, Kasturi Jayant, Sanjay Hingmire, Aruna Chiwate, Uma Divate, Shachi Vashist, Gauravi Mishra, Radhika Jadhav, Maqsood Siddiqi, Subha Sankaran, Priya Ramesh Prabhu, Thiraviam Pillai Rameshwari Ammal Kannan, Rintu Varghese, Surendra S Shastri, Devasena Anantharaman, Tarik Gheit, Massimo Tommasino, Catherine Sauvaget, M Radhakrishna Pillai, Rengaswamy Sankaranarayanan

Abstract

Background: A randomised trial designed to compare three and two doses of quadrivalent human papillomavirus (HPV) vaccine in adolescent girls in India was converted to a cohort study after suspension of HPV vaccination in trials by the Indian Government. In this Article, the revised aim of the cohort study was to compare vaccine efficacy of single dose to that of three and two doses in protecting against persistent HPV 16 and 18 infection at 10 years post vaccination.

Methods: In the randomised trial, unmarried girls aged 10-18 years were recruited from nine centres across India and randomly assigned to either two doses or three doses of the quadrivalent HPV vaccine (Gardasil [Merck Sharp & Dohme, Whitehouse Station, NJ, USA]; 0·5 mL administered intramuscularly). After suspension of recruitment and vaccination, the study became a longitudinal, prospective cohort study by default, and participants were allocated to four cohorts on the basis of the number vaccine doses received per protocol: the two-dose cohort (received vaccine on days 1 and 180 or later), three-dose cohort (days 1, 60, and 180 or later), two-dose default cohort (days 1 and 60 or later), and the single-dose default cohort. Participants were followed up yearly. Cervical specimens were collected from participants 18 months after marriage or 6 months after first childbirth, whichever was earlier, to assess incident and persistent HPV infections. Married participants were screened for cervical cancer as they reached 25 years of age. Unvaccinated women age-matched to the married vaccinated participants were recruited to serve as controls. Vaccine efficacy against persistent HPV 16 and 18 infections (the primary endpoint) was analysed for single-dose recipients and compared with that in two-dose and three-dose recipients after adjusting for imbalance in the distribution of potential confounders between the unvaccinated and vaccinated cohorts. This trial is registered with ISRCTN, ISRCTN98283094, and ClinicalTrials.gov, NCT00923702.

Findings: Vaccinated participants were recruited between Sept 1, 2009, and April 8, 2010 (date of vaccination suspension), and followed up over a median duration of 9·0 years (IQR 8·2-9·6). 4348 participants had three doses, 4980 had two doses (0 and 6 months), and 4949 had a single dose. Vaccine efficacy against persistent HPV 16 and 18 infection among participants evaluable for the endpoint was 95·4% (95% CI 85·0-99·9) in the single-dose default cohort (2135 women assessed), 93·1% (77·3-99·8) in the two-dose cohort (1452 women assessed), and 93·3% (77·5-99·7) in three-dose recipients (1460 women assessed).

Interpretation: A single dose of HPV vaccine provides similar protection against persistent infection from HPV 16 and 18, the genotypes responsible for nearly 70% of cervical cancers, to that provided by two or three doses.

Funding: Bill & Melinda Gates Foundation.

Conflict of interest statement

Declaration of interests PB has received research funding from GlaxoSmithKline through the Chittaranjan National Cancer Institute (Kolkata, India) during his previous position at the institute. NB has received research funding through her institute from GlaxoSmithKline and Merck. SJ has received funds from GlaxoSmithKline through the Jehangir Clinical Development Center (Pune, India) for a human papillomavirus vaccine study. All other authors declare no competing interests.

© 2021 World Health Organization; licensee Elsevier. This is an Open Access article published under the CC BY 3.0 IGO license which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. In any use of this article, there should be no suggestion that WHO endorses any specific organisation, products or services. The use of the WHO logo is not permitted. This notice should be preserved along with the article's original URL.

Figures

Figure 1
Figure 1
Study flow chart HC II=Hybrid Capture II. HPV=human papillomavirus.
Figure 2
Figure 2
Incidence of HPV 16 and 18 (A), and HPV 31, 33, and 45 (B) HPV=human papillomavirus.

References

    1. Brisson M, Kim JJ, Canfell K. Impact of HPV vaccination and cervical screening on cervical cancer elimination: a comparative modelling analysis in 78 low-income and lower-middle-income countries. Lancet. 2020;395:575–590.
    1. WHO Immunization, vaccines and biologicals.
    1. WHO Immunization coverage. July 15, 2021.
    1. Bruni L, Saura-Lázaro A, Montoliu A. HPV vaccination introduction worldwide and WHO and UNICEF estimates of national HPV immunization coverage 2010–2019. Prev Med. 2021;144
    1. UNICEF Human papillomavirus (HPV) vaccine: supply and demand update. October, 2020.
    1. Orumaa M, Kjaer SK, Dehlendorff C. The impact of HPV multi-cohort vaccination: real-world evidence of faster control of HPV-related morbidity. Vaccine. 2020;38:1345–1351.
    1. Sankaranarayanan R, Prabhu PR, Pawlita M. Immunogenicity and HPV infection after one, two, and three doses of quadrivalent HPV vaccine in girls in India: a multicentre prospective cohort study. Lancet Oncol. 2016;17:67–77.
    1. Sankaranarayanan R, Basu P, Kaur P. Current status of human papillomavirus vaccination in India's cervical cancer prevention efforts. Lancet Oncol. 2019;20:e637–e644.
    1. Sankaranarayanan R, Joshi S, Muwonge R. Can a single dose of human papillomavirus (HPV) vaccine prevent cervical cancer? Early findings from an Indian study. Vaccine. 2018;36:4783–4791.
    1. Basu P, Sankaranarayanan R. Atlas of colposcopy: principles and practice. 2017.
    1. Eklund C, Forslund O, Wallin KL, Dillner J. Continuing global improvement in human papillomavirus DNA genotyping services: the 2013 and 2014 HPV LabNet international proficiency studies. J Clin Virol. 2018;101:74–85.
    1. Tsang SH, Sampson JN, Schussler J. Durability of cross-protection by different schedules of the bivalent HPV vaccine: the CVT Trial. J Natl Cancer Inst. 2020;112:1030–1037.
    1. Rothman KJ, Boice JD, Austin H. Epidemiology Resources; Boston, MA: 1982. Epidemiologic analysis with a programmable calculator.
    1. Sullivan LM, Massaro JM, D'Agostino RB., Sr Presentation of multivariate data for clinical use: the Framingham Study risk score functions. Stat Med. 2004;23:1631–1660.
    1. International Agency for Research on Cancer/WHO . vol 7. IARC; Lyon: 2014. Primary end-points for prophylactic HPV vaccine trials. IARC Working Group report.
    1. Bosch FX, Qiao YL, Castellsagué X. Chapter 2. The epidemiology of human papillomavirus infection and its association with cervical cancer. Int J Gynaecol Obstet. 2006;94(suppl 1):S8–21.
    1. Kreimer AR, Struyf F, Del Rosario-Raymundo MR. 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. Basu P, Muwonge R, Bhatla N. Two-dose recommendation for human papillomavirus vaccine can be extended up to 18 years—updated evidence from Indian follow-up cohort study. Papillomavirus Res. 2019;7:75–81.
    1. Kreimer AR, Sampson JN, Porras C. Evaluation of durability of a single dose of the bivalent HPV vaccine: the CVT Trial. J Natl Cancer Inst. 2020;112:1038–1046.
    1. Bachmann MF, Jennings GT. Vaccine delivery: a matter of size, geometry, kinetics and molecular patterns. Nat Rev Immunol. 2010;10:787–796.
    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. Rodriguez AM, Zeybek B, Vaughn M. Comparison of the long-term impact and clinical outcomes of fewer doses and standard doses of human papillomavirus vaccine in the United States: a database study. Cancer. 2020;126:1656–1667.
    1. Brotherton JM, Budd A, Rompotis C. Is one dose of human papillomavirus vaccine as effective as three?: A national cohort analysis. Papillomavirus Res. 2019;8
    1. Verdoodt F, Dehlendorff C, Kjaer SK. Dose-related effectiveness of quadrivalent human papillomavirus vaccine against cervical intraepithelial neoplasia: a Danish nationwide cohort study. Clin Infect Dis. 2020;70:608–614.
    1. Markowitz LE, Naleway AL, Klein NP. Human papillomavirus vaccine effectiveness against HPV infection: evaluation of one, two, and three doses. J Infect Dis. 2020;221:910–918.
    1. Zeybek B, Lin YL, Kuo YF, Rodriguez AM. The impact of varying numbers of quadrivalent human papillomavirus vaccine doses on anogenital warts in the United States: a database study. J Low Genit Tract Dis. 2018;22:189–194.
    1. Whitworth HS, Gallagher KE, Howard N. Efficacy and immunogenicity of a single dose of human papillomavirus vaccine compared to no vaccination or standard three and two-dose vaccination regimens: a systematic review of evidence from clinical trials. Vaccine. 2020;38:1302–1314.
    1. Shinkafi-Bagudu Z. Global partnerships for HPV vaccine must look beyond national income. JCO Glob Oncol. 2020;6:1746–1748.
    1. Ginsburg O, Basu P, Kapambwe S, Canfell K. Eliminating cervical cancer in the COVID-19 era. Nat Can. 2021;2:133–134.
    1. Prem K, Choi YH, Bénard E. Global impact and cost-effectiveness of one-dose versus two-dose human papillomavirus vaccination schedules: a comparative modelling analysis. medRxiv. 2021 doi: 10.1101/2021.02.08.21251186. published online Feb 8. (preprint).

Source: PubMed

3
Tilaa