1
|
Feyzyab H, Milani A, Agi E, Hashemi M, Bolhassani A. Investigation of the Potency of KALA and REV Cell-Penetrating Peptides for In Vitro/In Vivo Delivery of an HPV Multiepitope DNA Construct. J Pept Sci 2025; 31:e70000. [PMID: 39853698 DOI: 10.1002/psc.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/02/2025] [Accepted: 01/09/2025] [Indexed: 01/26/2025]
Abstract
Developing human papillomavirus (HPV) therapeutic DNA vaccines requires an effective delivery system, such as cell-penetrating peptides (CPPs). In the current study, the multiepitope DNA constructs harboring the immunogenic and conserved epitopes of the L1, L2, and E7 proteins of HPV16/18 (pcDNA-L1-L2-E7 and pEGFP-L1-L2-E7) were delivered using KALA and REV CPPs with different properties in vitro and in vivo. Herein, after confirmation of the REV/DNA and KALA/DNA complexes, their stability was investigated against DNase I and serum protease. Then, their entry into HEK-293T eukaryotic cells was analyzed by qualitative and quantitative methods. Finally, anti-tumor effects of the peptide/DNA complexes were investigated in the C57BL/6 mouse model. Based on the obtained data, the REV/DNA and KALA/DNA complexes at the N/P ratio of 5:1 demonstrated successful penetration into HEK-293T cells. Furthermore, in vivo studies represented that the REV/DNA (survival rate: 75%) and KALA/DNA (survival rate: 50%) complexes provided significant protection against C3 tumors in mice. Indeed, REV CPP exhibited a higher survival rate and lower tumor volume than KALA CPP, 50 days after the C3 challenge. These findings represented the potential of KALA and REV CPPs, especially REV, as promising gene delivery systems for developing HPV therapeutic DNA vaccine candidates.
Collapse
Affiliation(s)
- Haleh Feyzyab
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Alireza Milani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Elnaz Agi
- Blood Diseases Research Center (BDRC), Iranian Comprehensive Hemophilia Care Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
2
|
Kusters JMA, Schim van der Loeff MF, Heijne JCM, King AJ, de Melker HE, Heijman T, Bogaards JA, van Benthem BHB. Changes in Genital Human Papillomavirus (HPV) Prevalence During 12 Years of Girls-Only Bivalent HPV Vaccination: Results From a Biennial Repeated Cross-sectional Study. J Infect Dis 2025; 231:e165-e176. [PMID: 39271142 PMCID: PMC11793022 DOI: 10.1093/infdis/jiae455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Between 2009-2021, bivalent human papillomavirus (HPV) vaccination was offered to girls in the Netherlands. We studied the impact of girls-only HPV vaccination on genital HPV prevalence among young adults. METHODS PASSYON (2009-2021) is a study among sexual health clinic clients aged 16-24 years old. Questionnaires elicited data on demographics, sexual behavior, and HPV vaccination status. Genital samples were analyzed using a PCR-based assay (SPF10-LiPA25). Prevalence trends of 12 high-risk genotypes were assessed as adjusted average annual change (aAAC), estimated using Poisson generalized estimating equations models. The relation between aAAC and phylogenetic distance to HPV-16/18 was assessed by means of regression and rank correlation analysis. Data were collected from 8889 females and 3300 heterosexual males (HMs). RESULTS Among females (irrespective of vaccination status), prevalences of HPV-16/18/31/33/35/45 decreased significantly over time. Increasing trends were observed for HPV-39/52/56. Among both HMs and unvaccinated females (54.3%), HPV-16/18 significantly declined, as did HPV-31 among HMs. Contrastingly, HPV-52/58 increased significantly among HMs and unvaccinated females. The type-specific aAAC correlated well with the phylogenetic distance to HPV-16/18. CONCLUSIONS During 12 years of girls-only bivalent HPV vaccination in the Netherlands, decreasing trends of the vaccine types and cross-protected types were observed among females. Herd protection of vaccine types was observed for HMs and unvaccinated females, and 1 cross-protected type for HMs. Increasing prevalence trends of HPV types with large phylogenetic distance to the vaccine types might indicate type replacement.
Collapse
Affiliation(s)
- Johannes M A Kusters
- Centre for Infectious Diseases Control, National Institute for Public Health and the Environment, Bilthoven
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam University Medical Center
| | - Maarten F Schim van der Loeff
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam University Medical Center
- Department of Infectious Diseases, Public Health Service of Amsterdam
| | - Janneke C M Heijne
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam University Medical Center
- Department of Infectious Diseases, Public Health Service of Amsterdam
| | - Audrey J King
- Centre for Infectious Diseases Control, National Institute for Public Health and the Environment, Bilthoven
| | - Hester E de Melker
- Centre for Infectious Diseases Control, National Institute for Public Health and the Environment, Bilthoven
| | - Titia Heijman
- Department of Infectious Diseases, Public Health Service of Amsterdam
| | - Johannes A Bogaards
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam University Medical Center
- Department of Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Birgit H B van Benthem
- Centre for Infectious Diseases Control, National Institute for Public Health and the Environment, Bilthoven
| |
Collapse
|
3
|
Jiamsiri S, Rhee C, Ahn HS, Seo HW, Klinsupa W, Park S, Lee J, Premsri N, Namwat C, Silaporn P, Excler JL, Kim DR, Chon Y, Sampson JN, Nilyanimit P, Vongpunsawad S, Poudyal N, Markowitz LE, Panicker G, Unger ER, Rerks-Ngarm S, Poovorawan Y, Lynch J. Community intervention of a single-dose or 2-dose regimen of bivalent human papillomavirus vaccine in schoolgirls in Thailand: vaccine effectiveness 2 years and 4 years after vaccination. J Natl Cancer Inst Monogr 2024; 2024:346-357. [PMID: 39529526 PMCID: PMC11555278 DOI: 10.1093/jncimonographs/lgae036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/17/2024] [Accepted: 06/22/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND With accumulating evidence of single-dose human papillomavirus (HPV) vaccine efficacy in young women, we conducted a community vaccine effectiveness study comparing HPV single-dose and 2-dose regimens (0 and 6 months) of a bivalent HPV vaccine among grade 8 schoolgirls (aged 13-14 years) in Thailand. METHODS In 2018, eligible grade 8 schoolgirls in Udon Thani (single dose) and Buri Ram (2 doses) provinces were offered HPV vaccine per assigned dose regimen. Concurrently, a cross-sectional survey for measuring baseline HPV prevalence was conducted in grade 10 (n = 2600) and grade 12 unvaccinated schoolgirls (n = 2000) in each province. HPV infection was assessed in first-void urine samples, tested by DNA polymerase chain reaction on the cobas 4800 system (Roche Molecular Diagnostics, Pleasanton, CA). All samples positive on the cobas system and an equal number of negative samples were also tested by Anyplex II HPV28 Detection (Seegene, Seoul, South Korea). The surveys were repeated in 2020 and 2022, when vaccinated grade 8 schoolgirls reached grade 10, and then subsequently grade 12, respectively. Vaccine effectiveness was estimated by comparing the weighted prevalence of HPV-16 or HPV-18 between grade-matched unvaccinated schoolgirls on the baseline survey (2018) and vaccinated schoolgirls in the year-2 (2020) and year-4 (2022) surveys. Adjustment methods were used in the analysis to account for potential differences in sexual behavior due to the noncontemporaneous comparison. RESULTS The prevalence of HPV-16 and HPV-18 on the baseline survey among unvaccinated grade 10/grade 12 schoolgirls was 2.90% (95% confidence interval [CI] = 2.54% to 3.31%)/3.98% (95% CI = 3.52% to 4.49%) for Udon Thani and 3.87% (95% CI = 3.46% to 4.34%)/6.13% (95% CI = 5.56% to 6.75%) for Buri Ram. On the year-2 survey, the prevalence among vaccinated grade 10 schoolgirls was 0.57% (95% CI = 0.42% to 0.77%) for Udon Thani and 0.31% (95% CI = 0.21% to 0.47%) for Buri Ram. The 2-year postvaccination crude vaccine effectiveness for the single-dose regimen was estimated at 80.4% (95% CI = 73.9% to 86.9%), and for the 2-dose regimen at 91.9% (95% CI = 88.5% to 95.4%). On the year-4 survey, the prevalence among vaccinated grade 12 schoolgirls was 0.37% (95% CI = 0.25% to 0.56%) for Udon Thani and 0.28% (95% CI = 0.18% to 0.45%) for Buri Ram. Four-year postvaccination crude vaccine effectiveness for the single-dose regimen was estimated at 90.6% (95% CI = 86.6% to 94.6%) and for the 2-dose regimen was estimated at 95.4% (95% CI = 93.2% to 97.6%). All adjustment methods minimally affected vaccine effectiveness for the single-dose and 2-dose regimens. At 4 years after vaccination, the difference in crude vaccine effectiveness between the single-dose and 2-dose regimens was ‒4.79% (95% CI = ‒9.32% to ‒0.25%), meeting the study's noninferiority criteria. CONCLUSIONS Our study demonstrated that both single-dose and 2-dose HPV vaccination significantly decreased HPV-16/18 point prevalence 2 years and 4 years after vaccination. Crude vaccine effectiveness at 4 years after vaccination was greater than 90% for both the single-dose and 2-dose regimens; the single-dose regimen was not inferior to the 2-dose regimen. These data show that a single dose of HPV vaccine provides high levels of protection when administered to schoolgirls younger than 15 years of age.
Collapse
Affiliation(s)
- Suchada Jiamsiri
- Department of Disease Control, Ministry of Public Health, Nonthaburi, Thailand
| | - Chulwoo Rhee
- International Vaccine Institute, Seoul, Republic of Korea
| | - Hyeon Seon Ahn
- International Vaccine Institute, Seoul, Republic of Korea
| | - Hyeong-Won Seo
- International Vaccine Institute, Seoul, Republic of Korea
| | - Worrawan Klinsupa
- Department of Disease Control, Ministry of Public Health, Nonthaburi, Thailand
| | - Sunju Park
- International Vaccine Institute, Seoul, Republic of Korea
| | - Jinae Lee
- International Vaccine Institute, Seoul, Republic of Korea
| | | | - Chawetsan Namwat
- Department of Disease Control, Ministry of Public Health, Nonthaburi, Thailand
| | - Patummal Silaporn
- Department of Disease Control, Ministry of Public Health, Nonthaburi, Thailand
| | | | - Deok-Ryun Kim
- International Vaccine Institute, Seoul, Republic of Korea
| | - Yun Chon
- International Vaccine Institute, Seoul, Republic of Korea
| | - Joshua N Sampson
- National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Pornjarim Nilyanimit
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Sompong Vongpunsawad
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nimesh Poudyal
- International Vaccine Institute, Seoul, Republic of Korea
| | | | - Gitika Panicker
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | | | - Yong Poovorawan
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Julia Lynch
- International Vaccine Institute, Seoul, Republic of Korea
| |
Collapse
|
4
|
Tsukamoto K, Yamashita A, Maeki M, Tokeshi M, Imai H, Fukao A, Fujiwara T, Okudera K, Mizuki N, Okuda K, Shimada M. Enhanced Broad-Spectrum Efficacy of an L2-Based mRNA Vaccine Targeting HPV Types 6, 11, 16, 18, with Cross-Protection Against Multiple Additional High-Risk Types. Vaccines (Basel) 2024; 12:1239. [PMID: 39591142 PMCID: PMC11598371 DOI: 10.3390/vaccines12111239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Current L1-based human papillomavirus (HPV) vaccines provide type-specific protection but offer limited cross-protection against non-vaccine HPV types. Therefore, developing a broad-spectrum HPV vaccine is highly desirable. METHODS In this study, we optimized mRNA constructs and developed a multivalent L2-based mRNA vaccine encoding L2 aa 2-130, which includes all known neutralizing epitopes from four prevalent HPV types (HPV-6, -11, -16, and -18). We evaluated its immunogenicity in a mouse model and compared the efficacy of a commercially available mRNA delivery reagent with a custom-synthesized lipid nanoparticle (LNP) formulation. RESULTS We identified that a construct containing E01 (a 5'-untranslated region) and SL2.7 (a poly(A) polymerase recruitment sequence) significantly increased protein expression. The L2-based mRNA vaccine induced robust and long-lasting humoral immune responses, with significant titers of cross-reactive serum IgG antibodies against L2 epitopes. Notably, the vaccine elicited cross-neutralizing antibodies and conferred cross-protective immunity not only against vaccine-targeted HPV types but also against non-vaccine HPV types, following intravaginal challenge in mice. We also found that LNP delivered mRNA more effectively in vivo. CONCLUSIONS The L2-based mRNA vaccine developed in this study shows significant potential for broad-spectrum protection against multiple HPV types. This approach offers a promising strategy for reducing the global burden of HPV-associated cancers.
Collapse
Affiliation(s)
- Kosuke Tsukamoto
- Department of Ophthalmology and Visual Science, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan; (K.T.)
| | - Akio Yamashita
- Department of Investigative Medicine, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishiharacho 903-0215, Japan
| | - Masatoshi Maeki
- Division of Applied Chemistry, Faculty of Engineering, Hokkaido University, Kita 13 Nishi 8, Kita-ku, Sapporo 060-8628, Japan
| | - Manabu Tokeshi
- Division of Applied Chemistry, Faculty of Engineering, Hokkaido University, Kita 13 Nishi 8, Kita-ku, Sapporo 060-8628, Japan
| | - Hirotatsu Imai
- Department of Investigative Medicine, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishiharacho 903-0215, Japan
| | - Akira Fukao
- Department of Biochemistry, Faculty of Pharmacy, Kinki University, Higashiosaka, Osaka 577-8502, Japan
| | - Toshinobu Fujiwara
- Department of Biochemistry, Faculty of Pharmacy, Kinki University, Higashiosaka, Osaka 577-8502, Japan
| | - Koji Okudera
- Department of Pathology, Faculty of Medicine, Saitama Medical University, Saitama 350-0495, Japan
| | - Nobuhisa Mizuki
- Department of Ophthalmology and Visual Science, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan; (K.T.)
| | - Kenji Okuda
- Department of Molecular Biodefense Research, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Masaru Shimada
- Department of Molecular Biodefense Research, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| |
Collapse
|
5
|
Navarro-Torné A, Anderson A, Panwar K, Ghys E, Benninghoff B, Weynants V, Beddows S, Checchi M. How has post-implementation surveillance of high-coverage vaccination with HPV16/18-AS04 vaccine in England added to evidence about its cross-protective effects? Vaccine 2024; 42:126215. [PMID: 39213982 DOI: 10.1016/j.vaccine.2024.126215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 05/08/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Bivalent human papillomavirus HPV16/18-AS04 vaccine (Cervarix, GSK) offers direct protection against HPV16/18. Results from randomised controlled trials showed cross protective effects and suggested that declines in some closely related HPV types could be expected in a population with high vaccination coverage. AIM To evaluate the evidence for cross-protection afforded by HPV16/18-AS04 from post-implementation surveillance in England, and how this complements clinical trial data and post-implementation observations in other countries. METHODS Evidence of cross-protection in young women offered vaccination with HPV16/18-AS04 was gathered from HPV surveillance in England. Data from clinical trials and other post-implementation studies were reviewed. RESULTS Surveillance using anonymised residual specimens in England found declines of 52.3%, 67.4% and 33.3% against grouped HPV-31/33/45 in 16-18, 19-21, and 22-24 year olds, respectively. Additionally, type-specific analysis found that the prevalence of HPV31 declined to below 1% across all age groups. Cross-protection has been monitored and maintained for over 10 years since the introduction of the vaccination programme. Cross-protection against HPV6/11 was not found in English surveillance outcomes. CONCLUSION Surveillance of type-specific infections in vaccine-eligible populations in England has generated clear evidence of cross-protective effects from HPV16/18-AS04 vaccination against high-risk HPV 31/33/45 infections, consistent with other post-implementation observations and confirming and in some ways exceeding expectations from clinical trials.
Collapse
|
6
|
Kusters JMA, van der Loeff MFS, van Benthem BHB, King AJ, de Melker HE, Heijman T, Heijne JCM. Effectiveness of bivalent HPV vaccination against genital HPV DNA-positivity of a catch-up campaign at age 13-16 years compared to routine vaccination at age 12 years: a biennial repeated cross-sectional study. BMC Med 2024; 22:469. [PMID: 39407233 PMCID: PMC11475922 DOI: 10.1186/s12916-024-03686-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND The Netherlands is one of few countries worldwide which has used the bivalent HPV vaccine for girls-only for over a decade. This allows assessment of vaccine effectiveness (VE) against female genital HPV DNA-positivity of this vaccine in an observational post-licencing real-world setting. Additionally, it is unclear whether catch-up vaccination campaigns result in similar VE as routine vaccination. Therefore, type-specific and grouped VE were assessed and compared for women who had been eligible for catch-up vaccination at 13-16 years with those who had been eligible for routine vaccination at 12 years. METHODS PASSYON is a Dutch biennial repeated cross-sectional (2011-2021) study among sexual health clinic clients aged 16-24 years old. Women provided self-collected vaginal samples, questionnaires on demographics and sexual behaviour were administered, and women self-reported HPV vaccination status. Samples were analysed using a PCR-based assay (SPF10-LiPA25). Type-specific and grouped VE estimates, adjusted with propensity score stratification, were assessed against genital positivity for 14 HPV types. VE for targeted and non-targeted genotypes were compared between women who had been eligible for the catch-up and those who had been eligible for routine vaccination. RESULTS The study included 4488 female participants who had been eligible for HPV vaccination and provided genital swabs (1561 eligible for catch-up, 2927 for routine vaccination). Very high VE against genital HPV-16 and HPV-18 was observed (resp. 93.5% and 89.5%) and significant cross-protection against six other genotypes (HPV-31/33/35/45/52/58), varying from 18.0% (HPV-52) to 79.6% (HPV-45). VE estimates were comparable between women who had been eligible for the catch-up campaign and those eligible for routine vaccination: VE HPV-16/HPV-18: 92.2% (95%CI: 87.9-94.9) vs. 91.8% (95%CI: 86.0-95.2). CONCLUSIONS In real-world settings, the VE of bivalent vaccine is high against targeted genotypes, with cross-protection against 6 other genotypes. Catch-up campaigns up to age 16 years can be as effective as routine vaccination at age 12, although it is recommendable to provide HPV vaccination at an age at which most are likely not sexually active yet. This may inform countries considering catch-up campaigns when introducing or extending the use of HPV vaccination within their national immunisation programmes.
Collapse
Affiliation(s)
- Johannes M A Kusters
- Centre for Infectious Diseases Control, National Institute for Public Health and the Environment, Antonie Van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, the Netherlands.
- Institute for Immunology and Infectious Diseases (AII), Amsterdam, UMC , Amsterdam, the Netherlands.
- Amsterdam Public Health Research Institute (APH), Amsterdam, the Netherlands.
| | - Maarten F Schim van der Loeff
- Institute for Immunology and Infectious Diseases (AII), Amsterdam, UMC , Amsterdam, the Netherlands
- Amsterdam Public Health Research Institute (APH), Amsterdam, the Netherlands
- Department of Infectious Diseases, Public Health Service of Amsterdam, Amsterdam, the Netherlands
- Amsterdam UMC, Department of Internal Medicine, Location University of Amsterdam, Amsterdam, the Netherlands
| | - Birgit H B van Benthem
- Centre for Infectious Diseases Control, National Institute for Public Health and the Environment, Antonie Van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, the Netherlands
| | - Audrey J King
- Centre for Infectious Diseases Control, National Institute for Public Health and the Environment, Antonie Van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, the Netherlands
| | - Hester E de Melker
- Centre for Infectious Diseases Control, National Institute for Public Health and the Environment, Antonie Van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, the Netherlands
| | - Titia Heijman
- Department of Infectious Diseases, Public Health Service of Amsterdam, Amsterdam, the Netherlands
| | - Janneke C M Heijne
- Institute for Immunology and Infectious Diseases (AII), Amsterdam, UMC , Amsterdam, the Netherlands
- Amsterdam Public Health Research Institute (APH), Amsterdam, the Netherlands
- Department of Infectious Diseases, Public Health Service of Amsterdam, Amsterdam, the Netherlands
- Amsterdam UMC, Department of Internal Medicine, Location University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
7
|
Liang LA, Tanaka LF, Radde K, Bussas U, Ikenberg H, Heideman DAM, Meijer CJLM, Blettner M, Klug SJ. Population-based age- and type-specific prevalence of human papillomavirus among non-vaccinated women aged 30 years and above in Germany. BMC Infect Dis 2024; 24:1008. [PMID: 39300354 DOI: 10.1186/s12879-024-09827-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 08/28/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND A persisting high-risk human papillomavirus (HR-HPV) infection is causal for cervical cancer; however, there is limited population-based data on the prevalence of HPV infections in Germany. We assessed the age and type-specific HPV prevalence, and associated risk factors in HPV unvaccinated women aged 30 and above. METHODS The MARZY prospective population-based cohort study was conducted between 2005 and 2012 in Mainz and Mainz-Bingen, Germany. Eligible women were randomly recruited from population registries and invited for cervical cancer screening (n = 5,275). A study swab (liquid-based cytology) was taken and HPV testing was performed with GP5+/6 + polymerase chain reaction (PCR) followed by genotyping. We assessed HPV types as HR-HPV, 'moderate' risk and low-risk (LR-HPV). Logistic regression was performed to identify factors associated with HPV infection, stratified by HPV types. RESULTS 2,520 women were screened with a valid PCR result. Overall HPV prevalence was 10.6% (n = 266), with 6.5% HR-HPV positive (n = 165), 1.5% 'moderate' risk type (n = 38) and 3.3% LR-HPV type (n = 84) positive. 8.9% had a single infection (n = 225) and 1.6% had multiple types (n = 41). The most common HR-HPV types were 16, 56, 52 and 31 and LR-HPV 90 and 42. Of 187 HR-HPV infections detected (among 165 women), 55.1% (n = 103) were with HPV types not covered by available bivalent or quadrivalent HPV vaccines. About 23% (n = 43) were of types not covered by the nonavalent vaccine (HPV 35, 39, 51, 56, 59). The HR and LR-HPV prevalence were highest in the age group 30-34 years (HR 9.8%, 'moderate' risk 3.0% and LR 5.6%), decreasing with increasing age. HR-HPV prevalence in women with normal cytology was 5.5%. In women with a high-grade squamous intraepithelial lesion (HSIL), prevalence was 66.7%. Women currently not living with a partner and current smokers had increased chances of an HR-HPV infection. CONCLUSION The overall population-based HPV prevalence was relatively high. An important share of prevalent HR-HPV infections constituted types not covered by current HPV vaccines. With the advent of HPV screening and younger vaccinated cohorts joining screening, HPV types should be monitored closely, also in older women who were not eligible for HPV vaccination.
Collapse
Affiliation(s)
- Linda A Liang
- Chair of Epidemiology, TUM School of Medicine and Health, Technical University of Munich, Georg-Brauchle-Ring 56, 80992, Munich, Germany
| | - Luana F Tanaka
- Chair of Epidemiology, TUM School of Medicine and Health, Technical University of Munich, Georg-Brauchle-Ring 56, 80992, Munich, Germany
- Center for International Health, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Kathrin Radde
- Chair of Epidemiology, TUM School of Medicine and Health, Technical University of Munich, Georg-Brauchle-Ring 56, 80992, Munich, Germany
| | - Ulrike Bussas
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
- German Cancer Research Center, Division of Preventive Oncology, Heidelberg, Germany
| | | | - Daniëlle A M Heideman
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Pathology, Amsterdam, The Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Chris J L M Meijer
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Pathology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Maria Blettner
- Institute for Medical Biostatistics, Epidemiology, and Informatics, University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Stefanie J Klug
- Chair of Epidemiology, TUM School of Medicine and Health, Technical University of Munich, Georg-Brauchle-Ring 56, 80992, Munich, Germany.
| |
Collapse
|
8
|
Castle PE. Looking Back, Moving Forward: Challenges and Opportunities for Global Cervical Cancer Prevention and Control. Viruses 2024; 16:1357. [PMID: 39339834 PMCID: PMC11435674 DOI: 10.3390/v16091357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024] Open
Abstract
Despite the introduction of Pap testing for screening to prevent cervical cancer in the mid-20th century, cervical cancer remains a common cause of cancer-related mortality and morbidity globally. This is primarily due to differences in access to screening and care between low-income and high-income resource settings, resulting in cervical cancer being one of the cancers with the greatest health disparity. The discovery of human papillomavirus (HPV) as the near-obligate viral cause of cervical cancer can revolutionize how it can be prevented: HPV vaccination against infection for prophylaxis and HPV testing-based screening for the detection and treatment of cervical pre-cancers for interception. As a result of this progress, the World Health Organization has championed the elimination of cervical cancer as a global health problem. However, unless research, investments, and actions are taken to ensure equitable global access to these highly effective preventive interventions, there is a real threat to exacerbating the current health inequities in cervical cancer. In this review, the progress to date and the challenges and opportunities for fulfilling the potential of HPV-targeted prevention for global cervical cancer control are discussed.
Collapse
Affiliation(s)
- Philip E Castle
- Divisions of Cancer Prevention and Cancer Epidemiology and Genetics, US National Cancer Institute, National Institutes of Health, 9609 Medical Center Dr., Room 5E410, Rockville, MD 20850, USA
| |
Collapse
|
9
|
Gray P, Mariz FC, Eklund C, Eriksson T, Faust H, Kann H, Müller M, Paavonen J, Pimenoff VN, Sehr P, Surcel HM, Dillner J, Waterboer T, Lehtinen M. Lack of detectable HPV18 antibodies in 14% of quadrivalent vaccinees in a longitudinal cohort study. NPJ Vaccines 2024; 9:146. [PMID: 39138224 PMCID: PMC11322158 DOI: 10.1038/s41541-024-00941-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 07/30/2024] [Indexed: 08/15/2024] Open
Abstract
Although HPV vaccines are highly efficacious, a notable proportion of quadrivalent vaccinees are HPV18 seronegative post-vaccination. We have investigated this findings' validity by comparing vaccine-induced antibody responses using two different immunoassays. 6558 16-17-year-old females participated in the FUTURE II (NCT00092534) and PATRICIA (NCT00122681) trials in 2002-2004. Both the quadrivalent and bivalent vaccine recipients (QVR and BVR) received three doses. Twelve-year follow-up for 648 vaccinees was conducted by the Finnish Maternity Cohort. The presence of neutralising and binding HPV antibodies was analysed via HPV pseudovirion-based neutralisation and pseudovirion-binding assays. Four percent and 14.3% of the QVRs were seronegative for neutralising and binding antibodies to HPV16 and HPV18, respectively. No BVRs were HPV16/18 seronegative post-vaccination. The antibody titres were strongly correlated between the assays, Pearson's correlation coefficient, r[HPV16] = 0.92 and 0.85, and r[HPV18] = 0.91 and 0.86 among the QVRs and BVRs respectively. Fourteen percent of QVRs lacked detectable HPV18 antibodies in long-term follow-up.
Collapse
Affiliation(s)
- Penelope Gray
- Center for Cervical Cancer Elimination, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
| | - Filipe Colaço Mariz
- Tumorvirus-Specific Vaccination Strategies, Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer Feld 242, 69120, Heidelberg, Germany
| | - Carina Eklund
- Center for Cervical Cancer Elimination, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Tiina Eriksson
- Tampere University, Faculty of Medicine and Health Technology, Tampere, Finland
- Wellbeing services county of Pirkanmaa, PIRHA, Tays Research Services, Tampere, Finland
| | - Helena Faust
- Medical Products Agency Läkemedelsverket, Uppsala, Sweden
| | - Hanna Kann
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Martin Müller
- Tumorvirus-Specific Vaccination Strategies, Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer Feld 242, 69120, Heidelberg, Germany
| | - Jorma Paavonen
- Medical Faculty, University of Helsinki, Helsinki, Finland
| | - Ville N Pimenoff
- Center for Cervical Cancer Elimination, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biobank Borealis of Northern Finland, University of Oulu, Oulu, Finland
| | - Peter Sehr
- EMBL-DKFZ Chemical Biology Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Heljä-Marja Surcel
- Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biobank Borealis of Northern Finland, University of Oulu, Oulu, Finland
| | - Joakim Dillner
- Center for Cervical Cancer Elimination, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Tim Waterboer
- Infections and Cancer Epidemiology, Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer Feld 242, 69120, Heidelberg, Germany
| | - Matti Lehtinen
- Center for Cervical Cancer Elimination, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Tampere University, Faculty of Medicine and Health Technology, Tampere, Finland
| |
Collapse
|
10
|
Whitworth HS, Mounier-Jack S, Choi EM, Gallagher KE, Howard N, Kelly H, Mbwanji G, Kreimer AR, Basu P, Barnabas R, Drolet M, Brisson M, Watson-Jones D. Efficacy and immunogenicity of a single dose of human papillomavirus vaccine compared to multidose vaccination regimens or no vaccination: An updated systematic review of evidence from clinical trials. Vaccine X 2024; 19:100486. [PMID: 38873638 PMCID: PMC11169951 DOI: 10.1016/j.jvacx.2024.100486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 06/15/2024] Open
Abstract
Objectives This study systematically reviewed the published literature from clinical trials on the efficacy and immunogenicity of single-dose HPV vaccination compared to multidose schedules or no HPV vaccination. Methods Four databases were searched for relevant articles published from Jan-1999 to Feb-2023. Articles were assessed for eligibility for inclusion using pre-defined criteria. Relevant data were extracted from eligible articles and a descriptive quality assessment was performed for each study. A narrative data synthesis was conducted, examining HPV infection, other clinical outcomes and immunogenicity responses by dose schedule. Results Fifteen articles reporting data from six studies (all in healthy young females) were included. One article was included from each of three studies that prospectively randomised participants to receive a single HPV vaccine dose versus one or more comparator schedule(s). The other 12 articles reported data from three studies that randomised participants to receive multidose HPV vaccine (or control vaccine) schedules; in those studies, some participants failed to complete their allocated schedule, and evaluations were conducted to compare participants who actually received one, two or three doses. Across all efficacy studies, the incidence or prevalence of HPV16/18 infection was very low among HPV-vaccinated participants, regardless of the number of doses received; with no evidence for a difference between dose groups. In immunogenicity studies, HPV16/18 antibody seropositivity rates were high among all HPV-vaccinated participants. Antibody levels were significantly lower with one dose compared to two or three doses, but levels with one dose were stable and sustained to 11 years post-vaccination. Conclusions Results from this review support recent World Health Organization recommendations allowing either one- or two-dose HPV vaccination in healthy young females. Longer-term efficacy and immunogenicity data from ongoing studies are awaited. Randomised trials of single-dose HPV-vaccination are urgently needed in other populations, e.g. boys, older females and people with HIV.
Collapse
Affiliation(s)
- Hilary S. Whitworth
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Sandra Mounier-Jack
- Faculty of Public Health and Policy, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Edward M. Choi
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Katherine E. Gallagher
- Faculty of Public Health and Policy, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Natasha Howard
- Faculty of Public Health and Policy, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Helen Kelly
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Gladys Mbwanji
- Mwanza Intervention Trials Unit, National Institute of Medical Research, Mwanza, Tanzania
| | - Aimée R Kreimer
- National Cancer Institute, National Institute of Health, Bethesda, MD, United States
| | - Partha Basu
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Ruanne Barnabas
- Division of Infectious Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, United States
| | - Mélanie Drolet
- Department of Social and Preventive Medicine, Laval University, Québec, Canada
| | - Marc Brisson
- Department of Social and Preventive Medicine, Laval University, Québec, Canada
| | - Deborah Watson-Jones
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Mwanza Intervention Trials Unit, National Institute of Medical Research, Mwanza, Tanzania
| |
Collapse
|
11
|
McGill F, Fields PJ, Bahadoor-Yetman A, Manglardi ES, Bailey R, Padala K, Lendore J, John-Ballantyne T, Lake S. Investigating under-reported human papillomavirus genotypes in Grenadian women through self-sampling for cervical cancer screening. Rev Panam Salud Publica 2024; 48:e62. [PMID: 39044773 PMCID: PMC11265311 DOI: 10.26633/rpsp.2024.62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/22/2024] [Indexed: 07/25/2024] Open
Abstract
Objective To compare the adequacy, agreement, and acceptability of Papanicolaou testing (cytology) for cervical cancer screening using self-collected samples compared to physician-collected samples in Grenada in the Caribbean. Furthermore, the study identifies the human papillomavirus (HPV) genotypes present among asymptomatic women testing positive for HPV, the etiologic cause of cervical cancer. Methods Participants were divided into two groups and two cervical samples were collected from the women in each group: a self-collected sample and a physician-collected sample. Cervical specimens were tested for cytology and HPV. HPV genotyping was performed on positive specimens. Results Self-collected samples were adequate and in agreement with physician-collected samples, showing no difference between the two sampling methods. Oncogenic high-risk HPV genotypes were identified in cervical samples which were positive for atypical squamous cells and low-grade squamous intraepithelial lesions. The high-risk HPV genotypes found, notably HPV 45 and 53, differed from those most commonly reported. Although the commonly reported high-risk genotypes HPV 16 and 18 were found, so were 31, 33, 35, 52, 66, 68, and 82. Conclusions Using self-collection facilitated the discovery of unexpected HPV genotypes among asymptomatic women in Grenada. These findings add new information to the literature regarding cervical cancer and neoplasia screening and HPV genotypes in the Caribbean. This genotype information may impact surveillance of women with low-grade lesions, HPV vaccine selection, and possibly further vaccine research. Research regarding HPV in Caribbean pathology samples of cervical neoplasia and cancer is needed.
Collapse
Affiliation(s)
- Frances McGill
- St. George’s UniversitySt. George’sGrenadaSt. George’s University, St. George’s, Grenada
| | - Paul J. Fields
- St. George’s UniversitySt. George’sGrenadaSt. George’s University, St. George’s, Grenada
| | - Aví Bahadoor-Yetman
- St. George’s UniversitySt. George’sGrenadaSt. George’s University, St. George’s, Grenada
| | - Erin Salter Manglardi
- St. George’s UniversitySt. George’sGrenadaSt. George’s University, St. George’s, Grenada
| | - Ronelle Bailey
- St. George’s UniversitySt. George’sGrenadaSt. George’s University, St. George’s, Grenada
| | - Kritika Padala
- St. George’s UniversitySt. George’sGrenadaSt. George’s University, St. George’s, Grenada
| | - Jessica Lendore
- St. George’s UniversitySt. George’sGrenadaSt. George’s University, St. George’s, Grenada
| | | | - Sasha Lake
- St. George’s UniversitySt. George’sGrenadaSt. George’s University, St. George’s, Grenada
| |
Collapse
|
12
|
Li Y, Tian S, Ai Y, Hu Z, Ma C, Fu M, Xu Z, Li Y, Liu S, Zou Y, Zhou Y, Jin J. A nanoparticle vaccine displaying varicella-zoster virus gE antigen induces a superior cellular immune response than a licensed vaccine in mice and non-human primates. Front Immunol 2024; 15:1419634. [PMID: 39081325 PMCID: PMC11286566 DOI: 10.3389/fimmu.2024.1419634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Herpes zoster (HZ), also known as shingles, remains a significant global health issue and most commonly seen in elderly individuals with an early exposure history to varicella-zoster virus (VZV). Currently, the licensed vaccine Shingrix, which comprises a recombinant VZV glycoprotein E (gE) formulated with a potent adjuvant AS01B, is the most effective shingles vaccine on the market. However, undesired reactogenicity and increasing global demand causing vaccine shortage, prompting the development of novel shingles vaccines. Here, we developed novel vaccine candidates utilising multiple nanoparticle (NP) platforms to display the recombinant gE antigen, formulated in an MF59-biosimilar adjuvant. In naïve mice, all tested NP vaccines induced higher humoral and cellular immune responses than Shingrix, among which, the gEM candidate induced the highest cellular response. In live attenuated VZV (VZV LAV)-primed mouse and rhesus macaque models, the gEM candidate elicited superior cell-mediated immunity (CMI) over Shingrix. Collectively, we demonstrated that NP technology remains a suitable tool for developing shingles vaccine, and the reported gEM construct is a highly promising candidate in the next-generation shingles vaccine development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jing Jin
- Patronus Biotech Co. Ltd., Guangzhou, China
| |
Collapse
|
13
|
Yang Y, Zhu J, Feng R, Han M, Chen F, Hu Y. Altered vaginal cervical microbiota diversity contributes to HPV-induced cervical cancer via inflammation regulation. PeerJ 2024; 12:e17415. [PMID: 38881859 PMCID: PMC11179633 DOI: 10.7717/peerj.17415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 04/28/2024] [Indexed: 06/18/2024] Open
Abstract
Background Cancer has surpassed infectious diseases and heart ailments, taking the top spot in the disease hierarchy. Cervical cancer is a significant concern for women due to high incidence and mortality rates, linked to the human papillomavirus (HPV). HPV infection leads to precancerous lesions progressing to cervical cancer. The cervix's external os, near the vagina, hosts various microorganisms. Evidence points to the link between vaginal microbiota and HPV-induced cervical cancer. Cervical cancer onset aligns with an imbalanced Th1/Th2 immune response, but the role of vaginal microbiota in modulating this imbalance is unclear. Methods In this study, we collected vaginal samples from 99 HPV-infected patients across varying degrees of lesions, alongside control groups. These samples underwent bacterial DNA sequencing. Additionally, we employed Elisa kits to quantify the protein expression levels of Th1/Th2 cytokines IL2, IL12, IL5, IL13, and TNFa within the centrifuged supernatant of vaginal-cervical secretions from diverse research subjects. Subsequently, correlation analyses were conducted between inflammatory factors and vaginal microbiota. Results Our findings highlighted a correlation between decreased Lactobacillus and increased Gardenerella presence with HPV-induced cervical cancer. Functionally, our predictive analysis revealed the predominant enrichment of the ABC transporter within the vaginal microbiota of cervical cancer patients. Notably, these microbiota alterations exhibited correlations with the production of Th1/Th2 cytokines, which are intimately tied to tumor immunity. Conclusions This study suggests the potential involvement of vaginal microbiota in the progression of HPV-induced cervical cancer through Th1/Th2 cytokine regulation. This novel insight offers a fresh perspective for early cervical cancer diagnosis and future prevention strategies.
Collapse
Affiliation(s)
- Yiheng Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jufan Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Renqian Feng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mengfei Han
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | | | - Yan Hu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
14
|
Shing JZ, Porras C, Pinheiro M, Herrero R, Hildesheim A, Liu D, Gail MH, Romero B, Schiller JT, Zúñiga M, Mishra S, Burdette L, Jones K, Schussler J, Ocampo R, Fang J, Liu Z, Lowy DR, Tsang SH, Rodríguez AC, Schiffman M, Haas CB, Carvajal LJ, Brown JR, Kreimer AR, Mirabello L. Differential long-term bivalent HPV vaccine cross-protection by variants in the Costa Rica HPV vaccine trial. NPJ Vaccines 2024; 9:101. [PMID: 38851816 PMCID: PMC11162434 DOI: 10.1038/s41541-024-00896-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024] Open
Abstract
The AS04-adjuvanted human papillomavirus (HPV)16/18 vaccine, an L1-based vaccine, provides strong vaccine efficacy (VE) against vaccine-targeted type infections, and partial cross-protection to phylogenetically-related types, which may be affected by variant-level heterogeneity. We compared VE against incident HPV31, 33, 35, and 45 detections between lineages and SNPs in the L1 region among 2846 HPV-vaccinated and 5465 HPV-unvaccinated women through 11-years of follow-up in the Costa Rica HPV Vaccine Trial. VE was lower against HPV31-lineage-B (VE=60.7%;95%CI = 23.4%,82.8%) compared to HPV31-lineage-A (VE=94.3%;95%CI = 83.7%,100.0%) (VE-ratio = 0.64;95%CI = 0.25,0.90). Differential VE was observed at several lineage-associated HPV31-L1-SNPs, including a nonsynonymous substitution at position 6372 on the FG-loop, an important neutralization domain. For HPV35, the only SNP-level difference was at position 5939 on the DE-loop, with significant VE against nucleotide-G (VE=65.0%;95%CI = 28.0,87.8) but not for more the common nucleotide-A (VE=7.4%;95%CI = -34.1,36.7). Because of the known heterogeneity in precancer/cancer risk across cross-protected HPV genotype variants by race and region, our results of differential variant-level AS04-adjuvanted HPV16/18 vaccine efficacy has global health implications.
Collapse
Affiliation(s)
- Jaimie Z Shing
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA.
| | - Carolina Porras
- Agencia Costarricense de Investigaciones Biomédicas (ACIB), formerly Proyecto Epidemiológico Guanacaste, Fundación INCIENSA, San José, Costa Rica
| | - Maísa Pinheiro
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Rolando Herrero
- Agencia Costarricense de Investigaciones Biomédicas (ACIB), formerly Proyecto Epidemiológico Guanacaste, Fundación INCIENSA, San José, Costa Rica
| | - Allan Hildesheim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Danping Liu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Mitchell H Gail
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Byron Romero
- Agencia Costarricense de Investigaciones Biomédicas (ACIB), formerly Proyecto Epidemiológico Guanacaste, Fundación INCIENSA, San José, Costa Rica
| | - John T Schiller
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Michael Zúñiga
- Agencia Costarricense de Investigaciones Biomédicas (ACIB), formerly Proyecto Epidemiológico Guanacaste, Fundación INCIENSA, San José, Costa Rica
| | - Sambit Mishra
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
- Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Laurie Burdette
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
- Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Kristine Jones
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
- Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - John Schussler
- Information Management Services Inc, Silver Spring, MD, USA
| | - Rebeca Ocampo
- Agencia Costarricense de Investigaciones Biomédicas (ACIB), formerly Proyecto Epidemiológico Guanacaste, Fundación INCIENSA, San José, Costa Rica
| | - Jianwen Fang
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Zhiwei Liu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Douglas R Lowy
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Sabrina H Tsang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | | | - Mark Schiffman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Cameron B Haas
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Loretto J Carvajal
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
- Agencia Costarricense de Investigaciones Biomédicas (ACIB), formerly Proyecto Epidemiológico Guanacaste, Fundación INCIENSA, San José, Costa Rica
| | - Jalen R Brown
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Aimée R Kreimer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Lisa Mirabello
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| |
Collapse
|
15
|
Murahwa AT, Mudzviti T, Mandishora RSD, Chatindo T, Chanetsa P, Pascoe M, Shamu T, Basera W, Luethy R, Williamson AL. Vaccine and Non-Vaccine HPV Types Presence in Adolescents with Vertically Acquired HIV Five Years Post Gardasil Quadrivalent Vaccination: The ZIMGARD Cohort. Viruses 2024; 16:162. [PMID: 38275972 PMCID: PMC10818519 DOI: 10.3390/v16010162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 12/26/2023] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Human papillomavirus (HPV) vaccination programs are a key intervention in protecting individuals against HPV-related disease. HIV1-infected individuals are at increased risk of HPV-associated cancers. This study was conducted to evaluate the potential role of prophylactic HPV vaccines in preventing new HPV infections among participants with perinatally acquired HIV who received the quadrivalent HPV vaccine at least five years before this study. METHODS This cross-sectional study was conducted at Newlands Clinic, Harare, Zimbabwe. The clinic provided the Gardasil quadrivalent HPV vaccine (4vHPV) to 624 adolescents living with HIV starting in December 2015. Vaginal and penile swabs were collected and tested for HPV types from the study participants who had received the 4vHPV vaccine 5-6 years before enrolment. RESULTS We present the results of 98 participants (44.6% female) vaccinated at a median age of 15 years (IQR 12-16). The mean amount of time since vaccination was 6 years (SD: ±0.4). The HPV-positive rate amongst the analyzed swabs was 69% (68/98). Among 30/98 (31%) HPV-positive participants, 13/98 (13%) had low-risk HPV types, and 17/98 (17%) had high-risk HPV types. Twelve participants tested positive for HPV18, only one participant tested positive for HPV16, and an additional four (4.3%) tested positive for either type 6 or 11, with respect to vaccine-preventable low-risk HPV types. CONCLUSION The Gardasil quadrivalent HPV vaccine (4vHPV) was expected to protect against infection with HPV types 16, 18, 6, and 11. We demonstrated a possible waning of immunity to HPV18 in 17% of the participants, and an associated loss in cross-protection against HPV45. We observed a relatively high prevalence of 'opportunistic non-vaccine HPV types' or 'ecological niche occupiers' in this cohort, and suggest further research on the involvement of these types in cervical and other genital cancers. Our study is one of the few, if not the first, to report on HPV vaccine immunoprotection among people living with HIV (PLWH), thereby setting a baseline for further studies on HPV vaccine effectiveness among PLWH.
Collapse
Affiliation(s)
- Alltalents T. Murahwa
- Institute of Infectious Disease and Molecular Medicine, Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa;
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Tinashe Mudzviti
- Newlands Clinic, Harare P.O. Box A178, Zimbabwe (T.C.); (P.C.); (M.P.); (T.S.); (R.L.)
- Department of Pharmacy and Pharmaceutical Sciences, University of Zimbabwe, Harare P.O. Box AI78, Zimbabwe
| | - Racheal S. Dube Mandishora
- Medical Microbiology Unit, Department of Laboratory Diagnostic and Investigative Sciences, University of Zimbabwe, Harare P.O. Box AI78, Zimbabwe;
- Center for Immunization and Infection Research in Cancer, Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, 33612 FL, USA
| | - Takudzwa Chatindo
- Newlands Clinic, Harare P.O. Box A178, Zimbabwe (T.C.); (P.C.); (M.P.); (T.S.); (R.L.)
| | - Peace Chanetsa
- Newlands Clinic, Harare P.O. Box A178, Zimbabwe (T.C.); (P.C.); (M.P.); (T.S.); (R.L.)
| | - Margaret Pascoe
- Newlands Clinic, Harare P.O. Box A178, Zimbabwe (T.C.); (P.C.); (M.P.); (T.S.); (R.L.)
| | - Tinei Shamu
- Newlands Clinic, Harare P.O. Box A178, Zimbabwe (T.C.); (P.C.); (M.P.); (T.S.); (R.L.)
- Institute of Social and Preventive Medicine, University of Bern, 3012 Bern, Switzerland
- Graduate School of Health Sciences, University of Bern, 3012 Bern, Switzerland
| | - Wisdom Basera
- Burden of Disease Research Unit, South African Medical Research Council, Cape Town 7925, South Africa;
| | - Ruedi Luethy
- Newlands Clinic, Harare P.O. Box A178, Zimbabwe (T.C.); (P.C.); (M.P.); (T.S.); (R.L.)
| | - Anna-Lise Williamson
- Institute of Infectious Disease and Molecular Medicine, Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa;
- SAMRC Gynaecological Cancer Research Centre, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| |
Collapse
|
16
|
Seyoum A, Seyoum B, Gure T, Alemu A, Alemayehu DH, Alemu A, Belachew A, Tefera DA, Aseffa A, Howe R, Mulu A, Mihret A. High rate of non-vaccine targeted high-risk HPV genotypes circulate among women in Eastern Ethiopia. Sci Rep 2024; 14:958. [PMID: 38200092 PMCID: PMC10781741 DOI: 10.1038/s41598-024-51594-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 01/07/2024] [Indexed: 01/12/2024] Open
Abstract
The World Health Organization [WHO] recommends a genotype-specific human papillomavirus [HPV] vaccination as a primary prevention strategy to control the burden of cervical cancer globally. In Ethiopia, where the non-vaccine-targeted HPV genotypes have not been adequately studied, a vaccination initiative was launched in 2018 targeting HPV-6,-11, -16, and -18 for girls aged 14-18 years. The co-existence of both vaccine-targeted and non-targeted genotypes is a serious concern, as it can accelerate cancer progression. Therefore, this study was conducted to determine the prevalence of non-vaccine-targeted HPV genotypes and assess the level of multiple infections with other genotypes in eastern Ethiopia. A health facility-based cross-sectional study including 110 women with positive HPV DNA results was conducted from April to August 2021. A structured questionnaire to collect demographic and clinical data was used. Cervical swabs were collected using L-shaped FLOQSwabs. Women's cytological profile was determined based on Pap smear test results. An automated nucleic acid extraction system using STARMag 96 ProPrep Universal Extraction Kit was utilized following the manufacturer's protocol. An amplification assay in real-time was employed to amplify and identify the HPV Late 1 [L1] gene, which is utilized for genotyping purposes. Following this, the collected data was entered into Epi data version 3.1 software, and the analysis was performed using STATA version 14. A total of 110 women [age range 30-60 years, mean age = 36.5 years and SD ± 6.9] had positive HPV DNA results and were included in the study. Among these, 108 women had valid co-testing [Pap test and HPV DNA test] results for further analysis, and the results of the remaining 2 women were rejected. Overall, the prevalence of non-vaccine-targeted HPV was 56 (51.8%, 95%CI [0.42, 0.61]), of which 28 women (25.4%, 95%CI [0.18, 0.34]) had a single non-vaccine HPV genotype infection. The remaining 29 women (26.4%, 95% CI: 0.190-0.355) experienced multiple infections. The non-vaccine-targeted genotypes of HPV-35 accounted for 11 cases (10%, 95%CI [0.06, 0.17]), HPV-68 was detected in 9 women (8.2%, 95%CI [0.04, 0.15]), HPV-56 and HPV-66 were both found in 8 cases each (7.3%, 95%CI [0.04, 0.14]) of the total. In addition, out of these 108 women, 93 (86.1%, 95%CI [0.78, 0.91]) had low-grade squamous intraepithelial lesions, 13 (12%, 95%CI [0.07, 0.20]) no intraepithelial lesion or malignancy, and two (1.9%, 95%CI [0.01, 0.07]) high-grade squamous intraepithelial lesions. Furthermore, there was no statistical difference [p = 0.755] between vaccine-targeted and non-vaccine-targeted genotypes as the primary cause of cervical lesions. In conclusion, the findings of the present study highlight the existence of a notable prevalence of multiple infections caused by non-vaccine-targeted HPV genotypes. Therefore, it is recommended that both the Federal and regional health bureaus to evaluate the range of hr HPV genotypes protected by the current HPV vaccine and explore the option of transitioning from the quadrivalent HPV vaccine to a novavalent vaccine that includes seven high-risk HPV genotypes.
Collapse
Affiliation(s)
- Ayichew Seyoum
- College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia.
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia.
| | - Berhanu Seyoum
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Tadesse Gure
- College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Ashenafi Alemu
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | | | - Addisu Alemu
- College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Anteneh Belachew
- College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | | | - Abraham Aseffa
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Rawleigh Howe
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | | | - Adane Mihret
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| |
Collapse
|
17
|
Qu W, Sui L, Li Y. Vaccine escape challenges virus prevention: The example of two vaccine-preventable oncogenic viruses. J Med Virol 2023; 95:e29184. [PMID: 37943176 DOI: 10.1002/jmv.29184] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/06/2023] [Accepted: 10/09/2023] [Indexed: 11/10/2023]
Abstract
Over the years, the pace of developing vaccines for HBV and HPV has never stopped. After more than 30 years of application, the HBV vaccine has reduced 80% of hepatocellular carcinoma (HCC). However, vaccine escape variants occur under selective pressure induced by widespread vaccination and antiviral therapy, which results in fulminant infection and horizontal transmission. Several mechanisms have been studied to explain HBV vaccine escape, including vaccine escape mutations (VEMs) in the major hydrophilic region, which leads to a decrease in the binding ability to neutralize antibodies and is the primary escape mechanism, protein conformational and N-linked glycosylation sites changes caused by VEMs, differences in genotype distribution, gene recombination, and some temporarily unknown reasons. However, effective solutions are still being explored. The HPV vaccine has also been proven to prevent 70%-90% of cervical cancer worldwide. Cases of HPV infection after being vaccinated have been observed in clinical practice. However, few researchers have paid attention to the mechanism of HPV vaccine escape. Thus, we reviewed the literature on vaccine escape of both HBV and HPV to discuss the mechanism of the virus escaping from vaccine protection and possible solutions to this problem. We analyzed the gap between studies of HPV and HBV and made prospects for further research in HPV vaccine escape.
Collapse
Affiliation(s)
- Wenjie Qu
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Long Sui
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yanyun Li
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
18
|
Hu YM, Bi ZF, Zheng Y, Zhang L, Zheng FZ, Chu K, Li YF, Chen Q, Quan JL, Hu XW, Huang XC, Zhu KX, Wang-Jiang YH, Jiang HM, Zang X, Liu DL, Yang CL, Pan HX, Zhang QF, Su YY, Huang SJ, Sun G, Huang WJ, Huang Y, Wu T, Zhang J, Xia NS. Immunogenicity and safety of an Escherichia coli-produced human papillomavirus (types 6/11/16/18/31/33/45/52/58) L1 virus-like-particle vaccine: a phase 2 double-blind, randomized, controlled trial. Sci Bull (Beijing) 2023; 68:2448-2455. [PMID: 37743201 DOI: 10.1016/j.scib.2023.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/23/2023] [Accepted: 09/14/2023] [Indexed: 09/26/2023]
Abstract
The Escherichia coli-produced human papillomavirus (HPV) 16/18 bivalent vaccine (Cecolin) has received prequalification by the World Health Organization based on its high efficacy and good safety profile. We aimed to evaluate the immunogenicity and safety of the second-generation nonavalent HPV 6/11/16/18/31/33/45/52/58 vaccine (Cecolin 9) through the randomized, blinded phase 2 clinical trial. Eligible healthy women aged 18-45 years were randomly (1:1) allocated to receive three doses of 1.0 mL (270 µg) of Cecolin 9 or placebo with a 0-1-6-month schedule. The primary endpoint was the seroconversion rate and geometric mean titer of neutralizing antibodies (nAbs) one month after the full vaccination course (month 7). The secondary endpoint was the safety profile including solicited adverse reactions occurring within 7 d, adverse events (AEs) occurring within 30 d after each dose, and serious adverse events (SAEs) occurring during the 7-month follow-up period. In total, 627 volunteers were enrolled and randomly assigned to Cecolin 9 (n = 313) or placebo (n = 314) group in Jiangsu Province, China. Almost all participants in the per-protocol set for immunogenicity (PPS-I) seroconverted for nAbs against all the nine HPV types at month 7, while two failed to seroconvert for HPV 11 and one did not seroconvert for HPV 52. The incidence rates of total AEs in the Cecolin 9 and placebo groups were 80.8% and 72.9%, respectively, with the majority of them being mild and recovering shortly. None of the SAEs were considered related to vaccination. In conclusion, the E. coli-produced 9-valent HPV (9vHPV) vaccine candidate was well tolerated and immunogenic, which warrants further efficacy studies in larger populations.
Collapse
Affiliation(s)
- Yue-Mei Hu
- Jiangsu Provincial Center for Disease Control and Prevention, Public Health Research Institute of Jiangsu Province, Nanjing 210009, China
| | - Zhao-Feng Bi
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Ya Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Li Zhang
- National Institutes for Food and Drug Control, Beijing 102629, China
| | | | - Kai Chu
- Jiangsu Provincial Center for Disease Control and Prevention, Public Health Research Institute of Jiangsu Province, Nanjing 210009, China
| | - Ya-Fei Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Qi Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Jia-Li Quan
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Xiao-Wen Hu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Xing-Cheng Huang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Kong-Xin Zhu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Ya-Hui Wang-Jiang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Han-Min Jiang
- Dongtai Center for Disease Control and Prevention, Yancheng 224200, China
| | - Xia Zang
- Dongtai Center for Disease Control and Prevention, Yancheng 224200, China
| | - Dong-Lin Liu
- Dongtai Center for Disease Control and Prevention, Yancheng 224200, China
| | - Chang-Lin Yang
- Dongtai Center for Disease Control and Prevention, Yancheng 224200, China
| | - Hong-Xing Pan
- Jiangsu Provincial Center for Disease Control and Prevention, Public Health Research Institute of Jiangsu Province, Nanjing 210009, China
| | - Qiu-Fen Zhang
- Xiamen Innovax Biotech Company, Xiamen 361027, China
| | - Ying-Ying Su
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Shou-Jie Huang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Guang Sun
- Xiamen Innovax Biotech Company, Xiamen 361027, China.
| | - Wei-Jin Huang
- National Institutes for Food and Drug Control, Beijing 102629, China.
| | - Yue Huang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China.
| | - Ting Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China.
| | - Jun Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Ning-Shao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| |
Collapse
|
19
|
Han L, Zhang B. Can prophylactic HPV vaccination reduce the recurrence of cervical lesions after surgery? Review and prospect. Infect Agent Cancer 2023; 18:66. [PMID: 37898754 PMCID: PMC10613367 DOI: 10.1186/s13027-023-00547-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 10/18/2023] [Indexed: 10/30/2023] Open
Abstract
Women with HSIL typically undergo conization/LEEP to remove cervical lesions, but the risk of HSIL lesions returning after surgical treatment remains higher than in the general population. HPV vaccination is essential to prevent cervical cancer. However, the effect of prophylactic HPV vaccination on reducing the risk of recurrent cervical lesions after surgical treatment remains unclear. This review aims to analyze and summarize the latest literature on the role of prophylactic HPV vaccine in reducing the recurrence of cervical lesions after surgery in patients with HSIL, and to review and update the history, efficacy, effectiveness and safety of HPV vaccine, focusing on the current status of global HPV vaccine implementation and obstacles.
Collapse
Affiliation(s)
- Ling Han
- Department of Obstetrics and Gynecology, The First College of Clinical Medical Science, China Three Gorges University, Yichang City, Hubei Province, People's Republic of China
- Department of Obstetrics and Gynecology, Yichang Central People's Hospital, Yichang City, Hubei Province, People's Republic of China
| | - Bingyi Zhang
- Department of Ultrasound Imaging, The First College of Clinical Medical Science,, China Three Gorges University, Jiefang Road 2, Yichang City, 443003, Hubei Province, People's Republic of China.
- Department of Ultrasound Imaging, Yichang Central People's Hospital, Jiefang Road 2, Yichang City, 443003, Hubei Province, People's Republic of China.
| |
Collapse
|
20
|
van Eer K, Middeldorp M, Dzebisasjvili T, Lamkaraf N, de Melker HE, Steenbergen RDM, King AJ. Effects of 2 and 3 Vaccinations With the Bivalent Human Papillomavirus (HPV) Vaccine on the Prevalence and Load of HPV in Clearing and Persistent Infections in Young Women. J Infect Dis 2023; 228:1012-1022. [PMID: 36988110 DOI: 10.1093/infdis/jiad080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/16/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Human papillomavirus (HPV) viral load (VL) is associated with persistence, which increases cervical cancer risk. The bivalent vaccine protects against oncogenic HPV-16/18 and cross-protects against several nonvaccine types. We examined the effect of 2-dose (2D) and 3-dose (3D) vaccination on HPV prevalence and VL in clearing infections and persistent infections, 6 years and 12 years postvaccination, respectively. METHODS Vaginal swabs collected from the "HPV Amongst Vaccinated and Non-vaccinated Adolescents" study (HAVANA, 3D-eligible) and HAVANA-2 (2D-eligble) participants were genotyped for HPV with the SPF10-DEIA-LiPA25 system. HPV VL was measured with type-specific quantitative polymerase chain reaction (qPCR). RESULTS HPV-16, -18, -31, -33, and -45 clearing and/or persistent infection prevalence and HPV-16, -18, and -31 VLs in clearing infections were significantly reduced in 3D-vaccinated women compared to unvaccinated women. Except for HPV-11 and -59 clearing infections, no significant VL differences were observed among vaccinated women, ≤6 and >6 years post-vaccination. Infection numbers were low in 2D-eligible women, with no HPV-16/18 in vaccinated women. No VL differences for the remaining types were found. CONCLUSIONS 3D vaccination reduces HPV prevalence in clearing infections and persistent infections and decreases HPV VLs in clearing infections, 12 years post-vaccination for vaccine and several nonvaccine types. 2D-eligible women had low infection numbers, with no HPV-16/18 among vaccinated women.
Collapse
Affiliation(s)
- Kahren van Eer
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, The Netherlands
| | - Marit Middeldorp
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, The Netherlands
| | - Tsira Dzebisasjvili
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, The Netherlands
| | - Najima Lamkaraf
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, The Netherlands
| | - Hester E de Melker
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, The Netherlands
| | - Renske D M Steenbergen
- Pathology, Amsterdam University Medical Center location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Audrey J King
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, The Netherlands
| |
Collapse
|
21
|
Angeletti A, Lugani F, La Porta E, Verrina E, Caridi G, Ghiggeri GM. Vaccines and nephrotic syndrome: efficacy and safety. Pediatr Nephrol 2023; 38:2915-2928. [PMID: 36512075 PMCID: PMC9745735 DOI: 10.1007/s00467-022-05835-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/09/2022] [Accepted: 11/14/2022] [Indexed: 12/15/2022]
Abstract
Vaccines represent the most important medical evolution in the last two centuries allowing prevention and formally eradication of a wide number of infectious diseases. Safety and effectiveness are main issues that still require an open discussion. A few clinical reports described a critical temporal relationship between vaccination and acute nephrotic syndrome, indirectly suggesting an association. For this review, the literature was reviewed to identify articles reporting associations of nephrotic syndrome with vaccines against a vast array of infectious diseases (including bacteria, virus and Sars-Cov-2). As specific aims, we evaluated effectiveness and safety in terms of occurrence of either "de novo" nephrotic syndrome in health subjects or "relapse" in those already affected by the disease. In total, 377 articles were found; 166 duplicates and 71 non-full text, animal studies or non-English language were removed. After excluding another 50 articles not containing relevant data on generic side effects or on relapses or new onset nephrotic syndrome, 90 articles met the search criteria. Overall, studies reported the effect of vaccines in 1015 patients, plus 4 nationwide epidemiologic investigations. Limited experience on vaccination of NS patients with measles, mumps, and rubella live attenuated vaccines does not allow any definitive conclusion on their safeness. VZV has been administered more frequently without side effects. Vaccines utilizing virus inactivated, recombinant, and toxoid can be utilized without risks in NS. Vaccines for influenza reduce the risk of infections during the pandemic and are associated with reduced risk of relapse of NS typically induced by the infection. Vaccines for SARS-CoV-2 (all kinds) offer a concrete approach to reduce the pandemic. "De novo" NS or recurrence are very rare and respond to common therapies.
Collapse
Affiliation(s)
- Andrea Angeletti
- Division of Nephrology, Dialysis and Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy.
| | - Francesca Lugani
- Division of Nephrology, Dialysis and Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Edoardo La Porta
- Division of Nephrology, Dialysis and Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Enrico Verrina
- Division of Nephrology, Dialysis and Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Gianluca Caridi
- Division of Nephrology, Dialysis and Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Gian Marco Ghiggeri
- Division of Nephrology, Dialysis and Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
22
|
Luttjeboer J, Simons J, Westra T, Wilschut J, Boersma C, Postma M, van der Schans J. An Updated Analysis of the Impact of HPV Vaccination Based on Long-term Effectiveness in the Netherlands. Infect Dis Ther 2023; 12:2135-2145. [PMID: 37563438 PMCID: PMC10505117 DOI: 10.1007/s40121-023-00851-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/18/2023] [Indexed: 08/12/2023] Open
Abstract
INTRODUCTION Vaccination against human papillomavirus (HPV) is considered the most effective strategy to protect women from cervical cancer. Three HPV vaccines are currently licensed in Europe and, although they are generally supported by favorable health economic outcomes, current models fall short in predicting vaccination benefits. Here, we aim to re-evaluate the health benefits of HPV vaccination, using updated long-term effectiveness data and emphasizing quality of life losses related to pre-cancer disease and treatment. METHODS We used a static Markov model that compared "only screening" (includes unvaccinated girls) and "vaccination" (assumes 100% vaccination coverage with the bivalent HPV vaccine). A lifetime cohort of 100,000 uninfected 12-year-old girls was included, in which the number of cases with cervical intraepithelial neoplasia grade 2 or higher/3 (CIN2+, CIN3), cervical cancer, and cervical cancer deaths per scenario were determined. Furthermore, the reduction in major excisional procedures, the preterm deliveries averted, and the related gain in quality-adjusted life years (QALYs) due to vaccination were estimated. RESULTS The bivalent vaccine showed larger reductions in CIN2+, CIN3, cervical cancer cases, cervical cancer deaths, and major excisional treatments, after including long-term efficacy and effectiveness data, compared to previous data. Moreover, we observed an increased amount of QALYs gained due to prevention of major excisional treatment and the negative side effects related to it. CONCLUSIONS Updated health economic models for HPV vaccination, using updated and long-term effectiveness data and including prevention of treatment-related side effects, demonstrate a substantial additional positive effect on vaccination outcomes. Indeed, extrapolation of the bivalent HPV vaccine's updated long-term effectiveness data against HPV-related cervical diseases shows that the positive effects of vaccination may be more substantial than previously estimated. There is a graphical abstract available for this article.
Collapse
Affiliation(s)
- Jos Luttjeboer
- University Medical Center, Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Joost Simons
- University Medical Center, Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands.
- GSK, Wavre, Belgium.
| | | | - Jan Wilschut
- University Medical Center, Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Cornelis Boersma
- Open University, Valkenburgerweg 177, 6419AT, Heerlen, The Netherlands
| | - Maarten Postma
- University Medical Center, Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Jurjen van der Schans
- University Medical Center, Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| |
Collapse
|
23
|
Bogani G, Ghelardi A, Sopracordevole F, Annoni M, Ciavattini A, Giannella L, De Vincenzo R, Cattani P, Barbero M, Vercellini P, Raspagliesi F, Bonanni P, Scambia G. Human papillomavirus (HPV) vaccination: a call for action in Italy. Int J Gynecol Cancer 2023; 33:1132-1139. [PMID: 36918227 DOI: 10.1136/ijgc-2023-004275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Human papillomavirus (HPV) is the most common sexually transmitted infection. The implementation of primary prevention aims to reduce the burden of HPV infection and HPV-related disease. However, HPV-related diseases are still a concern, even in high-income countries. Approximately 570 000 new cervical cancer cases are diagnosed in Italy every year. Prophylactic HPV vaccines have been developed to minimize the spread of HPV. Growing evidence supports the administration of HPV vaccines (even just one dose) in reducing the prevalence of HPV infection and HPV-related disease including cancers. HPV vaccines are characterized by a high level of efficacy (>95%) in women who are naïve to HPV; however, they do not increase clearance in patients with ongoing HPV infection. With more than 200 million doses administered to date, HPV vaccines are considered to be safe and effective at preventing HPV-related infections and cancers. In this review we aim to review the current evidence regarding HPV vaccination and to describe trends in HPV vaccination coverage in Italy. In Italy, vaccination against HPV has been included in the National Immunization Plan (NIP) since 2007-2008. Using data abstracted from the Italian Ministry of Health, we analyzed changes in HPV vaccination coverage. We observed that HPV vaccines are underutilized and coverage rates are decreasing. Looking at the target population (females and males aged 11-12 years) in Italy, a decrease in coverage rates was observed. A call for action, improved HPV awareness, and education are the key elements to enhance the widespread adoption of HPV vaccination.
Collapse
Affiliation(s)
- Giorgio Bogani
- Department of Gynecologic Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | | | - Francesco Sopracordevole
- Gynecological Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Marco Annoni
- National Research Council of Italy, Roma, Italy
- Fondazione Umberto Veronesi, Milano, Italy
| | - Andrea Ciavattini
- Woman's Health Sciences Department, Polytechnic University of Marche, Ancona, Italy
| | - Luca Giannella
- Woman's Health Sciences Department, Polytechnic University of Marche, Ancona, Italy
| | - Rosa De Vincenzo
- Gynecologic Oncology, Catholic University of the Sacred Heart, Roma, Italy
| | - Paolo Cattani
- Italian Society of Colposcopy and Cervico-Vaginal Pathology, Roma, Italy
| | - Maggiorino Barbero
- Department of Obstetrics and Gynecology, Azienda Sanitaria Locale di Asti, Asti, Italy
| | - Paolo Vercellini
- Department of Obstetrics and Gynecology, Policlinico of Milan Mangiagalli Center, Milano, Italy
| | | | - Paolo Bonanni
- Department of Obstetrics and Gynecology, Università degli Studi di Firenze, Firenze, Italy
| | - Giovanni Scambia
- Dipartimento Scienze della Salute della Donna e del Bambino, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| |
Collapse
|
24
|
Mwenda V, Jalang'o R, Miano C, Bor JP, Nyangasi M, Mecca L, Were V, Kariithi E, Pecenka C, Schuind A, Abbas K, Clark A. Impact, cost-effectiveness, and budget implications of HPV vaccination in Kenya: A modelling study. Vaccine 2023:S0264-410X(23)00546-7. [PMID: 37296015 DOI: 10.1016/j.vaccine.2023.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND Sub-Saharan Africa has the highest rate of cervical cancer cases and deaths worldwide. Kenya introduced a quadrivalent HPV vaccine (GARDASIL, hereafter referred to as GARDASIL-4) for ten-year-old girls in late 2019 with donor support from Gavi, the Vaccine Alliance. As Kenya may soon graduate from Gavi support, it is important to evaluate the potential cost-effectiveness and budget impact of the current HPV vaccine, and potential alternatives. METHODS We used a proportionate outcomes static cohort model to evaluate the annual budget impact and lifetime cost-effectiveness of vaccinating ten-year-old girls over the period 2020-2029. We included a catch-up campaign for girls aged 11-14 years in 2020. We estimated cervical cancer cases, deaths, disability adjusted life years (DALYs), and healthcare costs (government and societal perspective) expected to occur with and without vaccination over the lifetimes of each cohort of vaccinated girls. For each of the four products available globally (CECOLIN©, CERVARIX©, GARDASIL-4©, and GARDASIL-9 ©), we estimated the cost (2021 US$) per DALY averted compared to no vaccine and to each other. Model inputs were obtained from published sources, as well as local stakeholders. RESULTS We estimated 320,000 cases and 225,000 deaths attributed to cervical cancer over the lifetimes of the 14 evaluated birth cohorts. HPV vaccination could reduce this burden by 42-60 %. Without cross-protection, CECOLIN had the lowest net cost and most attractive cost-effectiveness. With cross-protection, CERVARIX was the most cost-effective. Under either scenario the most cost-effective vaccine had a 100 % probability of being cost-effective at a willingness-to-pay threshold of US$ 100 (5 % of Kenya's national gross domestic product per capita) compared to no vaccination. Should Kenya reach its target of 90 % coverage and graduate from Gavi support, the undiscounted annual vaccine program cost could exceed US$ 10 million per year. For all three vaccines currently supported by Gavi, a single-dose strategy would be cost-saving compared to no vaccination. CONCLUSION HPV vaccination for girls is highly cost-effective in Kenya. Compared to GARDASIL-4, alternative products could provide similar or greater health benefits at lower net costs. Substantial government funding will be required to reach and sustain coverage targets as Kenya graduates from Gavi support. A single dose strategy is likely to have similar benefits for less cost.
Collapse
Affiliation(s)
- Valerian Mwenda
- National Cancer Control Program, Ministry of Health, Nairobi, Kenya.
| | - Rose Jalang'o
- National Vaccines and Immunization Program, Ministry of Health, Nairobi, Kenya
| | - Christine Miano
- National Vaccines and Immunization Program, Ministry of Health, Nairobi, Kenya
| | - Joan-Paula Bor
- National Cancer Control Program, Ministry of Health, Nairobi, Kenya
| | - Mary Nyangasi
- National Cancer Control Program, Ministry of Health, Nairobi, Kenya
| | - Lucy Mecca
- National Vaccines and Immunization Program, Ministry of Health, Nairobi, Kenya
| | - Vincent Were
- Kenya Medical Research Institute, Nairobi, Kenya
| | | | | | | | - Kaja Abbas
- London School of Hygiene and Tropical Medicine, London, UK
| | - Andrew Clark
- London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
25
|
Kheirvari M, Liu H, Tumban E. Virus-like Particle Vaccines and Platforms for Vaccine Development. Viruses 2023; 15:1109. [PMID: 37243195 PMCID: PMC10223759 DOI: 10.3390/v15051109] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Virus-like particles (VLPs) have gained a lot of interest within the past two decades. The use of VLP-based vaccines to protect against three infectious agents-hepatitis B virus, human papillomavirus, and hepatitis E virus-has been approved; they are very efficacious and offer long-lasting immune responses. Besides these, VLPs from other viral infectious agents (that infect humans, animals, plants, and bacteria) are under development. These VLPs, especially those from human and animal viruses, serve as stand-alone vaccines to protect against viruses from which the VLPs were derived. Additionally, VLPs, including those derived from plant and bacterial viruses, serve as platforms upon which to display foreign peptide antigens from other infectious agents or metabolic diseases such as cancer, i.e., they can be used to develop chimeric VLPs. The goal of chimeric VLPs is to enhance the immunogenicity of foreign peptides displayed on VLPs and not necessarily the platforms. This review provides a summary of VLP vaccines for human and veterinary use that have been approved and those that are under development. Furthermore, this review summarizes chimeric VLP vaccines that have been developed and tested in pre-clinical studies. Finally, the review concludes with a snapshot of the advantages of VLP-based vaccines such as hybrid/mosaic VLPs over conventional vaccine approaches such as live-attenuated and inactivated vaccines.
Collapse
Affiliation(s)
| | | | - Ebenezer Tumban
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX 79106, USA
| |
Collapse
|
26
|
Padavu S, Kumar BK, Kumar A, Rai P. In-silico Analysis of Human Papillomavirus – 45 E6, E7 & L1 Proteins as Potential Immunogens. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2023; 17:554-566. [DOI: 10.22207/jpam.17.1.53] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
Abstract
Globally, cervical cancer is the fourth most common cancer among women. After being cloned from a recurring cervical lesion in 1987, Human papillomavirus (HPV) type-45 was identified as a high-risk HPV type. It is the third most common cancer-causing HPV subtype, after HPV-16 and HPV-18. Immunogenic epitopes and structural features provide the most useful information for vaccine development. Computational algorithms provide quick, simple, trustworthy, and cost-efficient methods for predicting immunogenic epitopes. In this study, both B and T cell epitopes have been identified as potential immunogens that can elicit a response from the host system. Three potential B-cell epitopes, i.e., SIAGQYRGQCNTCCDQ, LQEIVLHLEPQNELDP, and DSTVYLPPPSVARVVS, were identified in this study. A potential epitope for E6 (ATLERTEVY) was predicted to 8 MHC-I alleles (HLA-A*30:02, HLA-B*15:01, HLA-A*01:01, HLA-A*26:01, HLA-A*32:01, HLA-B*35:01, HLA-B*58:01, HLA-A*11:01) and for L1 epitope (NVFPIFLQM) was predicted for 4 MHC-I alleles (HLA-A*30:02, HLA-A*32:01, HLA-B*53:01, HLA-B*51:01). To conclude, the epitopes identified here might potentially be useful for developing a cervical cancer vaccine against HPV-45 strains, but in vitro and in vivo trials are needed to validate their safety and efficacy.
Collapse
|
27
|
Li C, Hall TG, Hall JJ, He WQ. Effectiveness of quadrivalent HPV vaccination in reducing vaccine-type and nonvaccine-type high risk HPV infection. Epidemiol Infect 2023; 151:e37. [PMID: 36789960 PMCID: PMC10028998 DOI: 10.1017/s0950268823000213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
This study aimed to assess human papillomavirus (HPV) vaccine effectiveness (VE) against both vaccine-type and nonvaccine-type high-risk HPV (hrHPV) infection, and duration of protection in United States. The study population was female participants aged 18-35 years with an HPV vaccination history and genital testing for HPV from the National Health and Nutrition Examination Survey, 2007-2016. Participants vaccinated before sexual debut were assessed against 13 nonvaccine-type hrHPV infection including 31/33/35/39/45/51/52/56/58/59/68/73/82. Multivariable logistic regression was used to estimate VE overall, by age at diagnosis, time since vaccination and lifetime sexual partners. A total of 3866 women were included in the analysis, with 23.3% (95% CI 21.3%-25.4%) having been vaccinated (≥1 dose). VE against vaccine-type HPV18/16/11/6 infection was 58% overall, which was mainly driven by those aged 18-22 years (VE = 64%) and 23-27 years (65%). Among participants aged 18-22 years vaccinated before sexual debut, the VE was 47% (23%-64%) against 13 nonvaccine-type hrHPV and 61% (95% CI 36%-77%) against 5 selected nonvaccine-type hrHPV35/39/52/58/59. Both direct effectiveness and cross-protection maintained effective for 5-10 years post vaccination. We also found the prevalence of ever diagnosed cervical cancer among vaccinated was significantly lower (0.46%, 4/874) than that among unvaccinated participants (1.27%, 38/2992). These findings highlight the potential of significant reduction of cervical cancer following the universal HPV vaccination programme.
Collapse
Affiliation(s)
- Chenxi Li
- Melbourne School of Population & Global Health, The University of Melbourne, Melbourne, 3053, Australia
| | - Thomas G Hall
- Children's Hospital at Westmead, Westmead, 2145, Australia
| | - John J Hall
- School of Population Health, UNSW Sydney, Sydney, 2052, Australia
| | - Wen-Qiang He
- Childrens Hospital at Westmead Clinical School, The University of Sydney, Sydney, 2145, Australia
| |
Collapse
|
28
|
Illah O, Olaitan A. Updates on HPV Vaccination. Diagnostics (Basel) 2023; 13:243. [PMID: 36673053 PMCID: PMC9857409 DOI: 10.3390/diagnostics13020243] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/11/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Cervical cancer still poses a significant global challenge. Developed countries have mitigated this challenge by the introduction of structured screening programmes and, more recently, the HPV vaccine. Countries that have successfully introduced national HPV vaccination programmes are on course for cervical cancer elimination in a few decades. In developing countries that lack structured screening and HPV vaccination programmes, cervical cancer remains a major cause of morbidity and mortality. The HPV vaccine is key to addressing the disproportionate distribution of cervical cancer incidence, with much to be gained from increasing vaccine coverage and uptake globally. This review covers the history and science of the HPV vaccine, its efficacy, effectiveness and safety, and some of the considerations and challenges posed to the achievement of global HPV vaccination coverage and the consequent elimination of cervical cancer.
Collapse
Affiliation(s)
- Ojone Illah
- Women’s Cancer Department, EGA Institute for Women’s Health, University College London, London WC1E 6BT, UK
| | | |
Collapse
|
29
|
Ma M, Xia B, Wang Z, Hao Y, Zhang T, Xu X. A novel C-terminal modification method enhanced the yield of human papillomavirus L1 or chimeric L1-L2 virus-like particles in the baculovirus system. Front Bioeng Biotechnol 2023; 10:1073892. [PMID: 36686228 PMCID: PMC9849392 DOI: 10.3389/fbioe.2022.1073892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/16/2022] [Indexed: 01/07/2023] Open
Abstract
Human papillomavirus (HPV) major capsid protein L1 virus-like particles (VLPs) produced in the baculovirus system showed excellent safety and immunogenicity, but the relatively high production cost stands as a substantial barrier to extensive commercialization, especially in producing multivalent vaccines. Here, a novel method, C-terminal basic amino acid (aa) substitution, was developed for increasing VLP and chimeric VLP (cVLP) production in this system. A series of mutants of five HPV types, including three L1 VLPs (6L1, 11L1, and 52L1) and two L1-L2 cVLPs (16L1-33L2, 58L1-16L2), were constructed. We found that most mutants exhibited higher protein expression in Sf9 cells, among which the yields of the superior mutants, 6L1CS4, 11L1CS3, 52L1m4∆N13CS1, 16L1-33L2 CS1, and 58L1-16L2 CS3, were up to 40, 35, 20, 35, and 60 mg/L, which respectively increased by 4.2-, 7.3-, 5-, 2.5-, and 3.4-fold, and they also showed robust immunogenicity and great stabilities. Additionally, we found that the increased level of steady-state mRNA may play a crucial role in promoting L1 protein expression. Our results demonstrated that this novel method was cost-effective and can be used to reduce the production costs of L1 VLPs and L1-L2 cVLPs to develop broadly protective and affordable multivalent HPV vaccines.
Collapse
Affiliation(s)
| | | | | | | | - Ting Zhang
- *Correspondence: Ting Zhang, ; Xuemei Xu,
| | - Xuemei Xu
- *Correspondence: Ting Zhang, ; Xuemei Xu,
| |
Collapse
|
30
|
Kitamura T, Suzuki M, Shigehara K, Fukuda K, Matsuyama T, Kume H. Prevalence of Human Papillomavirus Types 16/18 and Effect of Vaccination among Japanese Female General Citizens in the Vaccine Crisis Era. Viruses 2023; 15:159. [PMID: 36680199 PMCID: PMC9863140 DOI: 10.3390/v15010159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
The Japanese government withdrew its recommendation for human papillomavirus (HPV) vaccination in June 2013 and resumed it in April 2022. This period is known as the vaccine crisis in Japan. This study aimed to elucidate the prevalence and genotype distribution of HPV among Japanese female citizens, and the effect of vaccination against HPV-16/18 in the era of the vaccine crisis. We recruited Japanese female citizens and asked them to provide self-collected samples from the vaginal wall using cotton swabs for HPV genotyping. Furthermore, we collected the participants' characteristics, including lifestyle and experience of vaccination against HPV, to determine the significant association with HPV infection. HPV-16/18 positivity was found in 5.6% (115/2044) of participants. The highest vaccination rate was observed in the age group of 20-24 years (60.6%), whereas the lowest HPV-16/18 positivity was observed in the age group of 45-49 years (2.8%), followed by the age group of 20-24 years (4.0%). Experience with HPV vaccination significantly reduced the risk of HPV-16/18 infection (adjusted odds ratio, 0.047; 95% confidence interval, 0.011-0.196). Vaccinated women were much less likely to be infected by HPV-16/18, regardless of the HPV vaccine type or the vaccination dose.
Collapse
Affiliation(s)
- Tadaichi Kitamura
- Japanese Foundation of Sexual Health Medicine, Tokyo 113-0034, Japan
| | - Motofumi Suzuki
- Department of Urology, Tokyo Metropolitan Bokutoh Hospital, Tokyo 130-8575, Japan
| | - Kazuyoshi Shigehara
- Department of Urology, Faculty of Medicine, Kanazawa University, Kanazawa 920-8641, Japan
| | - Kazuko Fukuda
- Japanese Foundation of Sexual Health Medicine, Tokyo 113-0034, Japan
| | - Taeko Matsuyama
- Department of Nursing, Tachikawa Faculty of Nursing, Tokyo Healthcare University, Tachikawa 190-8590, Japan
| | - Haruki Kume
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| |
Collapse
|
31
|
Shadab R, Lavery JV, McFadden SM, Elharake JA, Malik F, Omer SB. Key ethical considerations to guide the adjudication of a single-dose HPV vaccine schedule. Hum Vaccin Immunother 2022; 18:1917231. [PMID: 34010096 PMCID: PMC8920253 DOI: 10.1080/21645515.2021.1917231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
There is a high burden of human papillomavirus (HPV) associated cancers in low- and middle-income countries (LMICs). Reducing the recommended dosing schedule from two doses to one makes the vaccine schedule logistically simpler and lowers the cost. This could make the distribution of the current vaccine supply more equitable and lead to the protection of more people. However, the clinical trials studying the efficacy of a single-dose schedule have not yet delivered final results. Against this background, the question is whether a single-dose HPV vaccine recommendation is appropriate now, and if so, what are the ethical considerations of such a recommendation? We developed three ethical recommendations: (1) adopt a holistic view of evidence to justify policy decisions; (2) prioritize the reduction in global disparities in decision-making at all levels; and (3) be transparent in the reporting of how key stakeholder interests have shaped the collection and interpretation of the evidence, and ultimate decisions. The complex discussion regarding the HPV single-dose vaccine schedule highlights the need for in-depth engagement globally to improve our understanding of country-specific contexts, and how those contexts influence decisions regarding the HPV vaccine single-dose recommendation.
Collapse
Affiliation(s)
- Ruha Shadab
- Yale Institute for Global Health, New Haven, CT, USA
| | - James V. Lavery
- Hubert Department of Global Health, Rollins School of Public Health and Center for Ethics, Emory University, Atlanta, GA, USA
| | - SarahAnn M. McFadden
- Yale Institute for Global Health, New Haven, CT, USA
- Department of Internal Medicine, Infectious Disease, Yale School of Medicine, New Haven, CT, USA
- CONTACT SarahAnn M. McFadden Yale Institute for Global Health, 1 Church St, Ste 340, New Haven, CT06510
| | - Jad A. Elharake
- Yale Institute for Global Health, New Haven, CT, USA
- Department of Health Policy and Management, Yale School of Public Health, New Haven, CT, USA
| | - Fauzia Malik
- Yale Institute for Global Health, New Haven, CT, USA
- Department of Health Policy and Management, Yale School of Public Health, New Haven, CT, USA
| | - Saad B. Omer
- Yale Institute for Global Health, New Haven, CT, USA
- Department of Internal Medicine, Infectious Disease, Yale School of Medicine, New Haven, CT, USA
- Yale School of Nursing, Orange, CT, USA
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| |
Collapse
|
32
|
Adebamowo SN, Befano B, Cheung LC, Rodriguez AC, Demarco M, Rydzak G, Chen X, Porras C, Herrero R, Kim JJ, Castle PE, Wentzensen N, Kreimer AR, Schiffman M, Campos NG. Different human papillomavirus types share early natural history transitions in immunocompetent women. Int J Cancer 2022; 151:920-929. [PMID: 35603904 PMCID: PMC9329241 DOI: 10.1002/ijc.34128] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 03/30/2022] [Accepted: 04/27/2022] [Indexed: 11/07/2022]
Abstract
Necessary stages of cervical carcinogenesis include acquisition of a carcinogenic human papillomavirus (HPV) type, persistence associated with the development of precancerous lesions, and invasion. Using prospective data from immunocompetent women in the Guanacaste HPV Natural History Study (NHS), the ASCUS-LSIL Triage Study (ALTS) and the Costa Rica HPV Vaccine Trial (CVT), we compared the early natural history of HPV types to inform transition probabilities for health decision models. We excluded women with evidence of high-grade cervical abnormalities at any point during follow-up and restricted the analysis to incident infections in all women and prevalent infections in young women (aged <30 years). We used survival approaches accounting for interval-censoring to estimate the time to clearance distribution for 20 529 HPV infections (64% were incident and 51% were carcinogenic). Time to clearance was similar across HPV types and risk classes (HPV16, HPV18/45, HPV31/33/35/52/58, HPV 39/51/56/59 and noncarcinogenic HPV types); and by age group (18-29, 30-44 and 45-54 years), among carcinogenic and noncarcinogenic infections. Similar time to clearance across HPV types suggests that relative prevalence can predict relative incidence. We confirmed that there was a uniform linear association between incident and prevalent infections for all HPV types within each study cohort. In the absence of progression to precancer, we observed similar time to clearance for incident infections across HPV types and risk classes. A singular clearance function for incident HPV infections has important implications for the refinement of microsimulation models used to evaluate the cost-effectiveness of novel prevention technologies.
Collapse
Affiliation(s)
- Sally N. Adebamowo
- Department of Epidemiology and Public Health, Greenebaum Comprehensive Cancer Center; University of Maryland School of Medicine, Baltimore, MD, United States
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, United States
| | - Brian Befano
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, United States
- Information Management Services Inc, Information Management, Calverton, NY, United States
| | - Li C. Cheung
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, United States
| | - Ana Cecilia Rodriguez
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, United States
| | - Maria Demarco
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, United States
| | - Greg Rydzak
- Information Management Services Inc, Information Management, Calverton, NY, United States
| | - Xiaojian Chen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, United States
| | - Carolina Porras
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica
| | - Rolando Herrero
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica
- Prevention and Implementation Group, International Agency for Research on Cancer, Lyon, France
| | - Jane J. Kim
- Center for Health Decision Science, Department of Health Policy and Management, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Philip E. Castle
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, United States
- Division of Cancer Prevention, National Cancer Institute, Rockville, MD, United States
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, United States
| | - Aimée R. Kreimer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, United States
| | - Mark Schiffman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, United States
| | - Nicole G. Campos
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, United States
- Center for Health Decision Science, Department of Health Policy and Management, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| |
Collapse
|
33
|
Bhattacharjee R, Kumar L, Dhasmana A, Mitra T, Dey A, Malik S, Kim B, Gundamaraju R. Governing HPV-related carcinoma using vaccines: Bottlenecks and breakthroughs. Front Oncol 2022; 12:977933. [PMID: 36176419 PMCID: PMC9513379 DOI: 10.3389/fonc.2022.977933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
Human papillomavirus (HPV) contributes to sexually transmitted infection, which is primarily associated with pre-cancerous and cancerous lesions in both men and women and is among the neglected cancerous infections in the world. At global level, two-, four-, and nine-valent pure L1 protein encompassed vaccines in targeting high-risk HPV strains using recombinant DNA technology are available. Therapeutic vaccines are produced by early and late oncoproteins that impart superior cell immunity to preventive vaccines that are under investigation. In the current review, we have not only discussed the clinical significance and importance of both preventive and therapeutic vaccines but also highlighted their dosage and mode of administration. This review is novel in its way and will pave the way for researchers to address the challenges posed by HPV-based vaccines at the present time.
Collapse
Affiliation(s)
- Rahul Bhattacharjee
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lamha Kumar
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, India
| | - Archna Dhasmana
- Himalayan School of Biosciences, Swami Rama Himalayan University, Dehradun, India
| | - Tamoghni Mitra
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal, India
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, Jharkhand, India
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
- *Correspondence: Bonglee Kim, ; Rohit Gundamaraju,
| | - Rohit Gundamaraju
- ER Stress and Mucosal Immunology Lab, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
- *Correspondence: Bonglee Kim, ; Rohit Gundamaraju,
| |
Collapse
|
34
|
Single-dose HPV vaccine immunity: is there a role for non-neutralizing antibodies? Trends Immunol 2022; 43:815-825. [PMID: 35995705 DOI: 10.1016/j.it.2022.07.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 07/28/2022] [Accepted: 07/30/2022] [Indexed: 11/22/2022]
Abstract
A single dose of human papillomavirus (HPV) vaccine against HPV infection (prerequisite for cervical cancer) appears to be as efficacious as two or three doses, despite inducing lower antibody titers. Neutralizing antibodies are thought to be the primary mediator of protection, but the threshold for protection is unknown. Antibody functions beyond neutralization have not been explored for HPV vaccines. Here, we discuss the immune mechanisms of HPV vaccines, with a focus on non-neutralizing antibody effector functions. In the context of single-dose HPV vaccination where antibody is limiting, we propose that non-neutralizing antibody functions may contribute to preventing HPV infection. Understanding the immunological basis of protection for single-dose HPV vaccination will provide a rationale for implementing single-dose HPV vaccine regimens.
Collapse
|
35
|
Shing JZ, Hu S, Herrero R, Hildesheim A, Porras C, Sampson JN, Schussler J, Schiller JT, Lowy DR, Sierra MS, Carvajal L, Kreimer AR. Precancerous cervical lesions caused by non-vaccine-preventable HPV types after vaccination with the bivalent AS04-adjuvanted HPV vaccine: an analysis of the long-term follow-up study from the randomised Costa Rica HPV Vaccine Trial. Lancet Oncol 2022; 23:940-949. [PMID: 35709811 PMCID: PMC9255557 DOI: 10.1016/s1470-2045(22)00291-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/04/2022] [Accepted: 05/10/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND In women vaccinated against human papillomavirus (HPV), reductions in cervical disease and related procedures results in more women having intact transformation zones, potentially increasing the risk of cervical lesions caused by non-vaccine-preventable HPV types, a phenomenon termed clinical unmasking. We aimed to evaluate HPV vaccine efficacy against cervical intraepithelial neoplasia grade 2 or worse (CIN2+) and cervical intraepithelial neoplasia grade 3 or worse (CIN3+) attributed to non-preventable HPV types in the long-term follow-up phase of the Costa Rica HPV Vaccine Trial (CVT). METHODS CVT was a randomised, double-blind, community-based trial done in Costa Rica. Eligible participants were women aged 18-25 years who were in general good health. Participants were randomly assigned (1:1) to receive an HPV 16 and 18 AS04-adjuvanted vaccine or control hepatitis A vaccine, using a blocked randomisation method (permuted block sizes of 14, 16, and 18). Vaccines in both groups were administered intramuscularly with 0·5 mL doses at 0, 1, and 6 months. Masking of vaccine allocation was maintained throughout the 4-year randomised trial phase, after which participants in the hepatitis A virus vaccine control group were provided the HPV vaccine and exited the study; a screening-only, unvaccinated control group was enrolled. The unvaccinated control group and HPV vaccine group were followed up for 7 years, during which treatment allocation was not masked. One of the prespecified primary endpoints for the long-term follow-up phase was precancers associated with HPV types not prevented by the vaccine, defined as histologically confirmed incident CIN2+ events or CIN3+ events attributed to any HPV type except HPV 16, 18, 31, 33, and 45. Our primary analytical period was years 7-11. Primary analyses were in all participants with at least one follow-up visit and excluded participants with a previous endpoint (ie, modified intention-to-treat cohort). Safety endpoints have been reported elsewhere. This trial is registered with ClinicalTrials.gov, NCT00128661 and NCT00867464. The randomised, masked trial phase is completed; an unmasked subset of women in the HPV-vaccinated group is under active investigation. FINDINGS Between June 28, 2004, and Dec 21, 2005, 7466 participants were enrolled (HPV vaccine group n=3727 and hepatitis A virus vaccine control group n=3739). Between March 30, 2009, and July 5, 2012, 2836 women enrolled in the new unvaccinated control group. The primary analytical cohort (years 7 to 11) included 2767 participants in the HPV vaccine group and 2563 in the unvaccinated group for the CIN2+ events endpoint assessment and 2826 participants in the HPV vaccine group and 2592 in the unvaccinated control group for the CIN3+ events endpoint assessment. Median follow-up during years 7 to 11 for women included for the CIN2+ events analysis was 52·8 months (IQR 44·0 to 60·7) for the HPV vaccine group and 49·8 months (42·0 to 56·9) for the unvaccinated control group. During years 7 to 11, clinical unmasking was observed with a negative vaccine efficacy against CIN2+ events attributed to non-preventable HPV types (-71·2% [95% CI -164·0 to -12·5]), with 9·2 (95% CI 2·1 to 15·6) additional CIN2+ events attributed to non-preventable HPV types per 1000 HPV-vaccinated participants versus HPV-unvaccinated participants. 27·0 (95% CI 14·2 to 39·9) fewer CIN2+ events irrespective of HPV type per 1000 vaccinated participants were observed during 11 years of follow-up. Vaccine efficacy against CIN3+ events attributed to non-preventable HPV types during years 7 to 11 was -135·0% (95% CI -329·8 to -33·5), with 8·3 (3·0 to 12·8) additional CIN3+ events attributed to non-preventable HPV types per 1000 vaccinated participants versus unvaccinated participants. INTERPRETATION Higher rates of CIN2+ events and CIN3+ events due to non-preventable HPV types in vaccinated versus unvaccinated participants suggests clinical unmasking could attenuate long-term reductions in high-grade disease following successful implementation of HPV vaccination programmes in screened populations. Importantly, the net benefit of vaccination remains considerable; therefore, HPV vaccination should still be prioritised as primary prevention for cervical cancer. FUNDING National Cancer Institute and National Institutes of Health Office of Research on Women's Health. TRANSLATION For the Spanish translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Jaimie Z Shing
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Shangying Hu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rolando Herrero
- Agencia Costarricense de Investigaciones Biomédicas (ACIB), Fundación INCIENSA, San José, Costa Rica; Early Detection and Prevention Section, International Agency for Research on Cancer, WHO, Lyon, France
| | - Allan Hildesheim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Carolina Porras
- Agencia Costarricense de Investigaciones Biomédicas (ACIB), Fundación INCIENSA, San José, Costa Rica
| | - Joshua N Sampson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - John T Schiller
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Douglas R Lowy
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mónica S Sierra
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Loretto Carvajal
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Agencia Costarricense de Investigaciones Biomédicas (ACIB), Fundación INCIENSA, San José, Costa Rica
| | - Aimée R Kreimer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
36
|
Tsang SH, Schiller JT, Porras C, Kemp TJ, Herrero R, Schussler J, Sierra MS, Cortes B, Hildesheim A, Lowy DR, Rodríguez AC, Romero B, Çuburu N, Shing JZ, Pinto LA, Sampson JN, Kreimer AR. HPV16 infection decreases vaccine-induced HPV16 antibody avidity: the CVT trial. NPJ Vaccines 2022; 7:40. [PMID: 35351898 PMCID: PMC8964739 DOI: 10.1038/s41541-022-00431-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 12/16/2021] [Indexed: 02/05/2023] Open
Abstract
The HPV vaccine has shown sustained efficacy and consistent stabilization of antibody levels, even after a single dose. We defined the HPV16-VLP antibody avidity patterns over 11 years among women who received one- or three doses of the bivalent HPV vaccine in the Costa Rica HPV Vaccine Trial. Absolute HPV16 avidity was lower in women who received one compared to three doses, although the patterns were similar (increased in years 2 and 3 and remained stable over the remaining 8 years). HPV16 avidity among women who were HPV16-seropositive women at HPV vaccination, a marker of natural immune response to HPV16 infection, was significantly lower than those of HPV16-seronegative women, a difference that was more pronounced among one-dose recipients. No differences in HPV16 avidity were observed by HPV18 serostatus at vaccination, confirming the specificity of the findings. Importantly, point estimates for vaccine efficacy against incident, six-month persistent HPV16 infections was similar between women who were HPV16 seronegative and seropositive at the time of initial HPV vaccination for both one-dose and three-dose participants. It is therefore likely that this lower avidity level is still sufficient to enable antibody-mediated protection. It is encouraging for long-term HPV-vaccine protection that HPV16 antibody avidity was maintained for over a decade, even after a single dose.
Collapse
Affiliation(s)
- Sabrina H Tsang
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - John T Schiller
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Carolina Porras
- Agencia Costarricense de Investigaciones Biomédicas (ACIB), formerly Proyecto Epidemiológico Guanacaste, Fundación INCIENSA, San José, Costa Rica
| | - Troy J Kemp
- HPV Immunology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Rolando Herrero
- Agencia Costarricense de Investigaciones Biomédicas (ACIB), formerly Proyecto Epidemiológico Guanacaste, Fundación INCIENSA, San José, Costa Rica
- Early Detection and Prevention Section, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | | | - Monica S Sierra
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Bernal Cortes
- Agencia Costarricense de Investigaciones Biomédicas (ACIB), formerly Proyecto Epidemiológico Guanacaste, Fundación INCIENSA, San José, Costa Rica
| | - Allan Hildesheim
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Douglas R Lowy
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Byron Romero
- Agencia Costarricense de Investigaciones Biomédicas (ACIB), formerly Proyecto Epidemiológico Guanacaste, Fundación INCIENSA, San José, Costa Rica
| | - Nicolas Çuburu
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jaimie Z Shing
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ligia A Pinto
- HPV Immunology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Joshua N Sampson
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Aimée R Kreimer
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
37
|
Logel M, Laurie C, El-Zein M, Guichon J, Franco EL. A review of ethical and legal aspects of gender-neutral human papillomavirus vaccination. Cancer Epidemiol Biomarkers Prev 2022; 31:919-931. [PMID: 35247878 DOI: 10.1158/1055-9965.epi-21-1256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/12/2022] [Accepted: 02/25/2022] [Indexed: 12/24/2022] Open
Abstract
While launching a campaign to eliminate cervical cancer, the World Health Organization called to halt human papillomavirus (HPV) gender-neutral vaccination (GNV) because of limited vaccine supply, raising ethical and legal questions about female-only vaccination versus GNV. We identified ethical and legal aspects of HPV GNV by searching MEDLINE for records up to February 19, 2021. We also provided an overview of HPV vaccines, the evolution of HPV vaccine recommendations in North America, and a timeline of male HPV vaccination introduction by searching PubMed, Google, and government websites. Four HPV vaccines are available: Cervarix®, Gardasil®, Gardasil®9, and Cecolin®. Vaccine recommendations in North America evolved from female only to eventually include males. Following the Food and Drug Administration's approval of the first HPV vaccine for males (2009), 35 countries began vaccinating males (2011-2020). Based on 56 eligible records out of 652, we identified the following constructs: lower male awareness of HPV and vaccination (n=13), limited economic resources (n=5), shared social responsibility (n=18), unprotected groups from female-only HPV vaccination (n=10), limited screening for HPV-associated noncervical cancers (n=6), consideration of ethical principles (n=17), and HPV vaccine mandates (n=5). Ethical and legal aspects must be considered when recommending vaccination for females only or GNV.
Collapse
|
38
|
Combita AL, Reyes V, Puerto D, Murillo R, Sánchez R, Nuñez M, Hernandez-Suarez GA, Wiesner C. Reduction in Vaccine HPV Type Infections in a Young Women Group (18-25 Years) Five Years after HPV Vaccine Introduction in Colombia. Cancer Prev Res (Phila) 2022; 15:55-66. [PMID: 34610993 PMCID: PMC9662904 DOI: 10.1158/1940-6207.capr-21-0063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/29/2021] [Accepted: 09/21/2021] [Indexed: 01/07/2023]
Abstract
In Colombia, the human papillomavirus (HPV) vaccine was launched in 2012 in the context of a school-based national vaccination program targeting girls ages 9 to 14 and offering catch-up vaccination for girls ages 14 to 17. In this study, we evaluated the program's impact on type-specific HPV infection by comparing HPV cervical prevalence among vaccinated and nonvaccinated women. This is a comparative cross-sectional study conducted 5 years after the quadrivalent HPV vaccination implementation in a sentinel Colombian City. This study included young women (18-25 years old) who had been vaccinated in the catch-up group and were attending universities and technical institutions, and women who attended primary health care facilities for Pap smear screening. The HPV prevalence of 1,287 unvaccinated women was compared with the prevalence of 1,986 vaccinated women. The prevalence of HPV16/18 infections was significantly lower in vaccinated compared with unvaccinated women (6.5% vs. 15.4%; P < 0.001), whereas for HPV6/11 infections, a decrease of 63.7% in vaccinated women (1.02% vs. 2.81%) was observed. The adjusted effectiveness to HPV16/18 was 61.4%; 95% CI, 54.3%-67.6%. However, the effectiveness against HPV16/18 was significantly higher among women vaccinated before their sexual debut 91.5%; 95% CI, 86.8-94.5, compared with effectiveness for vaccination after their sexual debut, 36.2%; 95% CI, 23.6-46.7. Five years after the introduction of HPV vaccines in Colombia, high effectiveness of HPV to prevent HPV16/18 infections is observed in the catch-up cohorts including virgin and sexually active women. PREVENTION RELEVANCE: Monitoring HPV vaccines post-licensure plays an important role in assessing the progress of immunization programs, demonstrating the impact of vaccines on the population, and providing data for policy needs. In Colombia, HPV vaccines showed effectiveness when administered before start of sexual activity, and two doses are sufficient to achieve good protection.
Collapse
Affiliation(s)
- Alba L. Combita
- Grupo de Investigación en Biología del Cáncer, Instituto Nacional de Cancerología (INC), Bogotá, Colombia.,Departamento de Microbiología, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia.,Corresponding Author: Alba L. Combita, Cancer Biology Research Group, National Cancer Institute of Colombia (INC), Calle 1 #9-85, Bogotá 111511, Colombia. Phone: 571-4320160, ext. 4212; Fax: 571-3341360; E-mail:
| | - Viviana Reyes
- Grupo de Investigación en Biología del Cáncer, Instituto Nacional de Cancerología (INC), Bogotá, Colombia
| | - Devi Puerto
- Grupo de Investigación en Salud Pública y Epidemiología, INC, Bogotá, Colombia
| | - Raúl Murillo
- Centro Javeriano de Oncología, Hospital Universitario San Ignacio, Bogotá, Colombia
| | | | - Marcela Nuñez
- GASPI. Grupo Apoyo y Seguimiento para la Investigación, INC, Bogotá, Colombia
| | | | - Carolina Wiesner
- Grupo de Investigación en Salud Pública y Epidemiología, INC, Bogotá, Colombia
| |
Collapse
|
39
|
Current view on novel vaccine technologies to combat human infectious diseases. Appl Microbiol Biotechnol 2022; 106:25-56. [PMID: 34889981 PMCID: PMC8661323 DOI: 10.1007/s00253-021-11713-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/19/2021] [Accepted: 11/24/2021] [Indexed: 02/06/2023]
Abstract
Inactivated and live attenuated vaccines have improved human life and significantly reduced morbidity and mortality of several human infectious diseases. However, these vaccines have faults, such as reactivity or suboptimal efficacy and expensive and time-consuming development and production. Additionally, despite the enormous efforts to develop vaccines against some infectious diseases, the traditional technologies have not been successful in achieving this. At the same time, the concerns about emerging and re-emerging diseases urge the need to develop technologies that can be rapidly applied to combat the new challenges. Within the last two decades, the research of vaccine technologies has taken several directions to achieve safe, efficient, and economic platforms or technologies for novel vaccines. This review will give a brief overview of the current state of the novel vaccine technologies, new vaccine candidates in clinical trial phases 1-3 (listed by European Medicines Agency (EMA) and Food and Drug Administration (FDA)), and vaccines based on the novel technologies which have already been commercially available (approved by EMA and FDA) with the special reference to pandemic COVID-19 vaccines. KEY POINTS: • Vaccines of the new generation follow the minimalist strategy. • Some infectious diseases remain a challenge for the vaccine development. • The number of new vaccine candidates in the late phase clinical trials remains low.
Collapse
|
40
|
Castle PE, Einstein MH, Sahasrabuddhe VV. Cervical cancer prevention and control in women living with human immunodeficiency virus. CA Cancer J Clin 2021; 71:505-526. [PMID: 34499351 PMCID: PMC10054840 DOI: 10.3322/caac.21696] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/07/2021] [Accepted: 08/10/2021] [Indexed: 12/16/2022] Open
Abstract
Despite being highly preventable, cervical cancer is the fourth most common cancer and cause of cancer death in women globally. In low-income countries, cervical cancer is often the leading cause of cancer-related morbidity and mortality. Women living with human immunodeficiency virus (HIV)/acquired immunodeficiency syndrome are at a particularly high risk of cervical cancer because of an impaired immune response to human papillomavirus, the obligate cause of virtually all cervical cancers. Globally, approximately 1 in 20 cervical cancers is attributable to HIV; in sub-Saharan Africa, approximately 1 in 5 cervical cancers is due to HIV. Here, the authors provide a critical appraisal of the evidence to date on the impact of HIV disease on cervical cancer risk, describe key methodologic issues, and frame the key outstanding research questions, especially as they apply to ongoing global efforts for prevention and control of cervical cancer. Expanded efforts to integrate HIV care with cervical cancer prevention and control, and vice versa, could assist the global effort to eliminate cervical cancer as a public health problem.
Collapse
Affiliation(s)
- Philip E. Castle
- Division of Cancer PreventionNational Cancer InstituteNational Institutes of HealthRockvilleMaryland
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteNational Institutes of HealthRockvilleMaryland
| | - Mark H. Einstein
- Department of Obstetrics, Gynecology, and Reproductive HealthRutgers New Jersey Medical SchoolNewarkNew Jersey
| | - Vikrant V. Sahasrabuddhe
- Division of Cancer PreventionNational Cancer InstituteNational Institutes of HealthRockvilleMaryland
| |
Collapse
|
41
|
Basu P, Malvi SG, Joshi S, Bhatla N, Muwonge R, Lucas E, Verma Y, Esmy PO, Poli URR, Shah A, Zomawia E, Pimple S, Jayant K, Hingmire S, Chiwate A, Divate U, Vashist S, Mishra G, Jadhav R, Siddiqi M, Sankaran S, Prabhu PR, Kannan TPRA, Varghese R, Shastri SS, Anantharaman D, Gheit T, Tommasino M, Sauvaget C, Pillai MR, Sankaranarayanan R. 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. Lancet Oncol 2021; 22:1518-1529. [PMID: 34634254 PMCID: PMC8560643 DOI: 10.1016/s1470-2045(21)00453-8] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/16/2021] [Accepted: 07/26/2021] [Indexed: 12/02/2022]
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.
Collapse
Affiliation(s)
- Partha Basu
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, Lyon, France.
| | - Sylla G Malvi
- Tata Memorial Centre Rural Cancer Project, Nargis Dutt Memorial Cancer Hospital, Barshi District Solapur, Maharashtra, India
| | - Smita Joshi
- Jehangir Clinical Development Centre, Jehangir Hospital Premises, Pune, India
| | - Neerja Bhatla
- Department of Obstetrics and Gynaecology, All India Institute of Medical Sciences, New Delhi, India
| | - Richard Muwonge
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, Lyon, France
| | - Eric Lucas
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, Lyon, France
| | - Yogesh Verma
- Sikkim Manipal Institute of Medical Sciences, Sikkim Manipal University, Gangtok, Sikkim, India
| | - Pulikkottil O Esmy
- Christian Fellowship Community Health Centre, Ambillikai, Dindigul District, Tamil Nadu, India
| | | | - Anand Shah
- Department of Community Oncology, Gujarat Cancer and Research Institute, M P Shah Cancer Hospital, Civil Hospital Campus, Asarwa, Ahmedabad, India
| | | | - Sharmila Pimple
- Department of Preventive Oncology, Centre for Cancer Epidemiology, Tata Memorial Centre, Homi Bhabha National Institute, Parel, Mumbai, India
| | - Kasturi Jayant
- Tata Memorial Centre Rural Cancer Project, Nargis Dutt Memorial Cancer Hospital, Barshi District Solapur, Maharashtra, India
| | - Sanjay Hingmire
- Tata Memorial Centre Rural Cancer Project, Nargis Dutt Memorial Cancer Hospital, Barshi District Solapur, Maharashtra, India
| | - Aruna Chiwate
- Tata Memorial Centre Rural Cancer Project, Nargis Dutt Memorial Cancer Hospital, Barshi District Solapur, Maharashtra, India
| | - Uma Divate
- Jehangir Clinical Development Centre, Jehangir Hospital Premises, Pune, India
| | - Shachi Vashist
- Department of Obstetrics and Gynaecology, All India Institute of Medical Sciences, New Delhi, India
| | - Gauravi Mishra
- Department of Preventive Oncology, Centre for Cancer Epidemiology, Tata Memorial Centre, Homi Bhabha National Institute, Parel, Mumbai, India
| | - Radhika Jadhav
- Jehangir Clinical Development Centre, Jehangir Hospital Premises, Pune, India
| | | | - Subha Sankaran
- Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram, Kerala, India
| | - Priya Ramesh Prabhu
- Human Biology Division, Fred Hutchinson Cancer Research Centre, Seattle, WA, USA
| | | | - Rintu Varghese
- Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram, Kerala, India
| | - Surendra S Shastri
- Department of Health Disparities Research, Division of Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Devasena Anantharaman
- Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram, Kerala, India
| | - Tarik Gheit
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, Lyon, France
| | - Massimo Tommasino
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, Lyon, France
| | - Catherine Sauvaget
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, Lyon, France
| | - M Radhakrishna Pillai
- Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram, Kerala, India
| | | |
Collapse
|
42
|
Hopkins KL, Jaffer M, Hlongwane KE, Otwombe K, Dietrich J, Cheyip M, Olivier J, Doherty T, Gray GE. Assessing national cervical cancer screening guidelines: Results from an HIV testing clinic also screening for cervical cancer and HPV in Soweto, South Africa. PLoS One 2021; 16:e0255124. [PMID: 34329334 PMCID: PMC8323926 DOI: 10.1371/journal.pone.0255124] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/09/2021] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE A screening centre in Soweto, South Africa (SA), investigated high-risk human papillomavirus (HR-HPV), HIV, cervical cancer risk amongst women. METHODS This cross-sectional study (June 2018-March 2019) describes screening results (Roche Linear Array HPV test and Pap smear liquid based cytology) and history of screening (known HIV status, antiretroviral therapy [ART] use, previous Pap smears). Data were stratified by age group (18-29, 30+ years), HIV status, Pap smear results and tested for statistical significance. RESULTS Of 280 women, 20.4% were HIV-positive, 18.2% had abnormal Pap smears, 41.8% had HR-HPV. Of older women, 48.2% (n = 78/162) had never had a Pap smear. Of younger women, 89.0% (n = 105/118) never had a Pap smear, but had significantly more low-grade squamous intraepithelial lesions (LSIL) and other HR-HPV infection than older women (12.7%[n = 15/118] vs 4.9%[n = 8/162], p = 0.0193; and 49.2%[n = 58/118] vs 29.0%[n = 47/162], p = 0.0006; respectively). HIV-positive women had more abnormal cytology results and infection with other HR-HPV types or co-infection with other HR-HPV type(s)/HPV-16 compared to HIV-negative women (35.1%[n = 20/57] vs 13.9%[n = 31/223], p = 0.0002; 56.1%[n = 32/57] vs 32.7%[n = 73/223], p = 0.001; and 12.3%[n = 7/57] vs 4.9%[n = 11/223], p = 0.044; respectively). Of 57 HIV-positive women, 45.6% (n = 26) already knew their HIV status; of which 69.2% were on ART and 34.6% never had a Pap smear. CONCLUSION South African women have high rates of HIV, Pap smear abnormalities and HR-HPV, with low cervical cancer screening coverage. SA cervical cancer screening policy excludes (undiagnosed) HIV-positive and HIV-negative women <30 years, both populations found to have high prevalence of HR-HPV. HPV-based primary screening from 25 years could improve outcomes.
Collapse
Affiliation(s)
- Kathryn L. Hopkins
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Maya Jaffer
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa
| | - Khuthadzo E. Hlongwane
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa
| | - Kennedy Otwombe
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa
| | - Janan Dietrich
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa
- Health Systems Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Mireille Cheyip
- Centers for Disease Control and Prevention, Pretoria, South Africa
| | - Jacobus Olivier
- Centers for Disease Control and Prevention, Pretoria, South Africa
| | - Tanya Doherty
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Health Systems Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Glenda E. Gray
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa
- Office of the President, South African Medical Research Council, Cape Town, South Africa
| |
Collapse
|
43
|
Rosalik K, Tarney C, Han J. Human Papilloma Virus Vaccination. Viruses 2021; 13:v13061091. [PMID: 34201028 PMCID: PMC8228159 DOI: 10.3390/v13061091] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/19/2021] [Accepted: 05/25/2021] [Indexed: 12/30/2022] Open
Abstract
Human papilloma virus (HPV) is the most common sexually transmitted infection worldwide causing a variety of benign and malignant conditions. A significant portion of the global population is infected with HPV, with the virus attributed to causing up to 5% of cancers worldwide. Bivalent, quadrivalent, and nine-valent vaccinations exist to aid in the prevention of these diseases and have been proven to be effective at preventing both benign and malignant disease. While vaccination is readily accessible in more developed countries, barriers exist to worldwide distribution and acceptance of vaccination. Vaccination and screening of HPV infection when used in combination are proven and predicted to decrease HPV related pathology. Improvements in vaccination formulations, for treatment as well as prevention, are actively being sought from a variety of mechanisms. Despite these advancements, and the data supporting their efficacy, there has been substantial delay in obtaining adequate vaccination coverage. In reviewing these challenges and looking forward to new vaccine development—especially within the current pandemic—it is clear from the challenges of HPV we require methods to more effectively encourage vaccination, ways to dispel vaccination myths as they occur, and implement better processes for vaccine distribution globally.
Collapse
Affiliation(s)
- Kendal Rosalik
- Madigan Army Medical Center, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, 9040A Jackson Ave, Joint Base Lewis-McChord, WA 98431, USA;
- Correspondence:
| | - Christopher Tarney
- Madigan Army Medical Center, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, 9040A Jackson Ave, Joint Base Lewis-McChord, WA 98431, USA;
| | - Jasmine Han
- General Leonard Wood Army Community Hospital, Department of Obstetrics and Gynecology, 4430 Missouri Ave, Ford Leonard Wood, MO 65473, USA;
| |
Collapse
|
44
|
Mariz FC, Gray P, Bender N, Eriksson T, Kann H, Apter D, Paavonen J, Pajunen E, Prager KM, Sehr P, Surcel HM, Waterboer T, Müller M, Pawlita M, Lehtinen M. Sustainability of neutralising antibodies induced by bivalent or quadrivalent HPV vaccines and correlation with efficacy: a combined follow-up analysis of data from two randomised, double-blind, multicentre, phase 3 trials. THE LANCET. INFECTIOUS DISEASES 2021; 21:1458-1468. [PMID: 34081923 DOI: 10.1016/s1473-3099(20)30873-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/24/2020] [Accepted: 11/03/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Quadrivalent and bivalent vaccines against oncogenic human papillomavirus (HPV) are used worldwide with different reported overall efficacies against HPV infections. Although protective concentrations of vaccine-induced antibodies are still not formally defined, we evaluated the sustainability of neutralising antibodies in vaccine trial participants 2-12 years after vaccination and the correlation with reported vaccine efficacy. METHODS We did a follow-up analysis of data from the Finnish cohorts of two international, randomised, double-blind, phase 3 trials of HPV vaccines, PATRICIA (bivalent, HPV16 and 18) and FUTURE II (quadrivalent, HPV6, 11, 16, and 18). In 2002 and 2004-05, respectively, Finnish girls aged 16-17 years participated in one of these two trials and consented to health registry follow-up with the Finnish Cancer Registry. The cohorts were also linked with the Finnish Maternity Cohort (FMC) that collects first-trimester serum samples from nearly all pregnant Finnish women, resulting in 2046 post-vaccination serum samples obtained during up to 12 years of follow-up. We obtained serum samples from the FMC-based follow-up of the FUTURE II trial (from the quadrivalent vaccine recipients) and the PATRICIA trial (from corresponding bivalent vaccine recipients who were aligned by follow-up time, and matched by the number of pregnancies). We assessed neutralising antibody concentrations (type-specific seroprevalence) to HPV6, 16, and 18, and cross-neutralising antibody responses to non-vaccine HPV types 31, 33, 45, 52, and 58 from 2 to 12 years after vaccination. FINDINGS Up to Dec 31, 2016, we obtained and analysed 577 serum samples from the quadrivalent vaccine recipients and 568 from the bivalent vaccine recipients. In 681 first-pregnancy serum samples, neutralising antibodies to HPV6, 16, and 18 were generally found up to 12 years after vaccination. However, 51 (15%) of 339 quadrivalent vaccine recipients had no detectable HPV18 neutralising antibodies 2-12 years after vaccination, whereas all 342 corresponding bivalent vaccine recipients had HPV18 neutralising antibodies.. In seropositive quadrivalent vaccine recipients, HPV16 geometric mean titres (GMT) halved by years 5-7 (GMT 3679, 95% CI 2377 to 4708) compared with years 2-4 (6642, 2371 to 13 717). Between 5 and 12 years after vaccination, GMT of neutralising antibodies to HPV16 and 18 were 5·7 times and 12·4 times higher, respectively, in seropositive bivalent vaccine recipients than in the quadrivalent vaccine recipients. Cross-neutralising antibodies to HPV31, 33, 45, 52, and 58 were more prevalent in the bivalent vaccine recipients but, when measurable, sustainable up to 12 years after vaccination with similar GMTs in both vaccine cohorts. Seroprevalence for HPV16, 31, 33, 52, and 58 significantly correlated with vaccine efficacy against persistent HPV infections in the bivalent vaccine recipients only (rs=0·90, 95% CI 0·09 to 0·99, p=0·037, compared with rs=0·62, 95% CI -0·58 to 0·97, p=0·27 for the quadrivalent vaccine recipients). Correlation of protection with prevalence of neutralising or cross-neutralising HPV antibodies was not significant in the quadrivalent vaccine recipients. INTERPRETATION The observed significant differences in the immunogenicity of the two vaccines are in line with the differences in their cross-protective efficacy. Protective HPV vaccine-induced antibody titres can be detected up to 12 years after vaccination. FUNDING Academy of Finland and Finnish Cancer Foundation.
Collapse
Affiliation(s)
- Filipe Colaço Mariz
- Tumorvirus-Specific Vaccination Strategies, Deutsches Krebsforschungszentrum, Heidelberg, Germany.
| | - Penelope Gray
- Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Noemi Bender
- Infections and Cancer Epidemiology, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Tiina Eriksson
- FICAN-Mid, Pirkanmaan Sairaanhoitopiiri, Research, Development and Innovation Centre Nuorisotutkimusasema, Tampere, Finland
| | - Hanna Kann
- Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | | | - Jorma Paavonen
- Department of Obstetrics and Gynecology, University of Helsinki, Helsinki, Finland
| | | | - Kristina M Prager
- Infections and Cancer Epidemiology, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Peter Sehr
- EMBL-DKFZ Chemical Biology Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Heljä-Marja Surcel
- Biobank Borealis of Northern Finland, Oulu University Hospital, Oulu, Finland; Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Tim Waterboer
- Infections and Cancer Epidemiology, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Martin Müller
- Tumorvirus-Specific Vaccination Strategies, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Michael Pawlita
- Infections and Cancer Epidemiology, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Matti Lehtinen
- Infections and Cancer Epidemiology, Deutsches Krebsforschungszentrum, Heidelberg, Germany; FICAN-Mid, Pirkanmaan Sairaanhoitopiiri, Research, Development and Innovation Centre Nuorisotutkimusasema, Tampere, Finland; Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
45
|
Systematic literature review of cross-protective effect of HPV vaccines based on data from randomized clinical trials and real-world evidence. Vaccine 2021; 39:2224-2236. [PMID: 33744051 DOI: 10.1016/j.vaccine.2020.11.076] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/07/2020] [Accepted: 11/30/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND The extent of cross-protection provided by currently licensed bivalent and quadrivalent HPV vaccines versus direct protection against HPV 31-, 33-, 45-, 52-, and 58-related disease is debated. A systematic literature review was conducted to establish the duration and magnitude of cross-protection in interventional and observational studies. METHODS PubMed and Embase databases were searched to identify randomized controlled trials (RCT) and observational studies published between 2008 and 2019 reporting on efficacy and effectiveness of HPV vaccines in women against non-vaccine types 31, 33, 45, 52, 58, and 6 and 11 (non-bivalent types). Key outcomes of interest were vaccine efficacy against 6- and 12-month persistent infection or genital lesions, and type-specific genital HPV prevalence or incidence. RCT data were analyzed for the according-to-protocol (bivalent vaccine) or negative-for-14-HPV-types (quadrivalent vaccine) efficacy cohorts. RESULTS Data from 23 RCTs and 33 observational studies evaluating cross-protection were extracted. RCTs assessed cross-protection in post-hoc analyses of small size subgroups. Among fully vaccinated, baseline HPV-naïve women, the bivalent vaccine showed statistically significant cross-protective efficacy, although with wide confidence intervals, against 6-month and 12-month persistent cervical infections and CIN2+ only consistently for HPV 31 and 45, with the highest effect observed for HPV 31 (range 64.6% [95% CI: 27.6 to 83.9] to 79.1% [97.7% CI: 27.6 to 95.9] for 6-month persistent infection; maximal follow-up 4.7 years). No cross-protection was shown in extended follow-up. The quadrivalent vaccine efficacy reached statistical significance for HPV 31 (46.2% [15.3-66.4]; follow-up: 3.6 years). Similarly, observational studies found consistently significant effectiveness only against HPV 31 and 45 with both vaccines. CONCLUSIONS RCTs and observational studies show that cross-protection is inconsistent across non-vaccine HPV types and is largely driven by HPV 31 and 45. Furthermore, existing data suggest that it wanes over time; its long-term durability has not been established.
Collapse
|
46
|
Taku O, Mbulawa ZZA, Phohlo K, Garcia-Jardon M, Businge CB, Williamson AL. Distribution of Human Papillomavirus (HPV) Genotypes in HIV-Negative and HIV-Positive Women with Cervical Intraepithelial Lesions in the Eastern Cape Province, South Africa. Viruses 2021; 13:v13020280. [PMID: 33670231 PMCID: PMC7916956 DOI: 10.3390/v13020280] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/29/2021] [Accepted: 02/08/2021] [Indexed: 12/14/2022] Open
Abstract
South African women have a high rate of cervical cancer cases, but there are limited data on human papillomavirus (HPV) genotypes in cervical intraepithelial neoplasia (CIN) in the Eastern Cape province, South Africa. A total of 193 cervical specimens with confirmed CIN from women aged 18 years or older, recruited from a referral hospital, were tested for HPV infection. The cervical specimens, smeared onto FTA cards, were screened for 36 HPV types using an HPV direct flow kit. HPV prevalence was 93.5% (43/46) in CIN2 and 96.6% (142/147) in CIN3. HIV-positive women had a significantly higher HPV prevalence than HIV-negative women (98.0% vs. 89.1%, p = 0.012). The prevalence of multiple types was significantly higher in HIV-positive than HIV-negative women (p = 0.034). The frequently detected genotypes were HPV35 (23.9%), HPV58 (23.9%), HPV45 (19.6%), and HPV16 (17.3%) in CIN2 cases, while in CIN3, HPV35 (22.5%), HPV16 (21.8%), HPV33 (15.6%), and HPV58 (14.3%) were the most common identified HPV types, independent of HIV status. The prevalence of HPV types targeted by the nonavalent HPV vaccine was 60.9% and 68.7% among women with CIN2 and CIN3, respectively, indicating that vaccination would have an impact both in HIV-negative and HIV-positive South African women, although it will not provide full protection in preventing HPV infection and cervical cancer lesions.
Collapse
Affiliation(s)
- Ongeziwe Taku
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa; (O.T.); (Z.Z.A.M.); (K.P.)
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa
| | - Zizipho Z. A. Mbulawa
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa; (O.T.); (Z.Z.A.M.); (K.P.)
- SAMRC Gynaecological Cancer Research Centre, University of Cape Town, Cape Town 7925, South Africa
- Department of Laboratory Medicine and Pathology, Walter Sisulu University, Mthatha 5100, South Africa
- National Health Laboratory Service, Nelson Mandela Academic Hospital, Mthatha 5100, South Africa
| | - Keletso Phohlo
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa; (O.T.); (Z.Z.A.M.); (K.P.)
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa
| | - Mirta Garcia-Jardon
- Department of Pathology, Walter Sisulu University and National Health Laboratory Service, Mthatha 5100, South Africa;
| | - Charles B. Businge
- Department of Obstetrics and Gynaecology, Nelson Mandela Academic Hospital, Mthatha 5100, South Africa;
- Department of Obstetrics and Gynaecology, Faculty of Health Sciences, Walter Sisulu University, Mthatha 5100, South Africa
| | - Anna-Lise Williamson
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa; (O.T.); (Z.Z.A.M.); (K.P.)
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa
- SAMRC Gynaecological Cancer Research Centre, University of Cape Town, Cape Town 7925, South Africa
- Correspondence: ; Tel.: +21-4066124
| |
Collapse
|
47
|
Porras C, Tsang SH, Herrero R, Guillén D, Darragh TM, Stoler MH, Hildesheim A, Wagner S, Boland J, Lowy DR, Schiller JT, Schiffman M, Schussler J, Gail MH, Quint W, Ocampo R, Morales J, Rodríguez AC, Hu S, Sampson JN, Kreimer AR. Efficacy of the bivalent HPV vaccine against HPV 16/18-associated precancer: long-term follow-up results from the Costa Rica Vaccine Trial. Lancet Oncol 2020; 21:1643-1652. [PMID: 33271093 PMCID: PMC8724969 DOI: 10.1016/s1470-2045(20)30524-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/17/2020] [Accepted: 08/20/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Oncogenic human papillomavirus (HPV) infections cause most cases of cervical cancer. Here, we report long-term follow-up results for the Costa Rica Vaccine Trial (publicly funded and initiated before licensure of the HPV vaccines), with the aim of assessing the efficacy of the bivalent HPV vaccine for preventing HPV 16/18-associated cervical intraepithelial neoplasia grade 2 or worse (CIN2+). METHODS Women aged 18-25 years were enrolled in a randomised, double-blind, controlled trial in Costa Rica, between June 28, 2004, and Dec 21, 2005, designed to assess the efficacy of a bivalent vaccine for the prevention of infection with HPV 16/18 and associated precancerous lesions at the cervix. Participants were randomly assigned (1:1) to receive an HPV 16/18 AS04-adjuvanted vaccine or control hepatitis A vaccine. Vaccines were administered intramuscularly in three 0·5 mL doses at 0, 1, and 6 months and participants were followed up annually for 4 years. After the blinded phase, women in the HPV vaccine group were invited to enrol in the long-term follow-up study, which extended follow-up for 7 additional years. The control group received HPV vaccine and was replaced with a new unvaccinated control group. Women were followed up every 2 years until year 11. Investigators and patients were aware of treatment allocation for the follow-up phase. At each visit, clinicians collected cervical cells from sexually active women for cytology and HPV testing. Women with abnormal cytology were referred to colposcopy, biopsy, and treatment as needed. Women with negative results at the last screening visit (year 11) exited the long-term follow-up study. The analytical cohort for vaccine efficacy included women who were HPV 16/18 DNA-negative at vaccination. The primary outcome of this analysis was defined as histopathologically confirmed CIN2+ or cervical intraepithelial neoplasia grade 3 or worse associated with HPV 16/18 cervical infection detected at colposcopy referral. We calculated vaccine efficacy by year and cumulatively. This long-term follow-up study is registered with ClinicalTrials.gov, NCT00867464. FINDINGS 7466 women were enrolled in the Costa Rica Vaccine Trial; 3727 received the HPV vaccine and 3739 received the control vaccine. Between March 30, 2009, and July 5, 2012, 2635 women in the HPV vaccine group and 2836 women in the new unvaccinated control group were enrolled in the long-term follow-up study. 2635 women in the HPV vaccine group and 2677 women in the control group were included in the analysis cohort for years 0-4, and 2073 women from the HPV vaccine group and 2530 women from the new unvaccinated control group were included in the analysis cohort for years 7-11. Median follow-up time for the HPV group was 11·1 years (IQR 9·1-11·7), 4·6 years (4·3-5·3) for the original control group, and 6·2 years (5·5-6·9) for the new unvaccinated control group. At year 11, vaccine efficacy against incident HPV 16/18-associated CIN2+ was 100% (95% CI 89·2-100·0); 34 (1·5%) of 2233 unvaccinated women had a CIN2+ outcome compared with none of 1913 women in the HPV group. Cumulative vaccine efficacy against HPV 16/18-associated CIN2+ over the 11-year period was 97·4% (95% CI 88·0-99·6). Similar protection was observed against HPV 16/18-associated CIN3-specifically at year 11, vaccine efficacy was 100% (95% CI 78·8-100·0) and cumulative vaccine efficacy was 94·9% (73·7-99·4). During the long-term follow-up, no serious adverse events occurred that were deemed related to the HPV vaccine. The most common grade 3 or worse serious adverse events were pregnancy, puerperium, and perinatal conditions (in 255 [10%] of 2530 women in the unvaccinated control group and 201 [10%] of 2073 women in the HPV vaccine group). Four women in the unvaccinated control group and three in the HPV vaccine group died; no deaths were deemed to be related to the HPV vaccine. INTERPRETATION The bivalent HPV vaccine has high efficacy against HPV 16/18-associated precancer for more than a decade after initial vaccination, supporting the notion that invasive cervical cancer is preventable. FUNDING US National Cancer Institute.
Collapse
Affiliation(s)
- Carolina Porras
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica.
| | - Sabrina H Tsang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Rolando Herrero
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica; Early Detection and Prevention Section, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Diego Guillén
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica
| | | | - Mark H Stoler
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
| | - Allan Hildesheim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Sarah Wagner
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD, USA
| | - Joseph Boland
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD, USA
| | - Douglas R Lowy
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - John T Schiller
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Mark Schiffman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | | | - Mitchell H Gail
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Wim Quint
- DDL Diagnostic Laboratory, Rijswijk, Netherlands
| | - Rebeca Ocampo
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica
| | - Jorge Morales
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica
| | | | - Shangying Hu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Joshua N Sampson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Aimée R Kreimer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
48
|
Kreimer AR, Sampson JN, Porras C, Schiller JT, Kemp T, Herrero R, Wagner S, Boland J, Schussler J, Lowy DR, Chanock S, Roberson D, Sierra MS, Tsang SH, Schiffman M, Rodriguez AC, Cortes B, Gail MH, Hildesheim A, Gonzalez P, Pinto LA. Evaluation of Durability of a Single Dose of the Bivalent HPV Vaccine: The CVT Trial. J Natl Cancer Inst 2020; 112:1038-1046. [PMID: 32091594 PMCID: PMC7566548 DOI: 10.1093/jnci/djaa011] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 11/18/2019] [Accepted: 12/19/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The authors investigated the durability of vaccine efficacy (VE) against human papillomavirus (HPV)16 or 18 infections and antibody response among nonrandomly assigned women who received a single dose of the bivalent HPV vaccine compared with women who received multiple doses and unvaccinated women. METHODS HPV infections were compared between HPV16 or 18-vaccinated women aged 18 to 25 years who received one (N = 112), two (N = 62), or three (N = 1365) doses, and age- and geography-matched unvaccinated women (N = 1783) in the long-term follow-up of the Costa Rica HPV Vaccine Trial. Cervical HPV infections were measured at two study visits, approximately 9 and 11 years after initial HPV vaccination, using National Cancer Institute next-generation sequencing TypeSeq1 assay. VE and 95% confidence intervals (CIs) were estimated. HPV16 or 18 antibody levels were measured in all one- and two-dose women, and a subset of three-dose women, using a virus-like particle-based enzyme-linked immunosorbent assay (n = 448). RESULTS Median follow-up for the HPV-vaccinated group was 11.3 years (interquartile range = 10.9-11.7 years) and did not vary by dose group. VE against prevalent HPV16 or 18 infection was 80.2% (95% CI = 70.7% to 87.0%) among three-dose, 83.8% (95% CI = 19.5% to 99.2%) among two-dose, and 82.1% (95% CI = 40.2% to 97.0%) among single-dose women. HPV16 or 18 antibody levels did not qualitatively decline between years four and 11 regardless of the number of doses given, although one-dose titers continue to be statistically significantly lower compared with two- and three-dose titers. CONCLUSION More than a decade after HPV vaccination, single-dose VE against HPV16 or 18 infection remained high and HPV16 or 18 antibodies remained stable. A single dose of bivalent HPV vaccine may induce sufficiently durable protection that obviates the need for more doses.
Collapse
Affiliation(s)
| | | | - Carolina Porras
- Agencia Costarricense de Investigaciones Biomédicas (ACIB), Formerly Proyecto Epidemiológico Guanacaste, Fundación INCIENSA, San José, Costa Rica
| | | | - Troy Kemp
- HPV Immunology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Rolando Herrero
- Early Detection and Prevention Section, International Agency for Research on Cancer, Lyon, France
| | - Sarah Wagner
- National Cancer Institute, NIH, Bethesda, MD, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD, USA
| | - Joseph Boland
- National Cancer Institute, NIH, Bethesda, MD, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD, USA
| | | | | | | | - David Roberson
- National Cancer Institute, NIH, Bethesda, MD, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD, USA
| | | | | | | | | | - Bernal Cortes
- Agencia Costarricense de Investigaciones Biomédicas (ACIB), Formerly Proyecto Epidemiológico Guanacaste, Fundación INCIENSA, San José, Costa Rica
| | | | | | - Paula Gonzalez
- Agencia Costarricense de Investigaciones Biomédicas (ACIB), Formerly Proyecto Epidemiológico Guanacaste, Fundación INCIENSA, San José, Costa Rica
| | - Ligia A Pinto
- HPV Immunology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | |
Collapse
|
49
|
Costa AP, Giraldo PC, Cobucci RN, Consolaro ML, Souza RP, Canário LB, Machado PR, Martins RR, Baptista PV, Jr JE, Gonçalves AK. Cross-Protective IgG and IgA Antibodies against Oncogenic and Non-Oncogenic HPV Genotypes. Asian Pac J Cancer Prev 2020; 21:2799-2804. [PMID: 32986383 PMCID: PMC7779425 DOI: 10.31557/apjcp.2020.21.9.2799] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 09/26/2020] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE The aim of the study was to describe the course of IgG/IgA immune response in women immunized with bivalent vaccine and in women non-vaccinated with HPV infection, as well as evaluating the cross-protection against non-vaccine HPV types. METHODS Serum and cervical mucus samples were collected from infected and vaccinated women for HPV detection/genotyping and for detection of IgG/IgA anti-HPV/VLP (Virus-like Particles) by ELISA. RESULTS The median absorbance detected in serum samples for anti-HPV-IgG antibodies was higher in vaccinated women when compared to HPV infected women (p <0.01), however, the median absorbance in cervical mucus samples for anti-HPV-IgA was higher in infected women when compared to vaccinated women (p <0.01). Additionally, our analyses also provided additional evidence for cross-protective efficacy of the HPV-16/18 vaccine against HPV-82, -6, -11, -13, -61, -72 and -74. CONCLUSION The IgG antibodies were significantly more detected in the serum of vaccinated women, while the IgA was found in greater quantities in cervical samples from those infected by the virus. In addition, there is evidence that the bivalent vaccine provides cross-protection against other non-oncogenic viral subtypes. .
Collapse
Affiliation(s)
- Ana Paula Costa
- Postgraduate Program in Health Sciences, Federal University of Rio Grande do Norte, Natal, Brazil.
| | - Paulo César Giraldo
- Department of Gynecology, and Obstetrics, State University of Campinas, Campinas, Brazil.
| | | | - Márcia Lopes Consolaro
- Clinical Cytology and STD Laboratory, Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, PR, Brazil.
| | - Raquel Pantarotto Souza
- Clinical Cytology and STD Laboratory, Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, PR, Brazil.
| | - Luanda Barbara Canário
- Department of Clinical Analysis and Toxicology, Federal University of Rio Grande do Norte, Natal, Brazil.
| | - Paula Renata Machado
- Department of Clinical Analysis and Toxicology, Federal University of Rio Grande do Norte, Natal, Brazil.
| | - Rand Randall Martins
- Department of Pharmacy, Federal University of Rio Grande do Norte, Natal, Brazil.
| | - Pedro Vieira Baptista
- Hospital Lusíadas Porto and Unidade de Tracto Genital Inferior, Serviço de Ginecologia e Obstetrícia, Centro Hospitalar de São João, Porto, Portugal.
| | - José Eleutério Jr
- Department of Female, Child and Adolescent Health, Federal University of Ceará, Fortaleza, Brazil.
| | - Ana Katherine Gonçalves
- Postgraduate Program in Health Sciences, Federal University of Rio Grande do Norte, Natal, Brazil.
| |
Collapse
|
50
|
Burger EA, Portnoy A, Campos NG, Sy S, Regan C, Kim JJ. Choosing the optimal HPV vaccine: The health impact and economic value of the nonavalent and bivalent HPV vaccines in 48 Gavi-eligible countries. Int J Cancer 2020; 148:932-940. [PMID: 32706907 DOI: 10.1002/ijc.33233] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 07/06/2020] [Accepted: 07/15/2020] [Indexed: 01/03/2023]
Abstract
The human papillomavirus (HPV) vaccines may provide some level of cross-protection against high-risk HPV genotypes not directly targeted by the vaccines. We evaluated the long-term health and economic impacts of routine HPV vaccination using either the nonavalent HPV vaccine or the bivalent HPV vaccine in the context of 48 Gavi-eligible countries. We used a multi-modeling approach to compare the bivalent with or without cross-protection and the nonavalent HPV vaccine. The optimal, that is, most cost-effective, vaccine was the vaccine with an incremental cost-effectiveness ratio below the per-capita gross domestic product (GDP) for each country. By 2100 and assuming 70% HPV vaccination coverage, a bivalent vaccine without cross-protection, a bivalent vaccine with favorable cross-protection and the nonavalent vaccine were projected to avert 14.9, 17.2 and 18.5 million cumulative cases of cervical cancer across all 48 Gavi-eligible countries, respectively. The relative value of the bivalent vaccine compared to the nonavalent vaccine increased assuming a bivalent vaccine conferred high cross-protection. For example, assuming a cost-effectiveness threshold of per-capita GDP, the nonavalent vaccine was optimal in 83% (n = 40) of countries if the bivalent vaccine did not confer cross-protection; however, the proportion of countries decreased to 63% (n = 30) if the bivalent vaccine conferred high cross-protection. For lower cost-effectiveness thresholds, the bivalent vaccine was optimal in a greater proportion of countries, under both cross-protection assumptions. Although the nonavalent vaccine is projected to avert more cases of cervical cancer, the bivalent vaccine with favorable cross-protection can prevent a considerable number of cases and would be considered a high-value vaccine for many Gavi-eligible countries.
Collapse
Affiliation(s)
- Emily A Burger
- Harvard T.H. Chan School of Public Health, Center for Health Decision Science, Boston, Massachusetts, USA.,Department of Health Management and Health Economics, University of Oslo, Oslo, Norway
| | - Allison Portnoy
- Harvard T.H. Chan School of Public Health, Center for Health Decision Science, Boston, Massachusetts, USA
| | - Nicole G Campos
- Harvard T.H. Chan School of Public Health, Center for Health Decision Science, Boston, Massachusetts, USA
| | - Stephen Sy
- Harvard T.H. Chan School of Public Health, Center for Health Decision Science, Boston, Massachusetts, USA
| | - Catherine Regan
- Harvard T.H. Chan School of Public Health, Center for Health Decision Science, Boston, Massachusetts, USA
| | - Jane J Kim
- Harvard T.H. Chan School of Public Health, Center for Health Decision Science, Boston, Massachusetts, USA
| |
Collapse
|