1
|
do Nascimento GR, da Silva Santos AC, Silva NNT, Guilmarães NS, Lima AA, Coura-Vital W. Prevalence of non-vaccine high-risk HPV cervical infections in vaccinated women: a systematic review and meta-analysis. BMC Infect Dis 2025; 25:131. [PMID: 39875836 PMCID: PMC11773943 DOI: 10.1186/s12879-025-10520-6] [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: 06/13/2024] [Accepted: 01/17/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Human Papillomavirus (HPV) is the most common sexually transmitted lower genital tract infection worldwide and the main etiological factor of cervical cancer (CC). Since 2006, vaccines have been implemented to reduce CC-related morbidity and mortality. This systematic review and meta-analysis aimed to evaluate the prevalence of cervical infections by non-vaccine high-risk HPV (HR-HPV) types in women vaccinated against types 16 and 18. METHOD This systematic review and meta-analysis used independent electronic databases - Lilacs, WHO, BDENF, State Department of Health SP, Health Information Locator, IRIS, Coleciona Sistema Único de Saúde, BINACIS, IBECS, CUMED and SciELO, on July 14, 2023. Observational studies that evaluated vaccinated and unvaccinated women against HR-HPV and the prevalence of cervical infection by types of HR-HPV were included. Intervention effects were expressed as prevalence ratios (PR). Forest plots were used to visualize vaccination effects. The study protocol was previously registered in PROSPERO, under code CRD42023440610. RESULTS Of the 7,051 studies, 31 met the analysis criteria. A total of 59,035 women were eligible for this systematic review. The results showed a high prevalence of non-vaccine HR-HPV types, regardless of vaccination status. For HPV 31/33/45 (PR = 0.60 [0.40-0.91]), HPV31 (PR = 0.47 [0.31-0.72]), and HPV 45 (PR = 0.38 [0.22-0.69]), a positive random effect was found. CONCLUSION The prevalence of non-vaccine HR-HPV cervical infection was high in women, regardless of vaccination status. For HPV types 31 and 45 and 31/33/45, the prevalence was lower in vaccinated women, suggesting a cross-protective effect of vaccines for these viral types.
Collapse
Affiliation(s)
- Glauciane Resende do Nascimento
- Programa de Pós-graduação em Ciências Farmacêuticas, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Ana Carolina da Silva Santos
- Programa de Pós-graduação em Ciências Farmacêuticas, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | | | | | - Angélica Alves Lima
- Programa de Pós-graduação em Ciências Farmacêuticas, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
- Departamento de Análises Clínicas, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Wendel Coura-Vital
- Programa de Pós-graduação em Ciências Farmacêuticas, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil.
- Departamento de Análises Clínicas, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Brazil.
| |
Collapse
|
2
|
Téblick L, Lipovac M, Molenberghs F, Delputte P, De Vos WH, Vorsters A. HPV-specific antibodies in female genital tract secretions captured via first-void urine retain their neutralizing capacity. Hum Vaccin Immunother 2024; 20:2330168. [PMID: 38567541 PMCID: PMC10993920 DOI: 10.1080/21645515.2024.2330168] [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: 02/15/2024] [Accepted: 03/11/2024] [Indexed: 04/04/2024] Open
Abstract
Human papillomavirus (HPV) vaccines, primarily relying on neutralizing antibodies, have proven highly effective. Recently, HPV-specific antibodies have been detected in the female genital tract secretions captured by first-void urine (FVU), offering a minimally invasive diagnostic approach. In this study, we investigated whether HPV16-specific antibodies present in FVU samples retain their neutralizing capacity by using pseudovirion-based neutralization assays. Paired FVU and serum samples (vaccinated n = 25, unvaccinated n = 25, aged 18-25) were analyzed using two orthogonal pseudovirion-based neutralization assays, one using fluorescence microscopy and the other using luminescence-based spectrophotometry. Results were compared with HPV16-specific IgG concentrations and correlations between neutralizing antibodies in FVU and serum were explored. The study demonstrated the presence of neutralizing antibodies in FVU using both pseudovirion-based neutralization assays, with the luminescence-based assay showing higher sensitivity for FVU samples, while the fluorescence microscopy-based assay exhibited better specificity for serum and overall higher reproducibility. High Spearman correlation values were calculated between HPV16-IgG and HPV16-neutralizing antibodies for both protocols (rs: 0.54-0.94, p < .001). Significant Spearman correlations between FVU and serum concentrations were also established for all assays (rs: 0.44-0.91, p < .01). This study demonstrates the continued neutralizing ability of antibodies captured with FVU, supporting the hypothesis that HPV vaccination may reduce autoinoculation and transmission risk to the sexual partner. Although further protocol optimizations are warranted, these findings provide a foundation for future research and larger cohort studies that could have implications for the optimal design, evaluation, and implementation of HPV vaccination programs.
Collapse
Affiliation(s)
- Laura Téblick
- Centre for the Evaluation of Vaccination, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Marijana Lipovac
- Centre for the Evaluation of Vaccination, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Freya Molenberghs
- Laboratory of Cell Biology and Histology, University of Antwerp, Antwerp, Belgium
| | - Peter Delputte
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, Antwerp, Belgium
| | - Winnok H. De Vos
- Laboratory of Cell Biology and Histology, University of Antwerp, Antwerp, Belgium
- Antwerp Centre for Advanced Microscopy, University of Antwerp, Antwerp, Belgium
- µNEURO Centre of Research Excellence, University of Antwerp, Antwerp, Belgium
| | - Alex Vorsters
- Centre for the Evaluation of Vaccination, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
3
|
Huang B, Zhang D, Wang H, Li S, Guo J, Cui S, Chen X, Hong Y, Wu Z. Systemic administration of a potent mouse monoclonal antibody provides multisite protection against HPV16 infection. Int Immunopharmacol 2024; 143:113272. [PMID: 39353396 DOI: 10.1016/j.intimp.2024.113272] [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: 03/17/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
Although preventive vaccines for Human Papillomaviruses (HPV) are available, a definitive cure for the viral infection itself is currently lacking. There is a sizable population that remains inaccessible to HPV vaccination due to reasons such as high costs or lack of availability of the vaccines. Therefore, there remains a significant population susceptible to HPV infection. Persistent multisite infections with high-risk HPV types can cause cancer at several different anatomic sites.Especially HPV16 is a key etiologic factor for cervical, other ano-genital and oropharyngeal cancers. Therefore, it is imperative to develop pharmaceutical interventions for the treatment of viral infections. In this study, a panel of 9 neutralizing antibodies was screened using the hybridoma technique, with 20F6 being identified as the representative antibody. The purified 20F6 exhibited an IC50 of 0.0011 μg/ml against HPV16, demonstrating potent viral inhibitory activity. Moreover, it displayed cross-neutralizing efficacy towards other Alphapapillom 9 subtypes including HPV31, HPV33, HPV52, and HPV58 with respective IC50 values of 2.0 μg/ml, 7.3 μg/ml, 1.7 μg/ml, and 3.0 μg/ml. 20F6 recognizes the linear epitope MSLW, the first four amino-acids located at the very N-terminus of the HPV16 L1 protein. Administration of 20F6, 24 h prior to and following HPV16 pseudo-virion (PSV) challenge, conferred protection against infection in mice at doses as low as 1 mg/kg. Following intraperitoneal administration of 20F6, neutralizing antibodies were consistently detected at both oral and vaginal sites, indicating that prophylactic systemic administration of 20F6 may confer efficient protection against multiple susceptible mucosal sites.
Collapse
Affiliation(s)
- Bilian Huang
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Doudou Zhang
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Hongyun Wang
- Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Siyu Li
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Jianjun Guo
- Yancheng Clinical College, Xuzhou Medical University, Yancheng, China
| | - Shengwei Cui
- School of Life Sciences, Ningxia University, Yinchuan, China
| | - Xiaoping Chen
- Yancheng Clinical College, Xuzhou Medical University, Yancheng, China.
| | - Ying Hong
- Nanjing International Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhiwei Wu
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China; Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
4
|
Travill DI, Machalek DA, Rees H, Mbulawa Z, Chikandiwa A, Munthali R, Petoumenos K, Kaldor JM, Delany-Moretlwe S. High prevalence of human papillomavirus (HPV) in unvaccinated adolescent girls in South Africa, particularly those living with HIV. Vaccine 2024; 42:126442. [PMID: 39423451 DOI: 10.1016/j.vaccine.2024.126442] [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: 06/05/2024] [Revised: 09/16/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
INTRODUCTION In 2014, South Africa implemented a national two-dose HPV vaccination programme using the bivalent vaccine for girls aged 9 years and older attending Grade 4 at public schools. We assessed HPV prevalence and risk factors among South African adolescent girls and young women (AGYW) aged 17-18 years who were ineligible for vaccination. METHODS From June to December 2019, we surveyed AGYW aged 17-18 years attending primary care clinics in four South African provinces. Consenting participants completed a questionnaire, underwent HIV counselling and testing, and self-collected a vaginal swab for HPV testing. Samples were tested by Seegene AnyPlex™ II HPV28. We used summary statistics to describe the population characteristics and logistic regression to examine the association between risk factors and high-risk HPV detection. RESULTS 910 participants were screened, 900 enrolled, 896 had valid HPV results, and 819 were unvaccinated and included in this study. Of these, 248 (30.3 %) were living with HIV and 597 (72.9 %) reported ever having vaginal sex. Overall, 463 (56.5 %) had at least one high-risk HPV detected, and 177 (21.6 %) had HPV16/18 detected. AGYW living with HIV had a higher prevalence of any high-risk HPV (65.3 % vs 52.7 %, p < 0.001) and HPV 16/18 (29.4 % vs 18.2 %, p < 0.001) compared to those without HIV. Multiple infections were also more common in participants living with HIV, with three or more high-risk HPV types detected in 32.3 % compared with 15.4 % of those without HIV (p < 0.001). In multivariate analyses, HIV status (p < 0.001) and higher number of lifetime sexual partners (p-trend<0.001) were associated with high-risk HPV detection. CONCLUSIONS High-risk HPV was very common in unvaccinated South Africa AGYW, especially among those living with HIV, highlighting the importance of HPV vaccination in settings with high HIV prevalence.
Collapse
Affiliation(s)
- Danielle I Travill
- Wits RHI, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Dorothy A Machalek
- Kirby Institute, University of New South Wales-Kensington Campus, Sydney, New South Wales, Australia; Centre for Women's Infectious Diseases, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Helen Rees
- Wits RHI, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Zizipho Mbulawa
- UCT-MRC Clinical Gynaecological Cancer Research Centre, University of Cape Town, Rondebosch, Western Cape, South Africa; Department of Laboratory Medicine and Pathology, Walter Sisulu University, Mthatha, Eastern Cape, South Africa
| | - Admire Chikandiwa
- Wits RHI, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Richard Munthali
- Wits RHI, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Kathy Petoumenos
- Kirby Institute, University of New South Wales-Kensington Campus, Sydney, New South Wales, Australia
| | - John M Kaldor
- Kirby Institute, University of New South Wales-Kensington Campus, Sydney, New South Wales, Australia
| | | |
Collapse
|
5
|
Zha R, Liao C, Lin D, Zhao L, Chen Y, Yao L, Li X, Yi B, Li T, Xiao J, Hu Y, Chen Z, Guo C, Lu J, Lu J. A Comprehensive Evaluation of the HPV Neutralizing Antibodies in Guangzhou, China: A Comparative Study on Various HPV Vaccines. Vaccines (Basel) 2024; 12:1286. [PMID: 39591188 PMCID: PMC11599073 DOI: 10.3390/vaccines12111286] [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: 10/14/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND The evaluation of HPV vaccine effectiveness is essential for informing public health strategies, yet there remains a gap in understanding humoral immune responses generated by different HPV vaccine formulations in regional populations. This study addresses this gap by evaluating the immunogenicity of the newly developed HPV vaccine Cecolin (Wantai), alongside various imported vaccines, including bivalent, quadrivalent, and nonavalent options available in China. METHODS From March 2023 to June 2024, a total of 352 participants were enrolled, including 87 females aged 9-14 years who received two doses of the bivalent HPV vaccine (Cecolin), 215 females aged 15-45 years who were fully vaccinated with various HPV vaccines, and 50 non-recipients. Follow-up assessments were conducted at six timepoints during the administration of Cecolin. Serum was collected at enrollment and at each follow-up visit for antibody assessments using a pseudovirion-based neutralization assay (PBNA). FINDINGS The longitudinal follow-up of females aged 9-14 years revealed a 100% conversion rate for neutralizing antibodies against HPV types 16 and 18 after the second dose, compared to 94.3% and 97.1% conversion rates six months after the first dose. Compared to participants who received full doses of quadrivalent and nonavalent vaccines, females who received two or three doses of Cecolin exhibited higher neutralizing antibody geometric mean titers (GMTs) and non-vaccine-type (HPV31 and HPV33) antibody seroconversion rates. INTERPRETATION The domestically produced HPV vaccine Cecolin in China demonstrates strong immunogenicity and holds promise for the large-scale vaccination of females in developing countries to prevent cervical cancer.
Collapse
Affiliation(s)
- Renyun Zha
- School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China (Z.C.); (C.G.)
- One Health Center of Excellence for Research & Training, Sun Yat-Sen University, Guangzhou 510080, China
| | - Conghui Liao
- School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China (Z.C.); (C.G.)
- One Health Center of Excellence for Research & Training, Sun Yat-Sen University, Guangzhou 510080, China
| | - Daner Lin
- Guangzhou Baiyun District Center for Disease Control and Prevention, Guangzhou 510445, China
| | - Lixuan Zhao
- Guangzhou Baiyun District Center for Disease Control and Prevention, Guangzhou 510445, China
| | - Yanfang Chen
- Guangzhou Baiyun District Center for Disease Control and Prevention, Guangzhou 510445, China
| | - Lin Yao
- Guangzhou Baiyun District Center for Disease Control and Prevention, Guangzhou 510445, China
| | - Xiaokang Li
- School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China (Z.C.); (C.G.)
- One Health Center of Excellence for Research & Training, Sun Yat-Sen University, Guangzhou 510080, China
| | - Boyang Yi
- Translational Cancer Research Center, Peking University First Hospital, Beijing 100034, China
| | - Ting Li
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325000, China;
| | - Jianpeng Xiao
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, China
| | - Yan Hu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Zeliang Chen
- School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China (Z.C.); (C.G.)
| | - Cheng Guo
- School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China (Z.C.); (C.G.)
| | - Jianyun Lu
- Guangzhou Baiyun District Center for Disease Control and Prevention, Guangzhou 510445, China
| | - Jiahai Lu
- School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China (Z.C.); (C.G.)
- One Health Center of Excellence for Research & Training, Sun Yat-Sen University, Guangzhou 510080, China
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325000, China;
- National Medical Products Administration Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Guangzhou 510080, China
- Hainan Key Novel Thinktank “Hainan Medical University ‘One Health’ Research Center”, Haikou 571199, China
- Research Institute of Sun Yat-Sen University in Shenzhen, Shenzhen 518057, China
- Key Laboratory of Tropical Diseases Control, Sun Yat-Sen University, Ministry of Education, Guangzhou 510080, China
- Institute of One Health, Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
6
|
Bei L, Gao S, Zhao D, Kou Y, Liang S, Wu Y, Zhang X, Meng D, Lu J, Luo C, Li X, Wang Y, Qiu H, Xie L. Immunogenicity Assessment of a 14-Valent Human Papillomavirus Vaccine Candidate in Mice. Vaccines (Basel) 2024; 12:1262. [PMID: 39591165 PMCID: PMC11599024 DOI: 10.3390/vaccines12111262] [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: 09/30/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Cervical cancer ranks as the fourth most common cancer affecting women globally, with HPV as the primary etiology agent. Prophylactic HPV vaccines have substantially reduced the incidence of cervical cancer. METHODS This study assessed the immunogenicity of SCT1000, a 14-valent recombinant virus-like particle (VLP) vaccine developed by Sinocelltech, Ltd. using pseudovirion-based neutralization assays (PBNAs) and total IgG Luminex immunoassays (LIAs). Currently in phase III clinical trials in China, SCT1000 targets the same HPV types as Gardasil 9®, plus five additional high-risk types, thereby covering twelve high-risk HPV types implicated in 96.4% of cervical cancer cases. RESULTS In murine models, a dose of 1.85 μg per mouse was identified as optimal for evaluating SCT1000's immunogenicity in a three-dose regimen, as measured by PBNA and total IgG LIA across all 14 HPV types. SCT1000 induced high levels of protective antibodies, which were sustained for at least four months following the third dose. The vaccine also demonstrated stable and consistent immunogenicity in mouse potency assays under both long-term and accelerated conditions. Additionally, our studies revealed a strong correlation between the two serological tests used. CONCLUSIONS SCT1000 elicited robust, durable, and consistent humoral immune responses across all 14 HPV types, indicating its potential as a broad-spectrum vaccine candidate against HPV types 6/11/16/18/31/33/35/39/45/51/52/56/58/59. The significant correlations observed between PBNA and total IgG LIA support the use of the Luminex-based total IgG method as a reliable and effective alternative for immunogenicity assessment in preclinical and future clinical vaccine development.
Collapse
Affiliation(s)
- Lei Bei
- Heilongjiang Pharmaceutical Research Institute, Jiamusi University, Jiamusi 154007, China; (L.B.); (H.Q.)
| | - Shuman Gao
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (S.G.)
| | - Dandan Zhao
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (S.G.)
| | - Yajuan Kou
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (S.G.)
| | - Siyu Liang
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (S.G.)
| | - Yurong Wu
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (S.G.)
| | - Xiao Zhang
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (S.G.)
| | - Dan Meng
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (S.G.)
| | - Jianbo Lu
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (S.G.)
| | - Chunxia Luo
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (S.G.)
| | - Xuefeng Li
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (S.G.)
| | - Yang Wang
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (S.G.)
| | - Hongbin Qiu
- Heilongjiang Pharmaceutical Research Institute, Jiamusi University, Jiamusi 154007, China; (L.B.); (H.Q.)
| | - Liangzhi Xie
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (S.G.)
- Beijing Key Laboratory of Monoclonal Antibody Research and Development, Sino Biological Inc., Beijing 100176, China
- Cell Culture Engineering Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| |
Collapse
|
7
|
Liu Y, Ai H. Comprehensive insights into human papillomavirus and cervical cancer: Pathophysiology, screening, and vaccination strategies. Biochim Biophys Acta Rev Cancer 2024; 1879:189192. [PMID: 39349261 DOI: 10.1016/j.bbcan.2024.189192] [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: 06/13/2024] [Revised: 09/07/2024] [Accepted: 09/23/2024] [Indexed: 10/02/2024]
Abstract
This article provides an in-depth review of the Human Papillomavirus (HPV), a predominant etiological factor in cervical cancer, exploring its pathophysiology, epidemiology, and mechanisms of oncogenesis. We examine the role of proteins, DNA methylation markers, and non-coding RNAs as predictive biomarkers in cervical cancer, highlighting their potential in refining diagnostic and prognostic practices. The evolution and efficacy of cervical cancer screening methods, including the Papanicolaou smear, HPV testing, cytology and HPV test, and colposcopy techniques, are critically analyzed. Furthermore, the article delves into the current landscape and future prospects of prophylactic HPV vaccines and therapeutic vaccines, underscoring their significance in the prevention and potential treatment of HPV-related diseases. This comprehensive review aims to synthesize recent advances and ongoing challenges in the field, providing a foundation for future research and clinical strategies in the prevention and management of cervical cancer.
Collapse
Affiliation(s)
- Ying Liu
- Department of Gynaecology and Obstetrics, The Third Affiliated Hospital of Jinzhou Medical University; Liaoning Provincial Key Laboratory of Follicular Development and Reproductive Health, Jinzhou Medical University, No. 2, Section 5, Heping Road, Linghe District, Jinzhou City, Liaoning Province, 121000, P.R. China
| | - Hao Ai
- Department of Gynaecology and Obstetrics, The Third Affiliated Hospital of Jinzhou Medical University; Liaoning Provincial Key Laboratory of Follicular Development and Reproductive Health, Jinzhou Medical University, No. 2, Section 5, Heping Road, Linghe District, Jinzhou City, Liaoning Province, 121000, P.R. China.
| |
Collapse
|
8
|
Bao W, He X, Huang Y, Liu R, Li Z. The Clinical Effectiveness of Single-Dose Human Papillomavirus Vaccination. Vaccines (Basel) 2024; 12:956. [PMID: 39339988 PMCID: PMC11436243 DOI: 10.3390/vaccines12090956] [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: 06/17/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
The human papillomavirus (HPV) vaccine was initially approved for a three-dose regimen. Due to resource limitations, budget constraints, low acceptance, and poor adherence, global vaccination coverage is only 15%. A single-dose regimen could simplify logistics, reduce costs, and improve accessibility. However, its clinical effectiveness remains debatable. This review systematically searched PubMed, Embase, and Cochrane Library, including 42 clinical studies, to assess the effectiveness of a single-dose HPV vaccination for preventing HPV infections, cervical abnormalities, and genital warts. We summarized the effectiveness of bivalent, quadrivalent, and nonavalent vaccines across different age groups and buffer periods, and analyzed the factors contributing to the inconsistency of results. The review also provides insights into designing robust future research to inform single-dose HPV vaccination policies and guidelines, highlighting the need for further research to refine vaccination strategies.
Collapse
Affiliation(s)
- Wanying Bao
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Obstetrics and Gynecology, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Xinlin He
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Obstetrics and Gynecology, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yue Huang
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Obstetrics and Gynecology, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Rongyu Liu
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Obstetrics and Gynecology, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Zhengyu Li
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Obstetrics and Gynecology, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
9
|
Bentz JL, Barney RE, Georgantzoglou N, Manxhuka-Kerliu S, Ibishi VA, Dacaj-Elshani B, Bejarano S, Palumbo PE, Suresh A, LaRochelle EPM, Keegan WP, Wilson TL, Dokus BJ, Hershberger KC, Gallagher TL, Allen SF, Palisoul SM, Steinmetz HB, Kennedy LS, Tsongalis GJ. High-Risk HPV Screening Initiative in Kosovo-A Way to Optimize HPV Vaccination for Cervical Cancer. Diseases 2024; 12:189. [PMID: 39195188 DOI: 10.3390/diseases12080189] [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: 05/29/2024] [Revised: 07/06/2024] [Accepted: 07/16/2024] [Indexed: 08/29/2024] Open
Abstract
Nearly all cervical cancers are caused by persistent high-risk human papillomavirus (hrHPV) infection. There are 14 recognized hrHPV genotypes (HPV 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59, 66, and 68), and hrHPV genotypes 16 and 18 comprise approximately 66% of all cases worldwide. An additional 15% of cervical cancers are caused by hrHPV genotypes 31, 33, 45, 52, and 58. Screening patients for hrHPV as a mechanism for implementation of early treatment is a proven strategy for decreasing the incidence of HPV-related neoplasia, cervical cancer in particular. Here, we present population data from an HPV screening initiative in Kosovo designed to better understand the prevalence of the country's HPV burden and local incidence of cervical cancer by hrHPV genotype. Nearly 2000 women were screened for hrHPV using a real-time polymerase chain reaction (real-time PCR) assay followed by melt curve analysis to establish the prevalence of hrHPV in Kosovo. Additionally, DNA was extracted from 200 formalin-fixed, paraffin embedded cervical tumors and tested for hrHPV using the same method. Cervical screening samples revealed a high prevalence of hrHPV genotypes 16 and 51, while cervical cancer specimens predominantly harbored genotypes 16, 18, and 45. This is the first comprehensive screening study for evaluating the prevalence of hrHPV genotypes in Kosovo on screening cervical brush samples and cervical neoplasms. Given the geographic distribution of hrHPV genotypes and the WHO's global initiative to eliminate cervical cancer, this study can support and direct vaccination efforts to cover highly prevalent hrHPV genotypes in Kosovo's at-risk population.
Collapse
Affiliation(s)
- Jessica L Bentz
- Dartmouth Hitchcock Medical Center, Department of Pathology and Laboratory Medicine, Lebanon, NH 03756, USA
- Theodore and Audrey Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
| | - Rachael E Barney
- Dartmouth Hitchcock Medical Center, Department of Pathology and Laboratory Medicine, Lebanon, NH 03756, USA
| | - Natalia Georgantzoglou
- Dartmouth Hitchcock Medical Center, Department of Pathology and Laboratory Medicine, Lebanon, NH 03756, USA
| | | | | | | | | | - Paul E Palumbo
- Dartmouth Hitchcock Medical Center, Department of Medicine, Lebanon, NH 03756, USA
| | | | - Ethan P M LaRochelle
- Dartmouth Hitchcock Medical Center, Department of Pathology and Laboratory Medicine, Lebanon, NH 03756, USA
| | - William P Keegan
- Dartmouth Hitchcock Medical Center, Department of Pathology and Laboratory Medicine, Lebanon, NH 03756, USA
| | - Teresa L Wilson
- Dartmouth Hitchcock Medical Center, Department of Pathology and Laboratory Medicine, Lebanon, NH 03756, USA
| | - Betty J Dokus
- Dartmouth Hitchcock Medical Center, Department of Pathology and Laboratory Medicine, Lebanon, NH 03756, USA
| | - Kenneth C Hershberger
- Dartmouth Hitchcock Medical Center, Department of Pathology and Laboratory Medicine, Lebanon, NH 03756, USA
| | - Torrey L Gallagher
- Dartmouth Hitchcock Medical Center, Department of Pathology and Laboratory Medicine, Lebanon, NH 03756, USA
| | - Samantha F Allen
- Dartmouth Hitchcock Medical Center, Department of Pathology and Laboratory Medicine, Lebanon, NH 03756, USA
| | - Scott M Palisoul
- Dartmouth Hitchcock Medical Center, Department of Pathology and Laboratory Medicine, Lebanon, NH 03756, USA
| | - Heather B Steinmetz
- Dartmouth Hitchcock Medical Center, Department of Pathology and Laboratory Medicine, Lebanon, NH 03756, USA
| | | | - Gregory J Tsongalis
- Dartmouth Hitchcock Medical Center, Department of Pathology and Laboratory Medicine, Lebanon, NH 03756, USA
- Theodore and Audrey Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
- Dartmouth Cancer Center, Lebanon, NH 03756, USA
| |
Collapse
|
10
|
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
|
11
|
Dias Neto NM, Moura Dias VGN, Christofolini DM. Is syphilis infection a risk factor for cervicovaginal HPV occurrence? A case-control study. J Infect Public Health 2024; 17:102472. [PMID: 38901118 DOI: 10.1016/j.jiph.2024.102472] [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: 01/23/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Syphilis and human papillomavirus (HPV) are sexually transmitted infections affecting women in the same risk group. Thus, the main objective of the present study was to investigate the prevalence of HPV in a population of women with and without syphilis and observe the characteristics of HPV cervical lesions when coinfection occurs. Sociodemographic factors associated with coinfection were also evaluated. METHODS This case-control study was conducted at a Brazilian HIV/STD testing and training center. Study groups were composed of women with (case) and without syphilis (control), paired by age. The presence of HPV, HPV subtype, and lesion severity were investigated. All women were subjected to a sociodemographic interview, clinical data collection, cell collection for cytopathological analysis, and a hybrid capture test for HPV diagnosis. The chi-square test was used for statistical analysis. RESULTS The sample consisted of 176 women, 88 in each group. The prevalence of HPV was 14.8 % in the case (n = 13) and 18.1 % in the control group (n = 16), and there was no statistically significant difference between them. Illiterate individuals were more prevalent in the control group (p = 0.023). Considering women with suggestive signs of STIs, 30 % (6) of the patients and controls had high-risk HPV, and 15 % (3) had coinfection. The cytopathological assessment showed no differences between the groups concerning cellular atypia. However, ASC-US and ASC-H (atypical squamous cells of undetermined significance and high-grade) were only found in women with coinfections, with 75 % of these patients testing positive for high-risk HPV. Considering the distribution of lesions on the cervix, the HSIL (high-grade intraepithelial lesion) was assessed in high-risk HPV patients, both cases and controls. CONCLUSIONS The prevalence of HPV was not increased in patients infected with syphilis. In addition, coinfection does not seem to be an aggravating factor for the presence of precursor lesions of cervical cancer.
Collapse
Affiliation(s)
| | | | - Denise Maria Christofolini
- Postgraduate Program of Health Sciences, Centro Universitário FMABC, Santo André, SP, Brazil; Genetics Laboratory, Instituto Ideia Fértil de Saúde Reprodutiva, Instituto Idéia, São Paulo, SP, Brazil
| |
Collapse
|
12
|
Gao S, Zhao D, Feng C, Kou Y, Lu J, Luo C, Li X, Wang Y, Xie L. Validation of a triple-color pseudovirion-based neutralization assay for immunogenicity assessment of a 14-valent recombinant human papillomavirus vaccine. J Med Virol 2024; 96:e29859. [PMID: 39145587 DOI: 10.1002/jmv.29859] [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: 12/05/2023] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 08/16/2024]
Abstract
Validation of bioanalytical methods is crucial, especially in the pharmaceutical industry, to determine their suitability for specific purposes and the accuracy of analytical results. The pseudovirion-based neutralization assay (PBNA) is considered the gold standard for detecting and quantifying neutralizing antibodies against human papillomavirus in vaccine development for disease prevention. This paper introduces an improved triple-color PBNA method, capable of simultaneous detection of two or three human papillomavirus (HPV types for use in the development of a 14-valent HPV vaccine candidate. The primary objective was to comprehensively validate the triple-color PBNA method for general vaccine immunogenicity assays. Results show that the method has good specificity, accuracy, precision, linearity, robustness, and applicability. This innovative triple-color PBNA offers an improved approach for large-scale immunogenicity assessment in vaccine development. This study lays a solid foundation that can serve as a guiding paradigm for assessing vaccine responses in preclinical and clinical phases, providing valuable insights to the field.
Collapse
Affiliation(s)
- Shuman Gao
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Dandan Zhao
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Caixia Feng
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Yajuan Kou
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Jianbo Lu
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Chunxia Luo
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Xuefeng Li
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Yang Wang
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Liangzhi Xie
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
- Beijing Key Laboratory of Monoclonal Antibody Research and Development, Sino Biological Inc., Beijing, China
- Cell Culture Engineering Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
13
|
Andijani O, Alsalhi S. The Non-inferiority of Human Papillomavirus Vaccine Immunogenicity Among Women Over 26 Years: A Systematic Review. Cureus 2024; 16:e65157. [PMID: 39176354 PMCID: PMC11339578 DOI: 10.7759/cureus.65157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2024] [Indexed: 08/24/2024] Open
Abstract
OBJECTIVE Demonstrate the immunogenicity of the human papillomavirus (HPV) vaccine for the older age group (above 26 years) to prevent HPV infection with high-risk types and argue for extending vaccination recommendations for the older age group. METHODS Two authors searched PubMed, Embase, and the Cochrane Library from inception to December 2023 to collect information on clinical trials of HPV vaccine immunogenicity. The database search strategy used a combination of subject terms and free terms. Two authors first identified studies by reading the title, abstract, and full texts and, subsequently, based on the inclusion criteria. Studies eligible to be included are the clinical trials using one of the following types of HPV vaccines: 2vHPV, 4vHPV, and 9vHPV, and measuring the immunogenicity by the geometric mean concentration or titer (GMC/T) and seroconversion rate (SCR) among healthy women aged 9 to 55 years who had never received a prophylactic HPV vaccine, known serostatus for HPV, non-immunocompetence, or non-pregnant. RESULTS This review included nine articles, seven RCTs, and two open-labeled studies. CONCLUSION In summary, we have demonstrated that the immunogenicity of the HPV vaccines is non-inferior in the older age group. Even though the GMT declines with age, the SCR is similar in all age groups regardless of the serostatus. The immunogenicity of the bivalent vaccine is superior to that of the quadrivalent vaccine for the older age group. Additionally, the vaccine is more efficient in women under the age of 26, but older women will benefit from it.
Collapse
Affiliation(s)
| | - Sara Alsalhi
- Preventive Medicine, Saudi Ministry of Health, Jeddah, SAU
| |
Collapse
|
14
|
Ng K, Morais S, Wissing MD, Burchell AN, Tellier PP, Coutlée F, Waterboer T, El-Zein M, Franco EL. Empirical sample-specific approaches to define HPV16 and HPV18 seropositivity in unvaccinated, young, sexually active women. Microbiol Spectr 2024; 12:e0022924. [PMID: 38687066 PMCID: PMC11324019 DOI: 10.1128/spectrum.00229-24] [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: 01/24/2024] [Accepted: 04/09/2024] [Indexed: 05/02/2024] Open
Abstract
Given low seroconversion rates following human papillomavirus (HPV) infection, fixed external cutoffs may lead to errors in estimating HPV seroprevalence. We evaluated finite mixture modeling (FMM) and group-based trajectory modeling (GBTM) among unvaccinated, sexually active, HPV-exposed women to determine study-specific HPV16 and HPV18 seropositivity thresholds. We included 399 women (aged 18-24 years) enrolled in the HPV Infection and Transmission Among Couples Through Heterosexual Activity (HITCH) cohort study between 2005 and 2011 in Montreal, Canada. Participants' blood samples from up to six visits spanning 2 years were tested by multiplex serology for antibodies [median fluorescence intensity (MFI)] specific to bacterially expressed HPV16 and HPV18 L1 glutathione S-transferase fusion proteins. We applied FMM and GBTM to baseline and longitudinal antibody titer measurements, respectively, to define HPV type-specific seronegative and seropositive distributions. Study-specific thresholds were generated as five standard deviations above the mean seronegative antibody titers, mimicking cutoffs (HPV16: 422 MFI; HPV18: 394 MFI) derived from an external population of sexually inactive, HPV DNA-negative Korean women (aged 15-29 years). Agreement (kappa) of study-specific thresholds was evaluated against external cutoffs. Seroprevalence estimates using FMM (HPV16: 27.5%-43.2%; HPV18: 21.7%-49.5%) and GBTM (HPV16: 11.8%-11.8%; HPV18: 9.9%-13.4%) thresholds exceeded those of external cutoffs (HPV: 10.2%; HPV18: 9.7%). FMM thresholds showed slight-to-moderate agreement with external cutoffs (HPV16: 0.26%-0.46%; HPV18: 0.20%-0.56%), while GBTM thresholds exhibited high agreement (HPV16: 0.92%-0.92%; HPV18: 0.82%-0.99%). Kappa values suggest that GBTM, used for longitudinal serological data, and otherwise FMM, for cross-sectional data, are robust methods for determining the HPV serostatus without prior classification rules.IMPORTANCEWhile human papillomavirus (HPV) seropositivity has been employed as an epidemiologic determinant of the natural history of genital HPV infections, only a fraction of women incidentally infected with HPV respond by developing significant antibody levels. HPV seropositivity is often determined by a dichotomous fixed cutoff based on the seroreactivity of an external population of women presumed as seronegative, given the lack of evidence of HPV exposure. However, considering the variable nature of seroreactivity upon HPV infection, which arguably varies across populations, such externally defined cutoffs may lack specificity to the population of interest, causing inappropriate assessment of HPV seroprevalence and related epidemiologic uses of that information. This study demonstrates that finite mixture modeling (FMM) and group-based trajectory modeling (GBTM) can be used to independently estimate seroprevalence or serve as the basis for defining study-specific seropositivity thresholds without requiring prior subjective assumptions, consequently providing a more apt internally valid discrimination of seropositive from seronegative individuals.
Collapse
Affiliation(s)
- Kristy Ng
- Division of Cancer
Epidemiology, McGill University,
Montreal, Quebec,
Canada
| | - Samantha Morais
- Division of Cancer
Epidemiology, McGill University,
Montreal, Quebec,
Canada
| | - Michel D. Wissing
- Division of Cancer
Epidemiology, McGill University,
Montreal, Quebec,
Canada
| | - Ann N. Burchell
- Department of Family
and Community Medicine and MAP Centre for Urban Health Solutions, Li Ka
Shing Knowledge Institute, St. Michael’s Hospital, Unity Health
Toronto, Toronto,
Ontario, Canada
- Department of Family
and Community Medicine, Faculty of Medicine, University of
Toronto, Toronto,
Ontario, Canada
| | | | - François Coutlée
- Division of Cancer
Epidemiology, McGill University,
Montreal, Quebec,
Canada
- Laboratoire de
Virologie Moléculaire, Centre de recherche du Centre Hospitalier
de l'Université de
Montréal, Montreal,
Quebec, Canada
- Départements de
Microbiologie, Infectiologie et Immunologie, et de
Gynécologie‐Obstétrique, Université de
Montréal, Montreal,
Quebec, Canada
- Départements de
Médecine, de Médecine clinique de Laboratoire et
d'Obstétrique‐Gynécologie, Centre Hospitalier de
l'Université de Montréal,
Montreal, Quebec,
Canada
| | - Tim Waterboer
- Infections and Cancer
Epidemiology Division, German Cancer Research
Center, Heidelberg,
Germany
| | - Mariam El-Zein
- Division of Cancer
Epidemiology, McGill University,
Montreal, Quebec,
Canada
| | - Eduardo L. Franco
- Division of Cancer
Epidemiology, McGill University,
Montreal, Quebec,
Canada
| |
Collapse
|
15
|
Quang C, Chung AW, Kemp TJ, Ratu T, Tuivaga E, Russell FM, Licciardi PV, Toh ZQ. Development of a human papillomavirus (HPV) multiplex immunoassay to profile HPV antibodies. J Med Virol 2024; 96:e29732. [PMID: 38874202 DOI: 10.1002/jmv.29732] [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: 03/18/2024] [Revised: 04/29/2024] [Accepted: 06/01/2024] [Indexed: 06/15/2024]
Abstract
Neutralizing antibodies (NAbs) are considered the primary mechanism of vaccine-mediated protection against human papillomaviruses (HPV), the causative agent of cervical cancer. However, the minimum level of NAb needed for protection is currently unknown. The HPV pseudovirion-based neutralization assay (PBNA) is the gold standard method for assessing HPV antibody responses but is time-consuming and labor-intensive. With the development of higher valency HPV vaccines, alternative serological assays with the capacity for multiplexing would improve efficiency and output. Here we describe a multiplex bead-based immunoassay to characterize the antibody responses to the seven oncogenic HPV types (HPV16/18/31/33/45/52/58) contained in the current licensed nonavalent HPV vaccine. This assay can measure antibody isotypes and subclasses (total IgG, IgM, IgA1-2, IgG1-4), and can be adapted to measure other antibody features (e.g., Fc receptors) that contribute to vaccine immunity. When tested with serum samples from unvaccinated and vaccinated individuals, we found high concordance between HPV-specific IgG using this multiplex assay and NAbs measured with PBNA. Overall, this assay is high-throughput, sample-sparing, and time-saving, providing an alternative to existing assays for the measurement and characterization of HPV antibody responses.
Collapse
Affiliation(s)
- Chau Quang
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- Vaccine Immunology, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Amy W Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Troy J Kemp
- HPV Serology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Tupou Ratu
- Ministry of Health and Medical Services, Suva, Fiji
| | | | - Fiona M Russell
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- Vaccine Immunology, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Paul V Licciardi
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- Vaccine Immunology, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Zheng Q Toh
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- Vaccine Immunology, Murdoch Children's Research Institute, Parkville, VIC, Australia
| |
Collapse
|
16
|
Movahed F, Darzi S, Mahdavi P, Salih Mahdi M, Qutaiba B Allela O, Naji Sameer H, Adil M, Zarkhah H, Yasamineh S, Gholizadeh O. The potential use of therapeutics and prophylactic mRNA vaccines in human papillomavirus (HPV). Virol J 2024; 21:124. [PMID: 38822328 PMCID: PMC11143593 DOI: 10.1186/s12985-024-02397-9] [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: 02/07/2024] [Accepted: 05/23/2024] [Indexed: 06/02/2024] Open
Abstract
Cervical cancer (CC) and other malignant malignancies are acknowledged to be primarily caused by persistent human papillomavirus (HPV) infection. Historically, vaccinations against viruses that produce neutralizing antibodies unique to the virus have been an affordable way to manage viral diseases. CC risk is decreased, but not eliminated, by HPV vaccinations. Since vaccinations have been made available globally, almost 90% of HPV infections have been successfully avoided. On the lesions and diseases that are already present, however, no discernible treatment benefit has been shown. As a result, therapeutic vaccines that elicit immune responses mediated by cells are necessary for the treatment of established infections and cancers. mRNA vaccines possess remarkable potential in combating viral diseases and malignancy as a result of their superior industrial production, safety, and efficacy. Furthermore, considering the expeditiousness of production, the mRNA vaccine exhibits promise as a therapeutic approach targeting HPV. Given that the HPV-encoded early proteins, including oncoproteins E6 and E7, are consistently present in HPV-related cancers and pre-cancerous lesions and have crucial functions in the progression and persistence of HPV-related diseases, they serve as ideal targets for therapeutic HPV vaccines. The action mechanism of HPV and HPV-related cancer mRNA vaccines, their recent advancements in clinical trials, and the potential for their therapeutic applications are highlighted in this study, which also offers a quick summary of the present state of mRNA vaccines. Lastly, we highlight a few difficulties with mRNA HPV vaccination clinical practice and provide our thoughts on further advancements in this quickly changing sector. It is expected that mRNA vaccines will soon be produced quickly for clinical HPV prevention and treatment.
Collapse
Affiliation(s)
- Fatemeh Movahed
- Department of Gynecology and Obstetrics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Satinik Darzi
- Department Of Obstetrics and Gynecology, Abnormal Uterine Bleeding Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Parya Mahdavi
- Department of Obstetrics and Gynecology, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | | | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | - Mohaned Adil
- Pharmacy college, Al-Farahidi University, Baghdad, Iraq
| | - Hasna Zarkhah
- Department of Obstetrics and Gynaecology, Tabriz University of Medical Siences, Tabriz, Iran.
| | - Saman Yasamineh
- Young Researchers and Elite Club, Tabriz Branch, Islamic Azad University, Tabriz, Iran.
| | | |
Collapse
|
17
|
Yang ST, Wang PH, Liu HH, Chang WH, Chou FW, Lee WL. Cervical cancer: Part I human papilloma virus vaccination in Taiwan. Taiwan J Obstet Gynecol 2024; 63:320-328. [PMID: 38802194 DOI: 10.1016/j.tjog.2024.04.005] [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] [Accepted: 04/09/2024] [Indexed: 05/29/2024] Open
Abstract
A significant decline in both incidence and prevalence of cervical cancers after widespread-introducing cervical screening strategy by Papanicolau test (Pap test) has been found in the world, but cervical cancer is still one of the most common female cancers, reporting the fourth prevalence and also one of the leading causes to result in main women-associated morbidity and mortality, particularly for those women living in low- and middle-income countries. Cervical cancer is one of the most important health concerns directly destroying the global health-care system, partly because of not only increasing the disability either secondary to diseases themselves of victims or mediated by treatment-related adverse events to the survivors but also acting as a leading cause of death of diseased patients worldwide, alarming the urgent need to do something to minimize the catastrophic diseases-related heavy socioeconomic burden. It is fortunate that cervical cancer is a preventable disease, based on its strong association with human papillomavirus (HPV) infection (more than 95%), particularly for those high-risk HPV (HR-HPV) and its high possibility by detecting HPV infection before the development of cervical cancer as well as an effective prevention by HPV vaccination. That is why WHO (World Health Organization) considers cervical cancer as a public problem and attempts to accelerate the elimination of cervical cancer program by three-pillar approach (90:70:90% targets), including (1) 90% of girls are fully vaccinated with HPV vaccine by 15 years of age; (2) 70% of women are screened with a high-performance test by 35 and 45 years of age and precancerous lesions are treated early; and (3) 90% of women identified with cervical diseases receive appropriate and adequate treatment. Herein, this review focuses on the HPV vaccination as Part I, including global recommendations and Taiwan government's policy for HPV vaccination.
Collapse
Affiliation(s)
- Szu-Ting Yang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Peng-Hui Wang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Female Cancer Foundation, Taipei, Taiwan; Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.
| | - Hung-Hsien Liu
- Department of Medical Imaging and Intervention, Tucheng Hospital, New Taipei City, Taiwan
| | - Wen-Hsun Chang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Nursing, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Hospital and Health Care Administration, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Fang-Wei Chou
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Hospital and Health Care Administration, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wen-Ling Lee
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Medicine, Cheng-Hsin General Hospital, Taipei, Taiwan.
| |
Collapse
|
18
|
Rizarullah, Aditama R, Giri-Rachman EA, Hertadi R. Designing a Novel Multiepitope Vaccine from the Human Papilloma Virus E1 and E2 Proteins for Indonesia with Immunoinformatics and Molecular Dynamics Approaches. ACS OMEGA 2024; 9:16547-16562. [PMID: 38617694 PMCID: PMC11007845 DOI: 10.1021/acsomega.4c00425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/04/2024] [Accepted: 03/08/2024] [Indexed: 04/16/2024]
Abstract
One of the deadliest malignant cancer in women globally is cervical cancer. Specifically, cervical cancer is the second most common type of cancer in Indonesia. The main infectious agent of cervical cancer is the human papilloma virus (HPV). Although licensed prophylactic vaccines are available, cervical cancer cases are on the rise. Therapy using multiepitope-based vaccines is a very promising therapy for cervical cancer. This study aimed to develop a multiepitope vaccine based on the E1 and E2 proteins of HPV 16, 18, 45, and 52 using in silico. In this study, we develop a novel multiepitope vaccine candidate using an immunoinformatic approach. We predicted the epitopes of the cytotoxic T lymphocyte (CTL) and helper T lymphocyte (HTL) and evaluated their immunogenic properties. Population coverage analysis of qualified epitopes was conducted to determine the successful use of the vaccine worldwide. The epitopes were constructed into a multiepitope vaccine by using AAY linkers between the CTL epitopes and GPGPG linkers between the HTL epitopes. The tertiary structure of the multiepitope vaccine was modeled with AlphaFold and was evaluated by Prosa-web. The results of vaccine construction were analyzed for B-cell epitope prediction, molecular docking with Toll like receptor-4 (TLR4), and molecular dynamics simulation. The results of epitope prediction obtained 4 CTL epitopes and 7 HTL epitopes that are eligible for construction of multiepitope vaccines. Prediction of the physicochemical properties of multiepitope vaccines obtained good results for recombinant protein production. The interaction showed that the interaction of the multiepitope vaccine-TLR4 complex is stable based on the binding free energy value -106.5 kcal/mol. The results of the immune response simulation show that multiepitope vaccine candidates could activate the adaptive and humoral immune systems and generate long-term B-cell memory. According to these results, the development of a multiepitope vaccine with a reverse vaccinology approach is a breakthrough to develop potential cervical cancer therapeutic vaccines.
Collapse
Affiliation(s)
- Rizarullah
- Biochemistry
and Biomolecular Engineering Research Division, Faculty of Mathematics
and Natural Sciences, Bandung Institute
of Technology, Jl. Ganesa No. 10, Bandung 40132, Indonesia
- Department
of Biochemistry, Faculty of Medicine, Abulyatama
University, Jl. Blangbintang Lama, Aceh Besar 23372, Indonesia
| | - Reza Aditama
- Biochemistry
and Biomolecular Engineering Research Division, Faculty of Mathematics
and Natural Sciences, Bandung Institute
of Technology, Jl. Ganesa No. 10, Bandung 40132, Indonesia
| | - Ernawati Arifin Giri-Rachman
- Genetics
and Molecular Biotechnology Research Division, School of Life Sciences
and Technology, Bandung Institute of Technology, Jl. Ganesa No. 10, Bandung 40132, Indonesia
| | - Rukman Hertadi
- Biochemistry
and Biomolecular Engineering Research Division, Faculty of Mathematics
and Natural Sciences, Bandung Institute
of Technology, Jl. Ganesa No. 10, Bandung 40132, Indonesia
| |
Collapse
|
19
|
Kamuyu G, Coelho da Silva F, Tenet V, Schussler J, Godi A, Herrero R, Porras C, Mirabello L, Schiller JT, Sierra MS, Kreimer AR, Clifford GM, Beddows S. Global evaluation of lineage-specific human papillomavirus capsid antigenicity using antibodies elicited by natural infection. Nat Commun 2024; 15:1608. [PMID: 38383518 PMCID: PMC10881982 DOI: 10.1038/s41467-024-45807-w] [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: 07/31/2023] [Accepted: 02/05/2024] [Indexed: 02/23/2024] Open
Abstract
Human Papillomavirus (HPV) type variants have been classified into lineages and sublineages based upon their whole genome sequence. Here we have examined the specificity of antibodies generated following natural infection with lineage variants of oncogenic types (HPV16, 18, 31, 33, 45, 52 and 58) by testing serum samples assembled from existing archives from women residing in Africa, The Americas, Asia or Europe against representative lineage-specific pseudoviruses for each genotype. We have subjected the resulting neutralizing antibody data to antigenic clustering methods and created relational antigenic profiles for each genotype to inform the delineation of lineage-specific serotypes. For most genotypes, there was evidence of differential recognition of lineage-specific antigens and in some cases of a sufficient magnitude to suggest that some lineages should be considered antigenically distinct within their respective genotypes. These data provide compelling evidence for a degree of lineage specificity within the humoral immune response following natural infection with oncogenic HPV.
Collapse
Affiliation(s)
- Gathoni Kamuyu
- Virus Reference Department, Public Health Microbiology Division, UK Health Security Agency, London, UK
| | - Filomeno Coelho da Silva
- Virus Reference Department, Public Health Microbiology Division, UK Health Security Agency, London, UK
| | - Vanessa Tenet
- International Agency for Research on Cancer (IARC/WHO) Early Detection, Prevention and Infections Branch, Lyon, France
| | - John Schussler
- Information Management Services Inc, Silver Spring, MD, USA
| | - Anna Godi
- Virus Reference Department, Public Health Microbiology Division, UK Health Security Agency, London, UK
| | - Rolando Herrero
- Agencia Costarricense de Investigaciones Biomédicas (ACIB) formerly Proyecto Epidemiológico Guanacaste, Fundación INCIENSA (FUNIN), San José, Costa Rica
| | - Carolina Porras
- Agencia Costarricense de Investigaciones Biomédicas (ACIB) formerly Proyecto Epidemiológico Guanacaste, Fundación INCIENSA (FUNIN), San José, Costa Rica
| | - Lisa Mirabello
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - John T Schiller
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Mónica S Sierra
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Aimée R Kreimer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Gary M Clifford
- International Agency for Research on Cancer (IARC/WHO) Early Detection, Prevention and Infections Branch, Lyon, France
| | - Simon Beddows
- Virus Reference Department, Public Health Microbiology Division, UK Health Security Agency, London, UK.
- Blood Safety, Hepatitis, Sexually Transmitted Infections and HIV Division, UK Health Security Agency, London, UK.
| |
Collapse
|
20
|
Abstract
ABSTRACT The incidence of human papillomavirus (HPV)-associated head and neck cancer (HNC) has been rapidly increasing in developed countries, with HPV-associated HNC now accounting for 70% of all HNC cases. An increased incidence has been noted particularly among males. The disparities in HPV vaccine uptake rates and the increasing number of individuals with HPV-associated HNC suggest a lack of public awareness of both HPV sequelae and prevention options. This review highlights the importance of prophylactic HPV vaccination for preventing HPV-associated HNC, particularly in males. Current evidence substantiates the need for gender-neutral HPV vaccination programs and reinforces the recommendations made by the CDC. This article raises awareness of the association between HPV and HNC as well as the effectiveness of HPV vaccination in HNC prevention.
Collapse
|
21
|
Vielot NA, Brewer NT. Optimizing Cancer Prevention in Adolescents by Improving HPV Vaccine Delivery. N C Med J 2023; 85:33-36. [PMID: 39374359 DOI: 10.18043/001c.91426] [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: 10/09/2024]
Abstract
Routine HPV vaccination can prevent six cancers. Best practices for achieving high adolescent HPV vaccination coverage include automatically scheduled appointments, presumptive provider recommendations, standing orders, feedback on vaccination rates, and incentives. Promising practices include starting HPV vaccine recommendations and vaccine registry forecasting at age 9 and school entry requirements.
Collapse
Affiliation(s)
- Nadja A Vielot
- Department of Family Medicine, University of North Carolina at Chapel Hill
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill
| | - Noel T Brewer
- Department of Health Behavior, Gillings School of Global Public Health, University of North Carolina at Chapel Hill
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill
| |
Collapse
|
22
|
Zeng Y, Moscicki AB, Woo H, Hsu CH, Kemp TJ, Pinto LA, Szabo E, Dimond E, Bauman J, Sahasrabuddhe VV, Chow HHS. HPV16/18 Antibody Responses After a Single Dose of Nonavalent HPV Vaccine. Pediatrics 2023; 152:e2022060301. [PMID: 37317810 PMCID: PMC10312231 DOI: 10.1542/peds.2022-060301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/06/2023] [Indexed: 06/16/2023] Open
Abstract
OBJECTIVES A single dose of human papillomavirus (HPV) vaccine would simplify logistics and reduce costs of vaccination programs worldwide. We conducted a phase IIa trial to determine the stability of HPV type-specific antibody responses after a single dose of the nonavalent HPV vaccine, Gardasil9. METHODS Two hundred-and-one healthy 9 to 11-year-old girls and boys were enrolled at 2 centers in the United States to receive a prime dose of the nonavalent vaccine at baseline, a delayed dose at month 24, and an optional third dose at month 30. Blood samples were collected to measure HPV type-specific antibodies at baseline and at 6, 12, 18, 24, and 30 months after the prime dose. The primary outcomes were serum HPV16 and HPV18 antibody responses. RESULTS In both girls and boys, geometric mean concentrations of HPV16 and HPV18 antibodies increased at 6 months, declined between months 6 to 12, and then remained stable and high (at 20- and 10-times those at baseline for HPV16 and HPV18, respectively) throughout months 12, 18, and 24 (prebooster) visits. Both HPV16 and HPV18 antibody responses demonstrated anamnestic boosting effect at 30-months after the delayed (24-month) booster dose. CONCLUSIONS A single dose of the nonavalent HPV vaccine induced persistent and stable HPV16 and HPV18 antibody responses up to 24 months. This study contributes important immunogenicity data to inform feasibility of the single dose HPV vaccination paradigm. Further research is needed to assess the long-term antibody stability and individual clinical and public health benefit of the single dose schedule.
Collapse
Affiliation(s)
| | - Anna-Barbara Moscicki
- Department of Pediatrics, University of California Los Angeles, Los Angeles, California
| | - Heide Woo
- Department of Pediatrics, University of California Los Angeles, Los Angeles, California
| | - Chiu-Hsieh Hsu
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona
| | - Troy J. Kemp
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Ligia A. Pinto
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Eva Szabo
- Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| | - Eileen Dimond
- Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| | - Julie Bauman
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona
| | | | - H-H Sherry Chow
- Department of Pediatrics, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
23
|
Kemp TJ, Hempel HA, Pan Y, Roy D, Cherry J, Lowy DR, Pinto LA. Assay Harmonization Study To Measure Immune Response to SARS-CoV-2 Infection and Vaccines: a Serology Methods Study. Microbiol Spectr 2023; 11:e0535322. [PMID: 37191544 PMCID: PMC10269912 DOI: 10.1128/spectrum.05353-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/31/2023] [Indexed: 05/17/2023] Open
Abstract
The Coronavirus disease 2019 (COVID-19) pandemic presented the scientific community with an immediate need for accurate severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) serology assays, resulting in an expansion of assay development, some without following a rigorous quality control and validation, and with a wide range of performance characteristics. Vast amounts of data have been gathered on SARS-CoV-2 antibody response; however, performance and ability to compare the results have been challenging. This study seeks to analyze the reliability, sensitivity, specificity, and reproducibility of a set of widely used commercial, in-house, and neutralization serology assays, as well as provide evidence for the feasibility of using the World Health Organization (WHO) International Standard (IS) as a harmonization tool. This study also seeks to demonstrate that binding immunoassays may serve as a practical alternative for the serological study of large sample sets in lieu of expensive, complex, and less reproducible neutralization assays. In this study, commercial assays demonstrated the highest specificity, while in-house assays excelled in antibody sensitivity. As expected, neutralization assays demonstrated high levels of variability but overall good correlations with binding immunoassays, suggesting that binding may be reasonably accurate as well as practical for the study of SARS-CoV-2 serology. All three assay types performed well after WHO IS standardization. The results of this study demonstrate there are high performing serology assays available to the scientific community to rigorously dissect antibody responses to infection and vaccination. IMPORTANCE Previous studies have shown significant variability in SARS-CoV-2 antibody serology assays, highlighting the need for evaluation and comparison of these assays using the same set of samples covering a wide range of antibody responses induced by infection or vaccination. This study demonstrated that there are high performing assays that can be used reliably to evaluate immune responses to SARS-CoV-2 in the context of infection and vaccination. This study also demonstrated the feasibility of harmonizing these assays against the International Standard and provided evidence that the binding immunoassays may have high enough correlation with the neutralization assays to serve as a practical proxy. These results represent an important step in standardizing and harmonizing the many different serological assays used to evaluate COVID-19 immune responses in the population.
Collapse
Affiliation(s)
- Troy J. Kemp
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Heidi A. Hempel
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Yuanji Pan
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Daisy Roy
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - James Cherry
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Douglas R. Lowy
- Laboratory of Cellular Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ligia A. Pinto
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| |
Collapse
|
24
|
Losada C, Samaha H, Scherer EM, Kazzi B, Khalil L, Ofotokun I, Rouphael N. Efficacy and Durability of Immune Response after Receipt of HPV Vaccines in People Living with HIV. Vaccines (Basel) 2023; 11:1067. [PMID: 37376456 DOI: 10.3390/vaccines11061067] [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: 04/10/2023] [Revised: 05/24/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
People living with HIV (PLH) experience higher rates of HPV infection as well as an increased risk of HPV-related disease, including malignancies. Although they are considered a high-priority group for HPV vaccination, there are limited data regarding the long-term immunogenicity and efficacy of HPV vaccines in this population. Seroconversion rates and geometric mean titers elicited by vaccination are lower in PLH compared to immunocompetent participants, especially in individuals with CD4 counts below 200 cells/mm3 and a detectable viral load. The significance of these differences is still unclear, as a correlate of protection has not been identified. Few studies have focused on demonstrating vaccine efficacy in PLH, with variable results depending on the age at vaccination and baseline seropositivity. Although waning humoral immunity for HPV seems to be more rapid in this population, there is evidence that suggests that seropositivity lasts at least 2-4 years following vaccination. Further research is needed to determine the differences between vaccine formulations and the impact of administrating additional doses on durability of immune protection.
Collapse
Affiliation(s)
- Cecilia Losada
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, Decatur, GA 30030, USA
| | - Hady Samaha
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, Decatur, GA 30030, USA
| | - Erin M Scherer
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, Decatur, GA 30030, USA
| | - Bahaa Kazzi
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, Decatur, GA 30030, USA
| | - Lana Khalil
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, Decatur, GA 30030, USA
| | - Ighovwerha Ofotokun
- Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Nadine Rouphael
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, Decatur, GA 30030, USA
| |
Collapse
|
25
|
Qiu D, Liu Y, Wang Z, Zhang Z, Nie M, Xia N, Li S, Zhao Q. Precision and correlation of ED 50 and endpoint titer method in measuring HPV vaccine immunogenicity. J Virol Methods 2023; 316:114716. [PMID: 36965633 DOI: 10.1016/j.jviromet.2023.114716] [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: 01/09/2023] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 03/27/2023]
Abstract
Cervical cancer, the second leading cause of cancer-related deaths among women, is caused by human papillomavirus (HPV), a sexually transmitted virus. Vaccination is an effective preventive measure against viral infections and subsequent development of cervical cancer. Enzyme-linked immunosorbent assay (ELISA) is commonly used to measure specific binding antibody titers and assess the immunogenicity of test vaccines in preclinical models or clinical volunteers. Two methods of deriving titers, the endpoint titer (ET) and the effective dilution producing a median maximal effective fold of dilution (ED50) with a cut-off value, are widely used. For HPV, a pseudovirion-based neutralization assay (PBNA) is used to measure functional antibody titers. The ELISA binding titers and functional PBNA titers were found to be well-correlated for all nine HPV types tested in the vaccine, consistent with previous studies on HPV 16/18. Comparing the PBNA results with the two titration methods, the ED50 method showed higher precision and a closer correlation with PBNA results, both for individual types and pooled data analysis for all nine types. When comparing the titration results of the ET method based on a cut-off value with the ED50 method using all the data points across the dilution series, the ED50 method demonstrated better precision and a stronger correlation with PBNA results.
Collapse
Affiliation(s)
- Dekui Qiu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian 361102, PR China; School of Public Health, Xiamen University, Xiamen, Fujian 361102, PR China
| | - Yue Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian 361102, PR China; School of Public Health, Xiamen University, Xiamen, Fujian 361102, PR China
| | - Zhiping Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian 361102, PR China; School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, PR China
| | - Zhigang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian 361102, PR China; School of Public Health, Xiamen University, Xiamen, Fujian 361102, PR China
| | - Meifeng Nie
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian 361102, PR China; School of Public Health, Xiamen University, Xiamen, Fujian 361102, PR China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian 361102, PR China; School of Public Health, Xiamen University, Xiamen, Fujian 361102, PR China; Xiang An Biomedicine Laboratory, Xiamen, Fujian 361102, PR China; The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, PR China
| | - Shaowei Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian 361102, PR China; School of Public Health, Xiamen University, Xiamen, Fujian 361102, PR China; Xiang An Biomedicine Laboratory, Xiamen, Fujian 361102, PR China.
| | - Qinjian Zhao
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
26
|
Conarty JP, Wieland A. The Tumor-Specific Immune Landscape in HPV+ Head and Neck Cancer. Viruses 2023; 15:1296. [PMID: 37376596 DOI: 10.3390/v15061296] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 05/26/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Human papillomaviruses (HPVs) are the causative agent of several anogenital cancers as well as head and neck cancers, with HPV+ head and neck squamous cell carcinoma (HNSCC) becoming a rapidly growing public health issue in the Western world. Due its viral etiology and potentially its subanatomical location, HPV+ HNSCC exhibits an immune microenvironment which is more inflamed and thus distinct from HPV-negative HNSCC. Notably, the antigenic landscape in most HPV+ HNSCC tumors extends beyond the classical HPV oncoproteins E6/7 and is extensively targeted by both the humoral and cellular arms of the adaptive immune system. Here, we provide a comprehensive overview of HPV-specific immune responses in patients with HPV+ HNSCC. We highlight the localization, antigen specificity, and differentiation states of humoral and cellular immune responses, and discuss their similarities and differences. Finally, we review currently pursued immunotherapeutic treatment modalities that attempt to harness HPV-specific immune responses for improving clinical outcomes in patients with HPV+ HNSCC.
Collapse
Affiliation(s)
- Jacob P Conarty
- Department of Otolaryngology, The Ohio State University, Columbus, OH 43210, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Andreas Wieland
- Department of Otolaryngology, The Ohio State University, Columbus, OH 43210, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
27
|
Morais S, Wissing MD, Khosrow-Khavar F, Burchell AN, Tellier PP, Coutlée F, Waterboer T, El-Zein M, Franco EL. Serologic Response to Human Papillomavirus Genotypes Among Unvaccinated Women: Findings From the HITCH Cohort Study. J Infect Dis 2023; 227:1173-1184. [PMID: 36322543 PMCID: PMC10175069 DOI: 10.1093/infdis/jiac437] [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: 08/24/2022] [Revised: 10/25/2022] [Accepted: 10/31/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Humoral immune responses may be critical for preventing, controlling, and/or eliminating human papillomavirus (HPV) infection. We analyzed humoral response to natural HPV infection considering phylogenetic relatedness among unvaccinated women. METHODS We included 399 young women attending university/college in Montreal, Canada who were participants of the HITCH cohort. Participants provided blood samples at baseline and 5 follow-up visits. Antibody response to bacterially expressed L1 and E6 glutathione S-transferase (GST) fusion proteins, and virus-like particles (VLP-L1) of Alphapapillomavirus types were measured using multiplex serology. We assessed correlations and associations between HPV types at baseline using Pearson correlation coefficients (r) and univariable linear regressions. RESULTS At baseline, > 40% were seropositive for GST-L1 antibodies of at least 1 HPV type. Strong correlations between GST-L1 were observed for α9 HPV types: 58-52 (r = 0.86), 58-33 (r = 0.75), 33-52 (r = 0.72), and between GST-E6: 52-11 (r = 0.84), 52-18 (r = 0.79), 58-33 (r = 0.78), 35-11 (r = 0.76). HPV16 VLP-L1 moderately explained variability in HPV16 GST-L1 (regression coefficient [b] = 0.38, R2 = 43.1%), and HPV45 GST-L1 in HPV18 GST-L1 (b = 0.68, R2 = 42.8%). GST-E6 antibodies accounted for a low to moderate proportion of variability in HPV16 and HPV18 GST-E6 (R2 = 6.4%-62.2%). CONCLUSIONS Associations between naturally induced HPV-specific antibodies depend on phylogenetic relatedness.
Collapse
Affiliation(s)
- Samantha Morais
- Division of Cancer Epidemiology, McGill University, Montreal, Quebec, Canada
| | - Michel D Wissing
- Division of Cancer Epidemiology, McGill University, Montreal, Quebec, Canada
| | | | - Ann N Burchell
- Department of Family and Community Medicine and MAP Centre for Urban Health Solutions, Li Ka Shing Knowledge Institute, St Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | | | - François Coutlée
- Division of Cancer Epidemiology, McGill University, Montreal, Quebec, Canada
- Laboratoire de Virologie Moléculaire, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, et Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Tim Waterboer
- Infections and Cancer Epidemiology Division, German Cancer Research Center, Heidelberg, Germany
| | - Mariam El-Zein
- Division of Cancer Epidemiology, McGill University, Montreal, Quebec, Canada
| | - Eduardo L Franco
- Division of Cancer Epidemiology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
28
|
Roy V, Jung W, Linde C, Coates E, Ledgerwood J, Costner P, Yamshchikov G, Streeck H, Juelg B, Lauffenburger DA, Alter G. Differences in HPV-specific antibody Fc-effector functions following Gardasil® and Cervarix® vaccination. NPJ Vaccines 2023; 8:39. [PMID: 36922512 PMCID: PMC10017795 DOI: 10.1038/s41541-023-00628-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 02/17/2023] [Indexed: 03/17/2023] Open
Abstract
Gardasil® (Merck) and Cervarix® (GlaxoSmithKline) both provide protection against infection with Human Papillomavirus 16 (HPV16) and Human Papillomavirus 18 (HPV18), that account for around 70% of cervical cancers. Both vaccines have been shown to induce high levels of neutralizing antibodies and are known to protect against progression beyond cervical intraepithelial neoplasia grade 2 (CIN2+), although Cervarix® has been linked to enhanced protection from progression. However, beyond the transmission-blocking activity of neutralizing antibodies against HPV, no clear correlate of protection has been defined that may explain persistent control and clearance elicited by HPV vaccines. Beyond blocking, antibodies contribute to antiviral activity via the recruitment of the cytotoxic and opsonophagocytic power of the immune system. Thus, here, we used systems serology to comprehensively profile Gardasil®- and Cervarix®- induced antibody subclass, isotype, Fc-receptor binding, and Fc-effector functions against the HPV16 and HPV18 major capsid protein (L1). Overall, both vaccines induced robust functional humoral immune responses against both HPV16 and HPV18. However, Cervarix® elicited higher IgG3 and antibody-dependent complement activating responses, and an overall more coordinated response between HPV16 and 18 compared to Gardasil®, potentially related to the distinct adjuvants delivered with the vaccines. Thus, these data point to robust Fc-effector functions induced by both Gardasil® and Cervarix®, albeit with enhanced coordination observed with Cervarix®, potentially underlying immunological correlates of post-infection control of HPV.
Collapse
Affiliation(s)
- Vicky Roy
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.,Institute of Virology, University Hospital Bonn, Bonn, Germany
| | - Wonyeong Jung
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Caitlyn Linde
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Emily Coates
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Julie Ledgerwood
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Pamela Costner
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Galina Yamshchikov
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hendrik Streeck
- Institute of Virology, University Hospital Bonn, Bonn, Germany
| | - Boris Juelg
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
| |
Collapse
|
29
|
Kovačević G, Božić Nedeljković B, Patić A, Radovanov J, Hrnjaković-Cvjetković I. Human papillomavirus-specific antibody status among unvaccinated subjects in the region of Vojvodina, Serbia. Cent Eur J Public Health 2023; 31:57-62. [PMID: 37086422 DOI: 10.21101/cejph.a7257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/04/2023] [Indexed: 04/23/2023]
Abstract
OBJECTIVES The aim of the study was to evaluate the immune status of young people from the Vojvodina province, Serbia, through the detection of IgG antibodies specific for the L1 protein of HPV types 6, 11, 16, and 18 contained in quadrivalent vaccine. METHODS The study enrolled 514 healthy persons of both genders, aged between 18 and 30 years. All potential participants were informed about the project's aims by trained interviewers before venous blood collection. Also, participants completed a specially designed anonymous questionnaire to identify socio-demographic characteristics and individual behaviours associated with HPV seroprevalence. VPL HPV L1-specific IgG antibodies were measured using a semi-quantitative HPV IgG ELISA kit (Dia.Pro, Italy). RESULTS A total of 472 (91.8%) young subjects had no detectable antibodies against high- and low-risk HPV types covered by the quadrivalent vaccine. A slightly higher number of seropositive individuals were detected in the age group of 26-30 years compared to younger than 25. Multivariate analysis showed that the number of lifetime sexual partners was the most powerful predictor of HPV seropositivity (OR = 3.483, 95% CI: 1.294-9.379). CONCLUSIONS Obtained data point out low levels of naturally induced HPV-specific serum antibodies among the target population in the Vojvodina province. The present work highlights the significance and potential benefits of HPV vaccination. Routine HPV vaccination should be the public health priority in our country and should be included in the national immunization programme as soon as possible.
Collapse
Affiliation(s)
- Gordana Kovačević
- Centre of Virology, Institute for Public Health of Vojvodina, Novi Sad, Serbia
| | | | - Aleksandra Patić
- Centre of Virology, Institute for Public Health of Vojvodina, Novi Sad, Serbia
- Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Jelena Radovanov
- Centre of Virology, Institute for Public Health of Vojvodina, Novi Sad, Serbia
| | - Ivana Hrnjaković-Cvjetković
- Centre of Virology, Institute for Public Health of Vojvodina, Novi Sad, Serbia
- Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| |
Collapse
|
30
|
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
|
31
|
Donkoh ET, Dassah ET, Owusu-Dabo E. Optimization of a protocol for the evaluation of antibody responses to human papillomavirus (HPV) vaccination in low-resource settings. BMC Womens Health 2022; 22:234. [PMID: 35710373 PMCID: PMC9204889 DOI: 10.1186/s12905-022-01821-y] [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: 09/22/2021] [Accepted: 06/06/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Available human papillomavirus (HPV) vaccines could have an important primary role in cervical cancer prevention once their long-term immunogenicity and safety are evaluated at the population level. The aim of this study was to optimize an assay to be used in evaluating the long-term durability of HPV vaccine response following a pilot vaccination of adolescent girls in Ghana. Methods A rapid, high-throughput, indirect enzyme-linked immunosorbent assay (ELISA) was optimized for the detection and quantitation of anti-HPV L1 (late expression protein: types 6, 11, 16 and 18) immunoglobulin G (IgG) in human serum (n = 89). The utility of the assay was demonstrated using serum collected from a cohort of pre-adolescent girls (n = 49) previously vaccinated with a quadrivalent vaccine and non-immune serum obtained from age-matched controls (n = 40). Results The assay showed good discrimination of antibody levels between cases and control sera: seroprevalence of anti-HPV IgG antibodies was significantly higher among vaccinated than unvaccinated girls for both HPV-16 (63.3% vs. 12.5%; p < 0.001) and HPV-18 (34.7% vs. 20.0%; p = 0.042), respectively. Thirty-six months after receiving the third dose of vaccine, significantly higher mean anti-HPV-16 (0.618 vs. 0.145), anti-HPV-18 (0.323 vs. 0.309), and anti-HPV-6 (1.371 vs. 0.981) antibody levels were measured, compared to unvaccinated girls (all p < 0.05). A correlation between optical density and antibody activity indicated assay sensitivity to increasing levels of antibody activity. Conclusion We have successfully optimized and implemented a robust and sensitive assay for the evaluation of antibody responses among immunized adolescent girls for monitoring future large-scale HPV vaccination studies in low-income settings. Our results demonstrated greater immunoglobulin G antibody activity within serum drawn from adolescent girls immunized 36 months prior. Supplementary Information The online version contains supplementary material available at 10.1186/s12905-022-01821-y.
Collapse
Affiliation(s)
- Emmanuel Timmy Donkoh
- Center for Research in Applied Biology, University of Energy and Natural Resources, Sunyani, Ghana.
| | - Edward Tieru Dassah
- Department of Population, Family and Reproductive Health, School of Public Health, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Ellis Owusu-Dabo
- Department of Global and International Health, School of Public Health, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| |
Collapse
|
32
|
Smahelova J, Hamsikova E, Ludvikova V, Vydrova J, Traboulsi J, Vencalek O, Lukeš P, Tachezy R. Outcomes After Human Papillomavirus Vaccination in Patients With Recurrent Respiratory Papillomatosis: A Nonrandomized Clinical Trial. JAMA Otolaryngol Head Neck Surg 2022; 148:654-661. [PMID: 35653138 DOI: 10.1001/jamaoto.2022.1190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance Recurrent respiratory papillomatosis (RRP) is a rare benign chronic disease of the larynx etiologically linked with the infection of low-risk human papillomavirus (HPV). Combination of surgical and immunomodulatory therapy has limited success. Possible use of prophylactic HPV vaccine that includes HPV-6 and HPV-11 antigens has been studied. Objective To evaluate if the HPV vaccination is associated with a lower number of recurrences requiring surgical intervention in patients with new and recurrent RRP. Design, Setting, and Participants This was a non-placebo-controlled intervention study. Enrollment data were collected from October 2011 to August 2013. The patients were followed up at 1 month, 12 months, and 5 years after the third dose of the vaccine and clinically monitored until December 31, 2018. Data were analyzed from 2019 to 2021. Altogether, 50 adults with active RRP were enrolled and followed up in referral centers. For the final outcome, follow-up data for 42 patients were available. Eight patients who did not fulfill the protocol were excluded. Interventions All patients received HPV vaccine as an adjuvant treatment and were clinically followed up. When RRP progression or a significant recurrent lesion was detected, surgical removal via direct laryngoscopy was indicated. No adjuvant therapy with antiviral or biological agents was used. Main Outcomes and Measures This study compared the prevaccination and postvaccination positivity for HPV-specific antibodies. The main outcome was the difference in the frequency of RRP recurrences in the prevaccination and postvaccination period. Results A total of 50 patients with RRP were enrolled (median [SD] age, 41.5 [12.3] years [range, 21-73 years]; 39 [78%] men and 11 [22%] women). After HPV vaccination, patients with previously no HPV-specific antibodies showed seroconversion, and all patients developed 100-fold higher levels of HPV vaccine type-specific antibodies compared with the prevaccination period. In patients with recurrent RRP, decreased frequency of recurrences requiring surgical treatment was present after vaccination (from 0.85 to 0.36 recurrences/y). No difference in postvaccination recurrences was found between patients with newly diagnosed and recurrent RRP. Conclusions and Relevance In this nonrandomized clinical trial, the frequency of RRP recurrences was significantly lower after HPV vaccination, and patients with RRP thus had a reduced burden of disease. Because no difference was detected in the frequency of recurrent postvaccination lesions in patients with new and recurrent disease, it appears that both groups showed equal benefit following HPV vaccination. These findings suggest that the earlier that patients with RRP receive HPV vaccine, the sooner they may show reduced burden of disease. Trial Registration EudraCT Identifier: 2011-002667-14; ClinicalTrials.gov Identifier: NCT01375868.
Collapse
Affiliation(s)
- Jana Smahelova
- Department of Genetics and Microbiology, Faculty of Science, BIOCEV, Charles University, Prague, Czech Republic.,Institute of Hematology and Blood Transfusion, National Reference Laboratory for Papillomaviruses and Polyomaviruses, Prague, Czech Republic
| | - Eva Hamsikova
- Institute of Hematology and Blood Transfusion, National Reference Laboratory for Papillomaviruses and Polyomaviruses, Prague, Czech Republic
| | - Viera Ludvikova
- Institute of Hematology and Blood Transfusion, National Reference Laboratory for Papillomaviruses and Polyomaviruses, Prague, Czech Republic
| | - Jitka Vydrova
- Prague Voice Centre/Medical Healthcom, Prague, Czech Republic
| | - Joseph Traboulsi
- Department of ENT/Head and Neck Surgery, Na Homolce Hospital, Prague, Czech Republic
| | - Ondrej Vencalek
- Department of Mathematical Analysis and Applications of Mathematics, Faculty of Science, Palacky University, Olomouc, Czech Republic
| | - Petr Lukeš
- Department of Otorhinolaryngology and Head and Neck Surgery, First Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic
| | - Ruth Tachezy
- Department of Genetics and Microbiology, Faculty of Science, BIOCEV, Charles University, Prague, Czech Republic.,Institute of Hematology and Blood Transfusion, National Reference Laboratory for Papillomaviruses and Polyomaviruses, Prague, Czech Republic
| |
Collapse
|
33
|
Ejezie CL, Osaghae I, Ayieko S, Cuccaro P. Adherence to the Recommended HPV Vaccine Dosing Schedule among Adolescents Aged 13 to 17 Years: Findings from the National Immunization Survey-Teen, 2019–2020. Vaccines (Basel) 2022; 10:vaccines10040577. [PMID: 35455325 PMCID: PMC9026751 DOI: 10.3390/vaccines10040577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/06/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022] Open
Abstract
The 9-valent human papillomavirus (9-vHPV) vaccine uptake rate among adolescents has improved over the years; however, little is known about the adherence to the recommended dosing schedule. This study examines the prevalence and factors associated with adherence to the recommended 9vHPV vaccination dosing schedule among adolescents aged 13 to 17 years. The cross-sectional study was conducted using the 2019–2020 National Immunization Survey-Teen. The parents of 34,619 adolescents were included in our analyses. The overall up-to-date (UTD) prevalence was 57.1%. The UTD prevalence was 60.0% among females and 54.2% among males. Adolescents aged 16 years had the highest UTD prevalence of 63.0%. The UTD prevalence was 61.6% among Hispanics and 54.7% among non-Hispanic Whites. Overall, compared to females, males had 14% lower odds of UTD. The odds of UTD were 1.91 times, 2.08 times, and 1.98 times higher among adolescents aged 15–17 years, respectively, compared to those aged 13 years. Moreover, region, poverty, insurance status, mothers’ educational level, and provider recommendation were associated with UTD. Our findings show that adherence to the recommended 9vHPV vaccine schedule is low in the US. Targeted public health efforts are needed to improve the rates of adherence to the recommended 9vHPV dose schedule.
Collapse
Affiliation(s)
- Chinenye Lynette Ejezie
- Department of Health Promotion and Behavioral Sciences, The University of Texas School of Public Health, 1200 Pressler St, Houston, TX 77030, USA; (S.A.); (P.C.)
- Department of Investigational Cancer Therapeutics, The University of MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: ; Tel.: +1-(832-513-3925)
| | - Ikponmwosa Osaghae
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- Department of Epidemiology, Human Genetics & Environmental Sciences, The University of Texas School of Public Health, Houston, TX 77030, USA
| | - Sylvia Ayieko
- Department of Health Promotion and Behavioral Sciences, The University of Texas School of Public Health, 1200 Pressler St, Houston, TX 77030, USA; (S.A.); (P.C.)
| | - Paula Cuccaro
- Department of Health Promotion and Behavioral Sciences, The University of Texas School of Public Health, 1200 Pressler St, Houston, TX 77030, USA; (S.A.); (P.C.)
| |
Collapse
|
34
|
Thurm C, Reinhold A, Borucki K, Kahlfuss S, Feist E, Schreiber J, Reinhold D, Schraven B. Homologous and Heterologous Anti-COVID-19 Vaccination Does Not Induce New-Onset Formation of Autoantibodies Typically Accompanying Lupus Erythematodes, Rheumatoid Arthritis, Celiac Disease and Antiphospholipid Syndrome. Vaccines (Basel) 2022; 10:vaccines10020333. [PMID: 35214790 PMCID: PMC8880348 DOI: 10.3390/vaccines10020333] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 02/04/2023] Open
Abstract
The COVID-19 pandemics has caused the death of almost six million people worldwide. In order to establish collective immunity, the first vaccines that were approved in Germany were the vector virus-based vaccine Vaxzevria and the mRNA vaccines Comirnaty and Spikevax, respectively. As it was reported that SARS-CoV-2 can trigger autoimmunity, it is of significant interest to investigate whether COVID-19 vaccines evoke the formation of autoantibodies and subsequent autoimmunity. Here, we analyzed immune responses after different vaccination regimens (mRNA/mRNA, Vector/Vector or Vector/mRNA) with respect to anti-SARS-CoV-2-specific immunity and the development of autoantibodies well known for their appearance in distinct autoimmune diseases. We found that anti-SARS-CoV-2 antibody levels were 90% lower after Vector/Vector vaccination compared to the other vaccinations and that Vector/mRNA vaccination was more effective than mRNA/mRNA vaccination in terms of IgM and IgA responses. However, until 4 months after booster vaccination we only detected increases in autoantibodies in participants with already pre-existing autoantibodies whereas vaccinees showing no autoantibody formation before vaccination did not respond with sustained autoantibody production. Taken together, our study suggests that all used COVID-19 vaccines do not significantly foster the appearance of autoantibodies commonly associated with lupus erythematodes, rheumatoid arthritis, Celiac disease and antiphospholipid-syndrome but provide immunity to SARS-CoV-2.
Collapse
Affiliation(s)
- Christoph Thurm
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany; (C.T.); (A.R.); (S.K.); (D.R.)
- ChaMP, Center for Health and Medical Prevention, Otto-von-Guericke-University, 39106 Magdeburg, Germany
| | - Annegret Reinhold
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany; (C.T.); (A.R.); (S.K.); (D.R.)
- ChaMP, Center for Health and Medical Prevention, Otto-von-Guericke-University, 39106 Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GC-I3), Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany; (E.F.); (J.S.)
| | - Katrin Borucki
- Institute of Clinical Chemistry and Pathobiochemistry, Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany;
| | - Sascha Kahlfuss
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany; (C.T.); (A.R.); (S.K.); (D.R.)
- ChaMP, Center for Health and Medical Prevention, Otto-von-Guericke-University, 39106 Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GC-I3), Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany; (E.F.); (J.S.)
- Institute of Medical Microbiology and Hospital Hygiene, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Eugen Feist
- Health Campus Immunology, Infectiology and Inflammation (GC-I3), Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany; (E.F.); (J.S.)
- Department of Rheumatology, Helios Specialist Hospital Vogelsang, 39245 Gommern, Germany
| | - Jens Schreiber
- Health Campus Immunology, Infectiology and Inflammation (GC-I3), Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany; (E.F.); (J.S.)
- Department of Pneumology, University Hospital, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | - Dirk Reinhold
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany; (C.T.); (A.R.); (S.K.); (D.R.)
- ChaMP, Center for Health and Medical Prevention, Otto-von-Guericke-University, 39106 Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GC-I3), Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany; (E.F.); (J.S.)
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany; (C.T.); (A.R.); (S.K.); (D.R.)
- ChaMP, Center for Health and Medical Prevention, Otto-von-Guericke-University, 39106 Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GC-I3), Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany; (E.F.); (J.S.)
- Correspondence: ; Tel.: +49-391-67-15800
| |
Collapse
|
35
|
Machalek D, Rees H, Chikandiwa A, Munthali R, Travill D, Mbulawa Z, Petoumenos K, Delany-Moretlwe S, Kaldor J. Impact of one and two human papillomavirus (HPV) vaccine doses on community-level HPV prevalence in South African adolescent girls: study protocol and rationale for a pragmatic before-after design. BMJ Open 2022; 12:e059968. [PMID: 35144959 PMCID: PMC8845310 DOI: 10.1136/bmjopen-2021-059968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/20/2022] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION Vaccines against human papillomavirus (HPV) are the key to controlling cervical cancer in low/middle-income countries (LMICs) where incidence is highest, but there have been limited data from these settings on programme impact on HPV prevalence, and none in a population with endemic HIV infection. Furthermore, for many LMICs, the currently recommended two-dose schedule is difficult to deliver at scale, so there is mounting interest in a single-dose schedule. METHODS AND ANALYSIS The Human Papillomavirus One and Two-Dose Population Effectiveness Study is a hybrid impact evaluation of the national South African HPV vaccination programme, which has targeted grade 4 girls aged at least 9 years in public schools with two doses of vaccine since 2014, and a single-dose vaccine 'catch-up' programme delivered in one district in 2019. Impacts of both schedules on the prevalence of type-specific HPV infection will be measured using repeat cross-sectional surveys in adolescent girls and young women aged 17-18 years recruited at primary healthcare clinics in the four provinces. A baseline survey in 2019 measured HPV prevalence in the cohort who were ineligible for vaccination because they were already above the target age or grade under either the national programme or the single-dose programme in the selected district. HPV prevalence surveys are repeated in 2021 in the selected district, and in 2023 in all four provinces. We will calculate prevalence ratios to compare the prevalence of HPV types 16 and 18 in the single-dose (2021) and two-dose (2023) cohorts, with the vaccine-ineligible (2019) cohort. ETHICS AND DISSEMINATION The project was approved by the University of the Witwatersrand Human Research Ethics Committee (HREC #181005), and the University of New South Wales HREC (#181-005). Findings will be disseminated through peer-reviewed journals, scientific meetings, reports and community forums.
Collapse
Affiliation(s)
- Dorothy Machalek
- Kirby Institute, University of New South Wales-Kensington Campus, Sydney, New South Wales, Australia
- Centre for Women's Infectious Diseases, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Helen Rees
- Wits RHI, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Admire Chikandiwa
- Wits RHI, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Richard Munthali
- Wits RHI, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Danielle Travill
- Wits RHI, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Zizipho Mbulawa
- UCT-MRC Clinical Gynaecological Cancer Research Centre, University of Cape Town, Rondebosch, Western Cape, South Africa
- Department of Laboratory Medicine and Pathology, Walter Sisulu University, Mthatha, Eastern Cape, South Africa
| | - Kathy Petoumenos
- Kirby Institute, University of New South Wales-Kensington Campus, Sydney, New South Wales, Australia
| | | | - John Kaldor
- Kirby Institute, University of New South Wales-Kensington Campus, Sydney, New South Wales, Australia
| |
Collapse
|
36
|
Syrjänen S, Waterboer T, Rintala M, Pawlita M, Syrjänen K, Louvanto K, Grenman S. Maternal HPV-antibodies and seroconversion to HPV in children during the first 3 years of life. Sci Rep 2022; 12:2227. [PMID: 35140326 PMCID: PMC8828864 DOI: 10.1038/s41598-022-06343-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/21/2022] [Indexed: 12/14/2022] Open
Abstract
To assess the dynamics of human papillomavirus (HPV) serology, we analyzed HPV6-,11-,16-,18-, and 45 antibodies in infants during the first 36 months of their life. Serial serum samples of 276/327 mother–child pairs were collected at baseline (mothers) and at months 1, 2, 6, 12, 24 and 36 (offspring), and tested for HPVL1-antibodies using the GST-L1 assay. Concordance between maternal and infant HPV-antibody levels remained high until month-6 (p < = 0.001), indicating maternal antibody transfer. At 1 month, 40–62% of the infants tested seropositive to any of the 5 HPV-types. Between 1–3 years of age, 53% (58/109) of the children born to HPV-seronegative mothers tested HPV-seropositive. Times to positive seroconversion varied between13.4 and 18.7 months, and times to negative seroconversion (decay) between 8.5 and 9.9 months. Significant independent predictors of infants’ seroconversion to LR-HPV were hand warts and mother’s history of oral warts and seroconversion to LR-HPV. No predictors of seroconversion to HR-HPV were identified. Maternal HPV-IgG-antibodies are transferred to her offspring and remain detectable for 6 months, corroborating the IgG molecule’s half-life. Seroconversion to HPV-genotypes 6, 11, 16 and 18 was confirmed among children born to HPV-seronegative mothers, implicating an immune response to these HPV-genotypes during early infancy.
Collapse
Affiliation(s)
- Stina Syrjänen
- Department of Oral Pathology, Institute of Dentistry, Faculty of Medicine, University of Turku, Lemminkäisenkatu 2, 20520, Turku, Finland. .,Department of Pathology, University of Turku, Turku University Hospital, Turku, Finland.
| | - Tim Waterboer
- Department of Genome Modifications and Carcinogenesis, Infection and Cancer Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marjut Rintala
- Department of Obstetrics and Gynecology, University of Turku, Turku University Hospital, Turku, Finland
| | - Michael Pawlita
- Department of Genome Modifications and Carcinogenesis, Infection and Cancer Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kari Syrjänen
- Department of Clinical Research, Biohit Oyj, Helsinki, Finland
| | - Karolina Louvanto
- Department of Oral Pathology, Institute of Dentistry, Faculty of Medicine, University of Turku, Lemminkäisenkatu 2, 20520, Turku, Finland.,Department of Obstetrics and Gynecology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Seija Grenman
- Department of Obstetrics and Gynecology, University of Turku, Turku University Hospital, Turku, Finland
| |
Collapse
|
37
|
Panwar K, Godi A, Cocuzza CE, Andrews N, Southern J, Turner P, Miller E, Beddows S. Multiplex Human Papillomavirus L1L2 virus-like particle antibody binding assay. MethodsX 2022; 9:101776. [PMID: 35813158 PMCID: PMC9260319 DOI: 10.1016/j.mex.2022.101776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/22/2022] [Indexed: 12/04/2022] Open
Abstract
A variety of in vitro techniques are available to estimate the level of antibodies present in human serum samples. Such tests are highly specific and are used to determine prior exposure to a pathogen or to estimate the magnitude, breadth and durability of individual and population level vaccine immunity. Multiplex (or multi-analyte) platforms are increasingly being used to evaluate immune responses against multiple antigens at the same time, usually at reduced per sample cost and a more efficient use of available samples. Consequently, multiplex serology is an essential component of a wide range of public health programmes. Human papillomavirus (HPV) serology is limited to a small number of academic, public health and vaccine manufacturer laboratories globally. Such platforms include indirect binding to the major (L1) capsid protein virus-like particles (VLP), monoclonal antibody competition against L1 VLP and indirect binding to L1 and L2 (minor capsid protein) VLP on multiplex (Luminex®, Meso Scale Discovery®) and standard (ELISA) platforms. The methodology described here utilizes a common multi-analyte platform and L1L2-based VLP expressed in house, which allows the simultaneous detection and quantification of antibody responses against nine vaccine-relevant HPV genotypes.
Collapse
Affiliation(s)
- Kavita Panwar
- Virus Reference Department, UK Health Security Agency, London, UK
| | - Anna Godi
- Virus Reference Department, UK Health Security Agency, London, UK
| | | | - Nick Andrews
- Statistics, Modelling and Economics Department, UK Health Security Agency, London, UK
| | - Jo Southern
- Immunisation and Vaccine-Preventable Diseases Division, UK Health Security Agency, London, UK
| | - Paul Turner
- Section of Paediatrics, Imperial College London, London, UK
| | - Elizabeth Miller
- Immunisation and Vaccine-Preventable Diseases Division, UK Health Security Agency, London, UK
| | - Simon Beddows
- Virus Reference Department, UK Health Security Agency, London, UK
- Blood Safety, Hepatitis, Sexually Transmitted Infections and HIV Division, UK Health Security Agency, London, UK
- Corresponding author at: Virus Reference Department, UK Health Security Agency, 61 Colindale Avenue, London NW9 5EQ, UK.
| |
Collapse
|
38
|
Papadopoli R, De Sarro C, Palleria C, Gallelli L, Pileggi C, De Sarro G. Serological Response to SARS-CoV-2 Messenger RNA Vaccine: Real-World Evidence from Italian Adult Population. Vaccines (Basel) 2021; 9:1494. [PMID: 34960240 PMCID: PMC8705669 DOI: 10.3390/vaccines9121494] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND This study aims to investigate the extent of the BNT162b2 mRNA vaccine-induced antibodies against SARS-CoV-2 in a large cohort of Italian subjects belonging to the early vaccinated cohort in Italy. METHODS A prospective study was conducted between December 2020 and May 2021. Three blood samples were collected for each participant: one at the time of the first vaccine dose (T0), one at the time of the second vaccine dose, (T1) and the third 30 days after this last dose (T2). RESULTS We enrolled 2591 fully vaccinated subjects; 16.5% were frail subjects, and 9.8% were over 80 years old. Overall, 98.1% of subjects were seropositive when tested at T2, and 76.3% developed an anti-S IgG titer ≥4160 AU/mL, which is adequate to develop viral neutralizing antibodies. Seronegative subjects at T1 were more likely to remain seronegative at T2 or to develop a low-intermediate anti-S IgG titer (51-4159 AU/mL). CONCLUSIONS In summary, vaccination leads to detectable anti-S IgG titer in nearly all vaccine recipients. Stratification of the seroconversion level could be useful to promptly identify high-risk groups who may not develop a viral neutralizing response, even in the presence of seroconversion, and therefore may remain at higher risk of infection, despite vaccination.
Collapse
Affiliation(s)
| | | | | | | | - Claudia Pileggi
- Department of Health Sciences, Campus “Salvatore Venuta”, University of Catanzaro “Magna Græcia”, 88100 Catanzaro, Italy; (R.P.); (C.D.S.); (C.P.); (L.G.); (G.D.S.)
| | | |
Collapse
|
39
|
Kamolratanakul S, Pitisuttithum P. Human Papillomavirus Vaccine Efficacy and Effectiveness against Cancer. Vaccines (Basel) 2021; 9:vaccines9121413. [PMID: 34960159 PMCID: PMC8706722 DOI: 10.3390/vaccines9121413] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 12/11/2022] Open
Abstract
Human papillomavirus (HPV) is the most common sexually transmitted infection, with 15 HPV types related to cervical, anal, oropharyngeal, penile, vulvar, and vaginal cancers. However, cervical cancer remains one of the most common cancers in women, especially in developing countries. Three HPV vaccines have been licensed: bivalent (Cervarix, GSK, Rixensart, Belgium), quadrivalent (Merck, Sharp & Dome (Merck & Co, Whitehouse Station, NJ, USA)), and nonavalent (Merck, Sharp & Dome (Merck & Co, Whitehouse Station, NJ, USA)). The current HPV vaccine recommendations apply to 9 years old and above through the age of 26 years and adults aged 27–45 years who might be at risk of new HPV infection and benefit from vaccination. The primary target population for HPV vaccination recommended by the WHO is girls aged 9–14 years, prior to their becoming sexually active, to undergo a two-dose schedule and girls ≥ 15 years of age, to undergo a three-dose schedule. Safety data for HPV vaccines have indicated that they are safe. The most common adverse side-effect was local symptoms. HPV vaccines are highly immunogenic. The efficacy and effectiveness of vaccines has been remarkably high among young women who were HPV seronegative before vaccination. Vaccine efficacy was lower among women regardless of HPV DNA when vaccinated and among adult women. Comparisons of the efficacy of bivalent, quadrivalent, and nonavalent vaccines against HPV 16/18 showed that they are similar. However, the nonavalent vaccine can provide additional protection against HPV 31/33/45/52/58. In a real-world setting, the notable decrease of HPV 6/11/16/18 among vaccinated women compared with unvaccinated women shows the vaccine to be highly effective. Moreover, the direct effect of the nonavalent vaccine with the cross-protection of bivalent and quadrivalent vaccines results in the reduction of HPV 6/11/16/18/31/33/45/52/58. HPV vaccination has been shown to provide herd protection as well. Two-dose HPV vaccine schedules showed no difference in seroconversion from three-dose schedules. However, the use of a single-dose HPV vaccination schedule remains controversial. For males, the quadrivalent HPV vaccine possibly reduces the incidence of external genital lesions and persistent infection with HPV 6/11/16/18. Evidence regarding the efficacy and risk of HPV vaccination and HIV infection remains limited. HPV vaccination has been shown to be highly effective against oral HPV type 16/18 infection, with a significant percentage of participants developing IgG antibodies in the oral fluid post vaccination. However, the vaccines’ effectiveness in reducing the incidence of and mortality rates from HPV-related head and neck cancers should be observed in the long term. In anal infections and anal intraepithelial neoplasia, the vaccines demonstrate high efficacy. While HPV vaccines are very effective, screening for related cancers, as per guidelines, is still recommended.
Collapse
|
40
|
dos Santos Ferreira CE, Gómez-Dantés H, Junqueira Bellei NC, López E, Nogales Crespo KA, O’Ryan M, Villegas J. The Role of Serology Testing in the Context of Immunization Policies for COVID-19 in Latin American Countries. Viruses 2021; 13:2391. [PMID: 34960660 PMCID: PMC8706237 DOI: 10.3390/v13122391] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/18/2021] [Accepted: 11/18/2021] [Indexed: 12/16/2022] Open
Abstract
This review aims to explore the role and value of serology testing in the context of COVID-19 immunization policies in Latin American countries and the barriers and challenges to the adequate use and uptake of this tool. It builds on a review of the academic literature, evidence, and existing policies, and includes a multistage process of discussion and feedback by a group of five experts. Regional and country-level evidence and resources from five focus countries-Argentina, Brazil, Chile, Colombia, and Mexico-were collected and analyzed. This review contains an overview of (1) the impact of the SARS-CoV-2 pandemic, the variants of concern and current testing strategies, (2) the introduction of COVID-19 vaccination, (3) the potential use of serology testing to support immunization initiatives, (4) the current frameworks for the use of serology testing in the region, and (5) the barriers and challenges to implementing serology testing in the context of COVID-19 immunization policies, including a discussion on the potential actions required to address these barriers and facilitate the uptake of this strategy in the region. Stakeholders can use elements of this document to guide timely decision-making, raise awareness, and inspire further studies.
Collapse
Affiliation(s)
- Carlos E. dos Santos Ferreira
- Clinical Pathology, Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil;
- Microbiology Sector, Federal University of São Paulo’s Central Laboratory Activities, São Paulo 04088-002, Brazil
- Brazilian Society of Clinical Pathology and Laboratory Medicine, Rio de Janeiro 22220-040, Brazil
| | | | | | - Eduardo López
- Department of Medicine, Hospital de Niños Gutiérrez, Buenos Aires C1425-EFD, Argentina;
- Pediatric Infectious Diseases Program, Faculty of Medicine, University of Buenos Aires, Buenos Aires C1121-ABG, Argentina
- Pediatrics and Vaccinology, Faculty of Medicine, University of Salvador, Buenos Aires C1055-AAG, Argentina
| | | | - Miguel O’Ryan
- Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago de Chile 8380000, Chile;
- Millennium Institute of Immunology and Immunotherapy, University of Chile, Santiago de Chile 8331150, Chile
- Chilean Academy of Medicine, Santiago de Chile 6500445, Chile
| | | |
Collapse
|
41
|
Panicker G, Rajbhandari I, Pathak HN, Brady AM, Unger ER. Multiplex immunoassay to measure antibody response to nine HPV vaccine types. J Immunol Methods 2021; 498:113136. [PMID: 34464605 PMCID: PMC11059205 DOI: 10.1016/j.jim.2021.113136] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/22/2021] [Accepted: 08/25/2021] [Indexed: 11/29/2022]
Abstract
Well-characterized HPV serology assays are required to evaluate performance of biosimilar candidate vaccines, reduced dosing schedules and novel administration methods. We report characterization of an expanded assay, M9ELISA, that detects antibodies to HPV virus-like particles (VLP) of nine types using direct IgG ELISA on the Meso Scale Discovery (MSD) electrochemiluminescence platform. The method is based on the previously published M4ELISA which detects antibodies to HPV6,11,16, and 18. It has been modified to add detection of antibodies to HPV31,33,45,52 and 58, and to streamline assay and reduce background. The M9ELISA plates were prepared with purified type specific L1 + L2 VLPs coated on 10-spot/well standard MSD microplates. Results of ELISA on three serial dilutions of serum were read on MSD imager, and titers calculated using the parallel line method. Evaluations included dynamic range, assay reproducibility, and stability over time. We compared M9ELISA results to those from a pseudovirion-based neutralization assay in sera from a mixed cohort of unvaccinated and vaccinated individuals (n = ~116) and to competitive Luminex immunoassay (cLIA) results in sera from a predominantly unvaccinated cohort (n = 4426). The linear range of the assay extended over 5 logs, with inter-assay reproducibility coefficient of variation ≤25% for all types. The pre-coated plates were stable for at least 2 years. Spearman correlation of antibody titers showed excellent correlation with PBNA (r = 0.86-0.97) and moderate correlation (r = 0.52-0.68) with cLIA. Thus, the M9ELISA can serve as a useful platform for high-throughput, sensitive and simultaneous quantitation of the antibody responses to nine HPV vaccine types.
Collapse
Affiliation(s)
- G Panicker
- Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, USA.
| | - I Rajbhandari
- Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, USA
| | - H N Pathak
- Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, USA
| | - A M Brady
- Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, USA
| | - E R Unger
- Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, USA
| |
Collapse
|
42
|
Hoes J, Pasmans H, Schurink-van 't Klooster TM, van der Klis FRM, Donken R, Berkhof J, de Melker HE. Review of long-term immunogenicity following HPV vaccination: Gaps in current knowledge. Hum Vaccin Immunother 2021; 18:1908059. [PMID: 34033518 PMCID: PMC8920133 DOI: 10.1080/21645515.2021.1908059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The licensed HPV vaccines are highly efficacious and induce high levels of neutralizing antibody levels, the assumed mediators of protection. However, a correlate of protection against HPV is lacking, and the evidence is still limited as to long-term persistence of antibodies, especially following reduced dosing schedules. The World Health Organization (WHO) urges immunization of young girls as part of the strategy to eliminate cervical cancer, thus long-lasting protection is required. The current review describes long-term follow-up regarding vaccine-induced seropositivity and antibody level development following the different vaccines and dosing schedules. Implications and opportunities of long-term vaccine-induced immune responses are discussed, such as the gaps in monitoring of long-term immunogenicity, the possibilities of reduced dosing schedules, and the importance of evidence for durable immunity.
Collapse
Affiliation(s)
- J Hoes
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands.,Department of Epidemiology and Data Science, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - H Pasmans
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - T M Schurink-van 't Klooster
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - F R M van der Klis
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - R Donken
- Department of Epidemiology and Data Science, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - J Berkhof
- Department of Epidemiology and Data Science, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - H E de Melker
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| |
Collapse
|
43
|
Donken R, Dobson SRM, Marty KD, Cook D, Sauvageau C, Gilca V, Dionne M, McNeil S, Krajden M, Money D, Kellner J, Scheifele DW, Kollmann T, Bettinger JA, Liu S, Singer J, Naus M, Sadarangani M, Ogilvie GS. Immunogenicity of 2 and 3 Doses of the Quadrivalent Human Papillomavirus Vaccine up to 120 Months Postvaccination: Follow-up of a Randomized Clinical Trial. Clin Infect Dis 2021; 71:1022-1029. [PMID: 31617568 PMCID: PMC7428395 DOI: 10.1093/cid/ciz887] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 09/04/2019] [Indexed: 02/06/2023] Open
Abstract
Background Several countries have implemented a 2-dose (2D) human papillomavirus (HPV) vaccination schedule for adolescents based on immunobridging studies. We compared immunogenicity of 2D vs 3-dose (3D) schedules of the quadrivalent vaccine (4vHPV) up to 10 years after the first dose. Methods Girls aged 9–13 years were randomized to receive 2D or 3D and were compared with women aged 16–26 receiving 3D at day 1 and months 7, 24, and 120 after the first dose. Antibody levels for HPV6/11/16/18 were evaluated using the competitive Luminex immunoassay (cLIA) and total immunoglobulin G assay. Geometric mean titers (GMTs) and seropositivity rates were compared between the different groups at different time points. Noninferiority of GMT ratios was defined as the lower bound of the 2-sided 95% confidence interval (CI) being greater than 0.5. Kinetics of antibody titers over time among study groups were examined. Results At 120 months, data from 35 2D girls, 38 3D girls, and 30 3D women were used for analyses. cLIA seropositivity rates were above 95% for all HPV vaccine types and all schedules, except HPV18, with the lowest seropositivity observed among 3D women (60.0%; 95% CI, 40.6%–77.3%). GMT ratios (cLIA) for both 2D and 3D girls were noninferior to 3 doses in women for HPV6/11/16/18. Trends were comparable between assays. Conclusions GMTs for HPV6/11/16/18 after 2D or 3D of 4vHPV in girls were noninferior to 3D in adult women up to 120 months postvaccination. This study demonstrates long-term immunogenicity of the 2D HPV vaccine schedule.
Collapse
Affiliation(s)
- Robine Donken
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.,Women's Health Research Institute, BC Women's Hospital and Health Service, Vancouver, British Columbia, Canada.,Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Simon R M Dobson
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kim D Marty
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Darrel Cook
- Public Health Laboratory, BC Centre for Disease Control, Vancouver, British Columbia, Canada
| | - Chantal Sauvageau
- Institut National de Sante Publique du Quebec, Quebec, Canada.,Department of Social and Preventive Medicine, Laval University, Quebec, Canada
| | - Vladimir Gilca
- Institut National de Sante Publique du Quebec, Quebec, Canada
| | - Marc Dionne
- Department of Social and Preventive Medicine, Laval University, Quebec, Canada
| | - Shelly McNeil
- Canadian Center for Vaccinology, IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Nova Scotia, Canada
| | - Mel Krajden
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Public Health Laboratory, BC Centre for Disease Control, Vancouver, British Columbia, Canada
| | - Deborah Money
- Women's Health Research Institute, BC Women's Hospital and Health Service, Vancouver, British Columbia, Canada.,Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - James Kellner
- Alberta Children's Hospital Infectious Diseases Epidemiology & Vaccine Evaluation Team, Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
| | - David W Scheifele
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.,Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tobias Kollmann
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.,Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Julie A Bettinger
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.,Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shuzhen Liu
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joel Singer
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Monika Naus
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Public Health Laboratory, BC Centre for Disease Control, Vancouver, British Columbia, Canada
| | - Manish Sadarangani
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Gina S Ogilvie
- Women's Health Research Institute, BC Women's Hospital and Health Service, Vancouver, British Columbia, Canada.,Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
44
|
Mboumba Bouassa RS, Gubavu C, Veyer D, Robin L, Gravier A, Hocqueloux L, Prazuck T, Péré H, Bélec L. High Prevalence of Cervical High-Risk Human Papillomavirus Harboring Atypical Genotypes in Human Immunodeficiency Virus -Infected and -Uninfected First-Generation Adult Immigrant Women Originating from Sub-Saharan Africa and Living in France. J Immigr Minor Health 2021; 23:308-319. [PMID: 32816173 PMCID: PMC7914190 DOI: 10.1007/s10903-020-01074-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Human papillomavirus (HPV)-related cervical lesions in first-generation immigrant African women in France should reflect the epidemiology of high-risk (HR)-human papillomavirus (HPV) infection in sub-Saharan Africa. First-generation immigrant African women attending the Centre Hospitalier Régional of Orléans, France, were prospectively subjected to endocervical swabs for HPV DNA PCR and Pap smear. Fifty women (mean age, 41.7 years) living in France (mean stay, 10.7 years) were enrolled, including 26.0% of HIV-negative women from general population and 74.0% of women with known HIV infection. Cervical HPV prevalence was 68.0%, with 56.0% of HR-HPV. HR-HPV -68 and -58 were the predominant genotypes (20.0% and 14.0%, respectively). HR-HPV-16 and HR-HPV-18 were infrequently detected. HIV-infected women showed a trend to be more frequently infected by HPV than HIV-negative women (70.3% versus 61.5%). Most women (84.0%) showed normal cytology, while the remaining (16.0%) exhibited cervical abnormalities and were frequently HIV-infected (87.5%). These observations highlight the unsuspected high burden of cervical HR-HPV infections mostly associated with atypical genotypes, HIV infection and cervical abnormalities in first-generation immigrant African women living in France.
Collapse
Affiliation(s)
- Ralph-Sydney Mboumba Bouassa
- Laboratoire de Virologie, Hôpital Européen Georges Pompidou, Paris, France
- Faculté de Médecine Paris Descartes, Université Paris Descartes (Paris V), Sorbonne Paris Cité, Paris, France
- École Doctorale en Infectiologie Tropicale, Franceville, Gabon
- INSERM U970, Paris Cardiovascular Research Centre, Université Paris-Descartes, Sorbonne Paris Cité, Hôpital Européen Georges Pompidou, Paris, France
| | - Camelia Gubavu
- Service Des Maladies Infectieuses Et Tropicales, Centre Hospitalier Régional D'Orléans, La Source, France
| | - David Veyer
- Laboratoire de Virologie, Hôpital Européen Georges Pompidou, Paris, France
- Faculté de Médecine Paris Descartes, Université Paris Descartes (Paris V), Sorbonne Paris Cité, Paris, France
| | - Leman Robin
- Laboratoire de Virologie, Hôpital Européen Georges Pompidou, Paris, France
- Faculté de Médecine Paris Descartes, Université Paris Descartes (Paris V), Sorbonne Paris Cité, Paris, France
| | - Anne Gravier
- Service Des Maladies Infectieuses Et Tropicales, Centre Hospitalier Régional D'Orléans, La Source, France
| | - Laurent Hocqueloux
- Service Des Maladies Infectieuses Et Tropicales, Centre Hospitalier Régional D'Orléans, La Source, France
| | - Thierry Prazuck
- Service Des Maladies Infectieuses Et Tropicales, Centre Hospitalier Régional D'Orléans, La Source, France
| | - Hélène Péré
- Laboratoire de Virologie, Hôpital Européen Georges Pompidou, Paris, France
- Faculté de Médecine Paris Descartes, Université Paris Descartes (Paris V), Sorbonne Paris Cité, Paris, France
- INSERM U970, Paris Cardiovascular Research Centre, Université Paris-Descartes, Sorbonne Paris Cité, Hôpital Européen Georges Pompidou, Paris, France
| | - Laurent Bélec
- Laboratoire de Virologie, Hôpital Européen Georges Pompidou, Paris, France.
- Faculté de Médecine Paris Descartes, Université Paris Descartes (Paris V), Sorbonne Paris Cité, Paris, France.
- INSERM U970, Paris Cardiovascular Research Centre, Université Paris-Descartes, Sorbonne Paris Cité, Hôpital Européen Georges Pompidou, Paris, France.
| |
Collapse
|
45
|
Infante V, Miyaji KT, Soarez PC, Sartori AMC. Systematic review and meta-analysis of HPV vaccination in women with systemic lupus erythematosus (SLE). Expert Rev Vaccines 2021; 20:309-318. [PMID: 33573404 DOI: 10.1080/14760584.2021.1889375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVE We conducted a systematic review and meta-analysis to access HPV vaccines' safety and immunogenicity in Systemic Lupus Erythematosus (SLE) women. METHODS The search was conducted in the most relevant databases. Meta-analyses to evaluate seroconversion rates for each HPV vaccine type and SLE flare rates after vaccination were performed. RESULTS We identified 3,467 articles; six papers referring to SLE population were included. Five articles that evaluated vaccine immunogenicity at 7th month after enrollment were included in the meta-analysis. Overall seroconversion rates among SLE participants were 89.3% (95%CI, 0.76-1.00) for HPV6; 92.4% (95%CI, 0.82-1.00) for HPV11; 96.4% (95%CI, 0.93-1.00) for HPV16; and 91.8% (95%CI, 0.85-1.00) for HPV18. Five studies were included in the qualitative analysis of vaccines safety. Pain at the injection site was the most common adverse event (AE). Just one study reported serious AE not related to the vaccine. Flare rate after HPV vaccination was 12,6% (95% CI, 0.04-0.21). CONCLUSION Few studies, small sample size, evaluated HPV vaccines in SLE women. Seroconversion rates in SLE women were like healthy women, but anti-HPV geometric mean titers (GMT) were slightly lower in SLE women. HPV vaccines were safe in this population.
Collapse
Affiliation(s)
- Vanessa Infante
- Departamento De Molestias Infecciosas E Parasitarias, Faculdade De Medicina Da Universidade De Sao Paulo, São Paulo, Brazil
| | - Karina Takesaki Miyaji
- Departamento De Molestias Infecciosas E Parasitarias, Faculdade De Medicina Da Universidade De Sao Paulo, São Paulo, Brazil
| | - Patricia Coelhode Soarez
- Departamento De Medicina Preventiva, Faculdade De Medicina Da Universidade De Sao Paulo, São Paulo, Brazil
| | - Ana Marli Christovam Sartori
- Departamento De Molestias Infecciosas E Parasitarias, Faculdade De Medicina Da Universidade De Sao Paulo, São Paulo, Brazil
| |
Collapse
|
46
|
Oumeslakht L, Ababou M, Badaoui B, Qmichou Z. Worldwide genetic variations in high-risk human papillomaviruses capsid L1 gene and their impact on vaccine efficiency. Gene 2021; 782:145533. [PMID: 33636291 DOI: 10.1016/j.gene.2021.145533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/14/2020] [Accepted: 02/09/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND Human papillomavirus is the most common sexually transmitted infection. It is associated with different cancers, mainly cervical cancer, which remains the fourth most frequent cancer among women worldwide; it is also related to anogenital (anus, vulvar, vagina, and penis) and oropharyngeal cancers. Vaccination against HPV infection is the major way of prevention, and it has demonstrated impressive efficacy in reducing cervical cancer incidence. Nowadays, all the licensed HPV recombinant vaccines were designed based on HPV major capsid L1 protein. However, some variations in the HPV L1 gene sequence may induce structural changes within the L1 protein, which may alter the affinity and interaction of monoclonal antibodies (MAbs) with L1 protein epitopes, and influence host immune response and recognition. Hence, the importance of accuracy in delineating epitopes relevant to vaccine design and defining genetic variations within antigenic regions in the L1 gene to predict its impact on prophylactic vaccine efficiency. The present review reports the sequence variations in HR-HPV L1 gene isolates from different countries around the world, which may help to understand the effect of HPV L1 gene variations on vaccine efficiency. METHODS Research studies were retrieved from PubMed, Google Scholar, Science direct, and the National Center for Biotechnology Information (NCBI) database. A total of 31 articles describing genetic variations within the major capsid L1 gene and conducted in Africa, Europe, America and Asia were found. Only 26 studies conducted on HPV16, 18, 31, 33, 58, 45 and 52 which are the targets of HPV prophylactic vaccines, and which reported genetic variations within the L1 gene, were selected and evaluated in this review. FINDINGS We found a total of 87, 49, 11, 7, 22, 3, and 17 non-synonymous single nucleotide polymorphisms (SNPs) within HPV16, HPV18, HPV31, HPV58, HPV45, and HPV52 L1 gene, respectively. Four mutations were frequently observed in HPV16 L1 sequences: T353P in the HI loop, H228D in the EF loop, T266A in the FG loop, and T292A in the FG loop. Two mutations in HPV58 L1 sequences: T375N in the HI loop and L150F in the DE loop. Three mutations in HPV33 L1 sequences: T56N in the BC loop, G133S in the DE loop, T266K in the FG loop. Other mutations were found in HPV18, HPV45, and HPV52 L1 sequences. Some were found in different countries, and others were specific to a given population. Furthermore, some variations were located on peptide binding epitopes and lead to a modification of epitopes, which may influence MAbs interactions. Others need further investigations due to the lack of studies. CONCLUSION This study investigated the major capsid L1 genetic diversity of HPV16, 18, 31, 33, 58, 45, and 52 circulating in different populations around the world. Further investigations should be conducted to confirm their effect on immunogenicity and prophylactic vaccine efficiency.
Collapse
Affiliation(s)
- Loubna Oumeslakht
- Medical Biotechnology Center, Moroccan Foundation for Advanced Science, Innovation and Research, MAScIR, Rabat, Morocco; Laboratory of Biodiversity, Ecology and Genome, Department of Biology, Faculty of Sciences Rabat, Mohammed V University, Rabat, Morocco
| | - Mouna Ababou
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences Rabat, Mohammed V University, Rabat, Morocco
| | - Bouabid Badaoui
- Laboratory of Biodiversity, Ecology and Genome, Department of Biology, Faculty of Sciences Rabat, Mohammed V University, Rabat, Morocco
| | - Zineb Qmichou
- Medical Biotechnology Center, Moroccan Foundation for Advanced Science, Innovation and Research, MAScIR, Rabat, Morocco.
| |
Collapse
|
47
|
Toh ZQ, He L, Chen C, Huang A, Russell FM, Garland SM, Reyburn R, Ratu T, Tuivaga E, Frazer IH, Mulholland EK, Licciardi PV. Measurement of Human Papillomavirus-Specific Antibodies Using a Pseudovirion-Based ELISA Method. Front Immunol 2020; 11:585768. [PMID: 33193410 PMCID: PMC7655971 DOI: 10.3389/fimmu.2020.585768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/05/2020] [Indexed: 11/23/2022] Open
Abstract
Human papillomavirus (HPV) vaccines are safe and effective in preventing HPV infection and cervical precancers. Neutralizing antibodies are thought to be the primary mechanism of protection for HPV vaccines, although the exact level required for protection has not been identified. Three common serological assays used in clinical trials to measure HPV antibodies are HPV pseudovirion-based neutralization assay (PBNA), competitive or total Luminex immunoassays (cLIA or LIA) and VLP-based enzyme linked immunosorbent assays (ELISA). While PBNA is the gold-standard for measuring neutralizing antibodies (NAb), it is labor intensive. Luminex immunoassay and VLP-ELISA are rapid and high throughput, but their reagents and equipment can be difficult to source. Nevertheless, data generated from these assays generally correlate well with PBNA. Here, we described a simplified high-throughput PsV-based ELISA for HPV antibody measurement, to circumvent some of the limitations of existing assays. Using this assay, we were able to differentiate HPV-specific IgG and IgM, and found a strong correlation between HPV-specific IgG and NAb levels, as previously determined by PBNA. This assay platform is simpler and less time-consuming than PBNA. In addition, the materials can be readily produced and obtained commercially. This assay can be used as an alternative method to measure HPV antibodies.
Collapse
Affiliation(s)
- Zheng Quan Toh
- New Vaccines, Murdoch Children's Research Institute, Infection and Immunity, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Laura He
- New Vaccines, Murdoch Children's Research Institute, Infection and Immunity, Parkville, VIC, Australia
| | - Catherine Chen
- New Vaccines, Murdoch Children's Research Institute, Infection and Immunity, Parkville, VIC, Australia
| | - Angela Huang
- New Vaccines, Murdoch Children's Research Institute, Infection and Immunity, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Fiona M Russell
- New Vaccines, Murdoch Children's Research Institute, Infection and Immunity, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Suzanne M Garland
- New Vaccines, Murdoch Children's Research Institute, Infection and Immunity, Parkville, VIC, Australia.,Department of Obstetrics and Gynecology, The University of Melbourne, Parkville, VIC, Australia.,Regional WHO HPV Reference Laboratory, Centre Women's Infectious Diseases Research, The Royal Women's Hospital, Parkville, VIC, Australia
| | - Rita Reyburn
- New Vaccines, Murdoch Children's Research Institute, Infection and Immunity, Parkville, VIC, Australia
| | - Tupou Ratu
- Public Health Services, Ministry of Health and Medical Services, Suva, Fiji
| | - Evelyn Tuivaga
- Public Health Services, Ministry of Health and Medical Services, Suva, Fiji
| | - Ian H Frazer
- Faculty of Medicine, Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - E Kim Mulholland
- New Vaccines, Murdoch Children's Research Institute, Infection and Immunity, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.,Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Paul V Licciardi
- New Vaccines, Murdoch Children's Research Institute, Infection and Immunity, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
48
|
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
|
49
|
Tsang SH, Basu P, Bender N, Herrero R, Kemp TJ, Kreimer AR, Müller M, Panicker G, Pawlita M, Pinto LA, Sampson JN, Sankaranarayanan R, Schussler J, Sehr P, Sierra MS, Unger ER, Waterboer T, Hildesheim A. Evaluation of serological assays to monitor antibody responses to single-dose HPV vaccines. Vaccine 2020; 38:5997-6006. [PMID: 32713678 PMCID: PMC7429278 DOI: 10.1016/j.vaccine.2020.07.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/02/2020] [Accepted: 07/10/2020] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Whether existing serological assays are sufficiently robust to measure the lower antibody levels expected following single-dose HPV vaccination is unknown. METHODS We evaluated seven assays measuring HPV-16/18 immunological responses overall and by number of doses in 530 serum samples from participants receiving varying doses of Cervarix or Gardasil up to 36-months post-vaccination. Serum was evaluated by simplex (HPV-16 ELISA, HPV-18 ELISA), multiplex (LIA-4, VLP-MIA, M9ELISA, GST-L1), and high-throughput pseudovirion-based neutralization assays (HT-PBNA), and results were compared to the gold standard HPV-16/18 secreted alkaline phosphatase neutralization assay (SEAP-NA). Reproducibility was assessed by the coefficient of variation (CV) and intraclass correlation coefficient (ICC). Percent agreement, Pearson correlation, and weighted-kappa were used to assess validity. Determinants of seronegativity were evaluated by chi-squared test. RESULTS HPV-16: Seropositivity range was 97.1-99.5% for single dose and 98.8-99.8% overall. CV range was 4.0-18.0% for single dose and 2.9-19.5% overall. ICC range was 0.77-0.99 for single dose and 0.74-0.99 overall. Correlation with SEAP-NA range was 0.43-0.85 for single dose and 0.51-0.90 overall. Weighted-kappa range was 0.34-0.82 for single dose and 0.45-0.84 overall. HPV-18: Seropositivity range was 63.9-94.7% for single dose and 86.2-97.9% overall. CV range was 8.1-18.2% for single dose and 4.6-18.6% overall. ICC range was 0.75-0.99 for single dose and 0.83-0.99 overall. Correlation with SEAP-NA range was 0.31-0.99 for single dose and 0.27-0.96 overall. Weighted-kappa range was 0.35-0.83 for single dose and 0.45-0.84 overall. HPV-16 seronegativity was <5% for all assays. HPV-18 seronegativity range was 5.5-17.3%. For LIA-4 and GST-L1 where the proportion of seronegativity was >10%, the strongest correlates of seronegativity were receiving a single vaccine dose and receiving Gardasil. CONCLUSIONS These results support the utility of existing serological assays to monitor antibody responses following single-dose HPV vaccination.
Collapse
Affiliation(s)
- Sabrina H Tsang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Partha Basu
- Screening Group, International Agency for Research on Cancer, Lyon, France
| | - Noemi Bender
- Infection, Inflammation & Cancer Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rolando Herrero
- 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
| | - Aimée R Kreimer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Martin Müller
- Infection, Inflammation & Cancer Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gitika Panicker
- Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Michael Pawlita
- Infection, Inflammation & Cancer Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ligia A Pinto
- HPV Immunology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Joshua N Sampson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Peter Sehr
- Infection, Inflammation & Cancer Program, German Cancer Research Center (DKFZ), Heidelberg, Germany; Chemical Biology Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Monica S Sierra
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Elizabeth R Unger
- Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Tim Waterboer
- Infection, Inflammation & Cancer Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Allan Hildesheim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
50
|
Trevisan A, Candeias JMG, Thomann P, Villa LL, Franco EL, Trottier H, The Ludwig-McGill Study Group. Correlation between cervical HPV DNA detection and HPV16 seroreactivity measured with L1-only and L1+L2 viral capsid antigens. J Med Microbiol 2020; 69:960-970. [PMID: 32510304 DOI: 10.1099/jmm.0.001213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Introduction. Persistent human papillomavirus (HPV) type 16 infection is the main causal agent of cervical cancer. Most HPV infections clear spontaneously within 1-2 years. Although not all infected women develop detectable HPV antibodies, about 60-70 % seroconvert and retain their antibodies at low levels.Aim. We investigated if cervical HPV16 DNA positivity was associated with HPV16 seroreactivity measured with two different antigen formulations. We assessed if associations were influenced by co-infection with other HPV types and HPV16 viral load.Methodology. We used baseline data for women participating in the Ludwig-McGill cohort, a longitudinal investigation of the natural history of HPV infection and cervical neoplasia. The study enrolled 2462 Brazilian women from 1993 to 1997 (pre-vaccination). ELISA assays were based on L1-only or L1+L2 virus-like particles (VLPs). Seroreactivity was expressed as normalized absorbance ratios. HPV genotyping and viral load were evaluated by PCR protocols. Pearson's r was used to measure correlations between interval-scaled variables. Serological accuracy in HPV16 DNA detection was assessed using receiver operating characteristic (ROC) curves. We analysed the association between HPV DNA positivity and HPV16 seroreactivity by linear regression.Results. Correlations between L1+L2 and L1-only VLPs for detection of HPV16 were poor (r=0.43 and 0.44 for dilutions 1 : 10 and 1 : 50, respectively). The protocol with the best accuracy was L1+L2 VLPs at serum dilution 1 : 10 (ROC area=0.73, 95 % CI: 0.65-0.85). HPV16 DNA positivity was correlated with HPV16 seroreactivity and was not influenced by co-infection or viral load. To a lesser degree, HPV16 seroreactivity was correlated with infection by other Alpha-9 papillomavirus species.Conclusion. HPV16 DNA positivity and HPV16 seroreactivity are strongly correlated. L1+L2 VLPs perform better than L1-only VLPs for detecting IgG antibodies to HPV16 in women infected with HPV16 or other Alpha-9 HPV species. This study advances our understanding of humoral immune responses against HPV16 by providing insights about the influence of VLP antigen composition to measure humoral immune response against naturally acquired HPV infection.
Collapse
Affiliation(s)
- Andrea Trevisan
- Sainte-Justine Hospital Research Center, Université de Montréal, Montreal, Canada.,Department of Social and Preventive Medicine, School of Public Health, Université de Montréal, Montreal, Canada
| | - João M G Candeias
- Department of Microbiology and Immunology, Institute of Biosciences, Universidade Estadual Paulista, Botucatu, Brazil
| | | | - Luisa L Villa
- Dept of Radiology and Oncology, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo, Brazil.,Instituto do Cancer do Estado de São Paulo, Universidade de São Paulo, Sao Paulo, Brazil
| | - Eduardo L Franco
- Division of Cancer Epidemiology, McGill University, Montreal, Canada
| | - Helen Trottier
- Department of Social and Preventive Medicine, School of Public Health, Université de Montréal, Montreal, Canada.,Sainte-Justine Hospital Research Center, Université de Montréal, Montreal, Canada
| | | |
Collapse
|