1
|
Carter JJ, Hurlburt NK, Scherer EM, Singh S, Rodarte JV, Smith RA, Lewis P, Kinzelman R, Kieltyka J, Cabãn ME, Wipf GC, Pancera M, Galloway DA. HPV16 neutralizing monoclonal antibodies show evidence for common developmental pathways and public epitopes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.646278. [PMID: 40236113 PMCID: PMC11996370 DOI: 10.1101/2025.03.31.646278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Antibodies to human papillomavirus (HPV) primarily recognize surface exposed residues on five loops of the major capsid protein (L1) that vary significantly among HPV types. We determined which loops were required for neutralization for 70 HPV16 specific human monoclonal antibodies (mAbs) cloned from participants who received an HPV vaccine, and we describe molecular features of those antibodies. Chimeric HPV16 pseudovirus (cpsV), each having one surface loop bearing multiple amino acid substitutions, were used to determine neutralization specificity. The HPV16-FG-loop was the loop most frequently required for neutralization (44 of 70, 62.9%), however, all other surface loops were used for neutralization by multiple mAbs: HI (13, 18.6%), DE (15, 21.4%), EF (six, 8.6%), BC (four, 5.7%). Antibodies that required multiple loops were common (17, 24.3%). Three mAbs (4.3%) required sequences on the c-terminus of L1 and for another three mAbs the neutralization specificity could not be determined. Two types of mAbs appeared to be overrepresented: ten mAbs used V H 2-70 IGHV paired with V L λ1-40, having characteristic mutations in complementarity determining region two (CDRL2). Cryogenic electron microscopy (Cryo-EM) revealed that two of these antibodies bound five Fabs per pentamer interacting with all five L1-surface loops. The other type of mAbs that appeared to be overrepresented were ten mAbs using V H 4-34, seven of which also used D H 3-16*02 with conserved CDRH3 sequences. Cryo-EM for one of these mAbs, that required the FG-loop for neutralization, was shown to bind one Fab per pentamer at the apex, interacting with the DE- and FG-loops, with sequences of the Fab CDRH3 inserted between the DE- and FG-loops from two protomers. These two types of mAbs were found repeatedly in the four participants suggesting that these antibodies shared developmental pathways and bound to similar immunodominant epitopes on the virus. Highlights Most human mAbs recognized L1 surface loops but three of 70 recognized sequences on the C-terminal arm of L1Some antibodies induced by HPV vaccination follow shared developmental pathways. Human monoclonal antibodies using V H 2-70/V L λ1-40 were found in all participants and bound with at a stoichiometry of five Fabs per capsomer. Human monoclonal antibodies using the diversity gene segment D3-16*02 were found in all participants and one Fab was shown to bind with a stoichiometry of one Fab per capsomer. In brief A panel of 70 HPV16 specific human monoclonal antibodies (mAbs), cloned from memory B cells or plasmablasts following HPV vaccination, was characterized by determining the surface loops of the major capsid protein (L1) required for neutralization and examined for shared molecular features. All five L1 loops were found to be used for neutralization by one or more antibodies, but the most frequent target of these antibodies was the FG loop followed by the HI and DE loops. Ten antibodies paired the heavy chain variable gene V H 2-70 with the light chain variable gene V L λ1-40 and these antibodies had conserved mutations in the CDRL2 region of V L λ1-40. Mutating the CDRL2 back to the predicted germline sequence significantly reduced neutralization. Cryo-EM analysis of two V H 2-70/V L λ1-40 mAbs showed five Fabs binding per L1 pentamer and a conserved epitope with Fabs interacting with all five variable loops across two adjacent protomers. Seven other mAbs had a heavy chain composed of the variable region V H 4-34 with the diversity gene D3-16*02 resulting in the sequence motif WSGYR in the CDRH3. Mutation of that sequence to alanine ablated HPV16 neutralization activity. A cryo-EM structure of one of these antibodies showed one Fab binding the pentamer apex with the WSGYR motif inserting between three loops from two protomers. Antibodies with paired V H 2-70/V L λ1-40 and the antibodies with CDRH3 containing the WSGYR sequence, were found in all four study participants suggesting that such antibodies may be commonly elicited following HPV vaccination.
Collapse
|
2
|
Wu H, Li L, Fu K, Shen Y, Lu Y, Liao Z, Liu Y, Zha W, Wu L, Zhang Y. Effects of different-valent vaccines against human papillomavirus (HPV) to prevent persistent HPV16/18 infections and CIN2+ in women: a systematic review and network meta-analysis. Int J Infect Dis 2025; 151:107363. [PMID: 39709117 DOI: 10.1016/j.ijid.2024.107363] [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: 09/16/2024] [Revised: 12/08/2024] [Accepted: 12/12/2024] [Indexed: 12/23/2024] Open
Abstract
OBJECTIVES To evaluate the efficacy of 2-valent, 4-valent and 9-valent HPV vaccination in preventing persistent HPV infections and cervical intraepithelial neoplasia grade 2 or higher (CIN2+) lesions among women with different infection statuses at baseline. METHODS PubMed, Web of Science, Cochrane, Embase and ClinicalTrials.gov were searched from their inception to March 2024. Randomized controlled trials (RCTs) and post hoc analyses of RCTs reporting the risk of persistent HPV infections and CIN2+ among vaccinated women were included. Odds ratios (ORs) with 95% confidence intervals (CIs) were calculated to summarize the intervention effects. RESULTS Eighteen RCTs and one post hoc analysis of RCTs were included. In the according-to-protocol (ATP) cohorts, the 4-valent vaccine was the most effective against HPV16/18-related persistent infections and CIN2+ (6-month persistent infections (6mPIs): OR 0.05, 95% CI [0.02, 0.15]; 12-month persistent infections (12mPIs): OR 0.02, 95% CI [0.00, 0.18]; CIN2+: OR 0.03 95% CI [0.01, 0.17]). For the total vaccination cohorts (TVCs), the 2-valent vaccine was most effective against HPV16/18-related 12mPIs and CIN2+ (12mPIs: OR 0.15, 95% CI [0.04, 0.63]; CIN2+: OR 0.52 95% CI [0.32, 0.87]), whereas the 4-valent vaccine was most effective against HPV16/18-related 6mPIs (OR 0.08, 95% CI [0.02, 0.28]). CONCLUSIONS Vaccination against HPV can significantly reduce the risk of persistent HPV16/18-related infections and CIN2+, regardless of the HPV infection prevaccination. In addition to 4- and 9-valent vaccines, 2-valent vaccines could also provide satisfactory protection against persistent HPV16/18-related infections and CIN2+, especially over the long term, and may constitute an alternative for government-led vaccination programs in developing countries.
Collapse
Affiliation(s)
- Haiyue Wu
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, China; Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lucia Li
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, China; Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Kun Fu
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, China; Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - YuFei Shen
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, China; Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yingnan Lu
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, China; Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zexi Liao
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, China; Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yingzhen Liu
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, China; Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wenting Zha
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Lisha Wu
- Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, China
| | - Yu Zhang
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, China; Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
3
|
Han Z, Wang S, Mu T, Zhao P, Song L, Zhang Y, Zhao J, Yin W, Wu Y, Wang H, Gong B, Ji M, Roden RBS, Yang Y, Klein M, Wu K. Vaccination with a Human Papillomavirus L2 Multimer Provides Broad Protection against 17 Human Papillomavirus Types in the Mouse Cervicovaginal Challenge Model. Vaccines (Basel) 2024; 12:689. [PMID: 38932417 PMCID: PMC11209485 DOI: 10.3390/vaccines12060689] [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: 04/21/2024] [Revised: 06/11/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
Human papillomavirus (HPV) is a prevalent cause of mucosal and cutaneous infections and underlying conditions ranging from benign warts to anogenital and oropharyngeal cancers affecting both males and females, notably cervical cancer. Cervical cancer is the fourth leading cause of cancer deaths among women globally and is the most impactful in low- and middle-income countries (LMICs), where the costs of screening and licensed L1-based HPV vaccines pose significant barriers to comprehensive administration. Additionally, the licensed L1-based HPV vaccines fail to protect against all oncogenic HPV types. This study generated three independent lots of an L2-based target antigen (LBTA), which was engineered from conserved linear L2-protective epitopes (aa11-88) from five human alphapapillomavirus genotypes in E. coli under cGMP conditions and adjuvanted with aluminum phosphate. Vaccination of rabbits with LBTA generated high neutralizing antibody titers against all 17 HPV types tested, surpassing the nine types covered by Gardasil®9. Passive transfer of naïve mice with LBTA antiserum revealed its capacity to confer protection against vaginal challenge with all 17 αHPV types tested. LBTA shows stability at room temperature over >1 month. Standard in vitro and in vivo toxicology studies suggest a promising safety profile. These findings suggest LBTA's promise as a next-generation vaccine with comprehensive coverage aimed at reducing the economic and healthcare burden of cervical and other HPV+ cancers in LMICs, and it has received regulatory approval for a first-in-human clinical study (NCT05672966).
Collapse
Affiliation(s)
- Zhenwei Han
- Project Management Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China;
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, China;
| | - Shen Wang
- Regulatory and Medical Affairs Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China; (S.W.); (L.S.); (B.G.); (M.J.)
| | - Ting Mu
- Innovative Discovery Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China; (T.M.); (Y.Z.); (H.W.)
| | - Ping Zhao
- Test Development Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China; (P.Z.); (Y.W.)
| | - Lingli Song
- Regulatory and Medical Affairs Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China; (S.W.); (L.S.); (B.G.); (M.J.)
| | - Ying Zhang
- Innovative Discovery Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China; (T.M.); (Y.Z.); (H.W.)
| | - Jin Zhao
- Test Development Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China; (P.Z.); (Y.W.)
| | - Wen Yin
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, China;
| | - Yue Wu
- Test Development Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China; (P.Z.); (Y.W.)
| | - Huan Wang
- Innovative Discovery Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China; (T.M.); (Y.Z.); (H.W.)
| | - Bo Gong
- Regulatory and Medical Affairs Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China; (S.W.); (L.S.); (B.G.); (M.J.)
| | - Min Ji
- Regulatory and Medical Affairs Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China; (S.W.); (L.S.); (B.G.); (M.J.)
| | - Richard B. S. Roden
- Departments of Pathology, Oncology and Gynecology and Obstetrics, The Johns Hopkins University, Baltimore, MD 21287, USA
| | - Yanping Yang
- Executive Office, Wuhan BravoVax Co., Ltd., Wuhan 430070, China;
- Executive Office, Shanghai BravoBio Co., Ltd., Shanghai 200000, China
| | - Michel Klein
- Executive Office, Wuhan BravoVax Co., Ltd., Wuhan 430070, China;
- Executive Office, Shanghai BravoBio Co., Ltd., Shanghai 200000, China
| | - Ke Wu
- Executive Office, Wuhan BravoVax Co., Ltd., Wuhan 430070, China;
- Executive Office, Shanghai BravoBio Co., Ltd., Shanghai 200000, China
| |
Collapse
|
4
|
Akhatova A, Azizan A, Atageldiyeva K, Ashimkhanova A, Marat A, Iztleuov Y, Suleimenova A, Shamkeeva S, Aimagambetova G. Prophylactic Human Papillomavirus Vaccination: From the Origin to the Current State. Vaccines (Basel) 2022; 10:1912. [PMID: 36423008 PMCID: PMC9696339 DOI: 10.3390/vaccines10111912] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 07/30/2023] Open
Abstract
Immunization is the most successful method in preventing and controlling infectious diseases, which has helped saving millions of lives worldwide. The discovery of the human papillomavirus (HPV) infection being associated with a variety of benign conditions and cancers has driven the development of prophylactic HPV vaccines. Currently, four HPV vaccines are available on the pharmaceutical market: Cervarix, Gardasil, Gardasil-9, and the recently developed Cecolin. Multiple studies have proven the HPV vaccines' safety and efficacy in preventing HPV-related diseases. Since 2006, when the first HPV vaccine was approved, more than 100 World Health Organization member countries reported the implementation of HPV immunization. However, HPV vaccination dread, concerns about its safety, and associated adverse outcomes have a significant impact on the HPV vaccine implementation campaigns all over the world. Many developed countries have successfully implemented HPV immunization and achieved tremendous progress in preventing HPV-related conditions. However, there are still many countries worldwide which have not created, or have not yet implemented, HPV vaccination campaigns, or have failed due to deficient realization plans associated with establishing successful HPV vaccination programs. Lack of proper HPV information campaigns, negative media reflection, and numerous myths and fake information have led to HPV vaccine rejection in many states. Thus, context-specific health educational interventions on HPV vaccination safety, effectiveness, and benefits are important to increase the vaccines' acceptance for efficacious prevention of HPV-associated conditions.
Collapse
Affiliation(s)
- Ayazhan Akhatova
- School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan
| | - Azliyati Azizan
- Department of Basic Sciences, College of Osteopathic Medicine, Touro University, Henderson, NV 89014, USA
| | - Kuralay Atageldiyeva
- Department of Medicine, School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan
- Clinical Academic Department of Internal Medicine, CF University Medical Center, Astana 10000, Kazakhstan
| | - Aiymkul Ashimkhanova
- Department of Medicine, School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan
| | - Aizada Marat
- Department of Obstetrics and Gynecology #1, NJSC “Astana Medical University”, Astana 010000, Kazakhstan
| | - Yerbolat Iztleuov
- Medical Center, Marat Ospanov West-Kazakhstan Medical University, Aktobe 030000, Kazakhstan
| | - Assem Suleimenova
- Kazakh Institute of Oncology and Radiology, Almaty 050000, Kazakhstan
| | - Saikal Shamkeeva
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University Hospital, 04103 Leipzig, Germany
| | - Gulzhanat Aimagambetova
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan
| |
Collapse
|
5
|
Murenzi G, Shumbusho F, Hansen N, Munyaneza A, Gage JC, Muhoza B, Kanyabwisha F, Pierz A, Tuyisenge P, Anastos K, Castle PE. Long-term human papillomavirus vaccination effectiveness and immunity in Rwandan women living with and without HIV: a study protocol. BMJ Open 2022; 12:e061650. [PMID: 36008069 PMCID: PMC9422845 DOI: 10.1136/bmjopen-2022-061650] [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] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Prophylactic human papillomavirus (HPV) vaccines have been shown to be highly effective in protecting women against cervical infections, high-grade abnormalities and cancer caused by the targeted HPV types. However, the evidence for their effectiveness in women living with HIV (WLWH) is less clear. METHODS WLWH and HIV-negative women who likely did (birth cohorts 1996 and later) and WLWH and HIV(-) negative who likely did not (birth cohorts before 1996) receive HPV vaccination (n=3028; 757 participants for each of the four groups). Between groups, we will compare cervicovaginal, anal and oral prevalent and 6-12 month persistent HPV6/11/16/18 infections as measured using a modified AmpFire HPV genotyping assay that tests for 15 high-risk or intermediate-risk HPV genotypes, HPV6 and HPV11. We will also compare the HPV immune response in HPV-vaccinated WLWH to HPV-vaccinated HIV-negative women using an anti-HPV16 and anti-HPV18 ELISA. Vaccination status will be confirmed through national vaccination records. ANALYSIS We will calculate point prevalence and prevalence of 6-12 month persisting infections by individual HPV-type specific infections and groups of infections for each anatomic site and for each group of women. Results will be stratified by age at vaccination, age at enrolment and the number of doses (3 vs 2) as well as other factors possibly associated with HPV prevalence. Differences in endpoints between groups, overall and between subgroups, will be tested for statistical significance (p<0.05) using Fisher's exact or Pearson χ2 test. Differences in geometric mean titres and seropositivity will be tested for statistical significance using the Mann-Whitney and Fisher's exact tests, respectively. ETHICS AND DISSEMINATION The study was approved by the Albert Einstein College of Medicine Institutional Review Board and the Rwanda National Ethics Committee. Results will be disseminated through publication in peer-reviewed journals.
Collapse
Affiliation(s)
- Gad Murenzi
- Einstein-Rwanda Research and Capacity Building Program, Rwanda Military Hospital, Kigali, Rwanda
- Einstein-Rwanda Research and Capacity Building Program, Research for Development (RD Rwanda), Kigali, Rwanda
| | - Fabienne Shumbusho
- Einstein-Rwanda Research and Capacity Building Program, Rwanda Military Hospital, Kigali, Rwanda
- Einstein-Rwanda Research and Capacity Building Program, Research for Development (RD Rwanda), Kigali, Rwanda
| | - Natasha Hansen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Athanase Munyaneza
- Einstein-Rwanda Research and Capacity Building Program, Rwanda Military Hospital, Kigali, Rwanda
- Einstein-Rwanda Research and Capacity Building Program, Research for Development (RD Rwanda), Kigali, Rwanda
| | - Julia C Gage
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Benjamin Muhoza
- Einstein-Rwanda Research and Capacity Building Program, Rwanda Military Hospital, Kigali, Rwanda
- Einstein-Rwanda Research and Capacity Building Program, Research for Development (RD Rwanda), Kigali, Rwanda
| | - Faustin Kanyabwisha
- Einstein-Rwanda Research and Capacity Building Program, Rwanda Military Hospital, Kigali, Rwanda
- Einstein-Rwanda Research and Capacity Building Program, Research for Development (RD Rwanda), Kigali, Rwanda
| | - Amanda Pierz
- Department of Community Health and Health Sciences, CUNY Graduate School of Public Health and Health Policy, New York, New York, USA
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Patrick Tuyisenge
- Einstein-Rwanda Research and Capacity Building Program, Rwanda Military Hospital, Kigali, Rwanda
- Einstein-Rwanda Research and Capacity Building Program, Research for Development (RD Rwanda), Kigali, Rwanda
| | - Kathryn Anastos
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Medicine, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, USA
| | - Philip E Castle
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
6
|
Nicoli F, Mantelli B, Gallerani E, Telatin V, Squarzon L, Masiero S, Gavioli R, Palù G, Barzon L, Caputo A. Effects of the age of vaccination on the humoral responses to a human papillomavirus vaccine. NPJ Vaccines 2022; 7:37. [PMID: 35292655 PMCID: PMC8924199 DOI: 10.1038/s41541-022-00458-0] [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: 05/24/2021] [Accepted: 02/11/2022] [Indexed: 11/11/2022] Open
Abstract
Adult vaccination programs are receiving increasing attention however, little is known regarding the impact of age on the maintenance of the immune response. We investigated this issue in the context of a human papillomavirus (HPV) vaccination program collecting real-world data on the durability of humoral immunity in 315 female subjects stratified according to vaccination age (adolescents and adults) and sampled at early or late time points after the last vaccine dose. HPV-specific IgGs, but not memory B cells, were induced and maintained at higher levels in subjects vaccinated during adolescence. Nonetheless, antibody functions waned over time to a similar degree in adolescents and adults. To shed light on this phenomena, we analyzed quantitative and qualitative properties of lymphocytes. Similar biochemical features were observed between B-cell subsets from individuals belonging to the two age groups. Long term humoral responses toward vaccines administered at an earlier age were comparably maintained between adolescents and adults. The percentages of naïve B and CD4+ T cells were significantly higher in adolescents, and the latter directly correlated with IgG titers against 3 out of 4 HPV types. Our results indicate that age-specific HPV vaccine responsiveness is mostly due to quantitative differences of immune cell precursors rather than qualitative defects in B cells. In addition, our results indicate that adults also have a good humoral immunogenic profile, suggesting that their inclusion in catch-up programmes is desirable.
Collapse
Affiliation(s)
- Francesco Nicoli
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121, Ferrara, Italy
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Barbara Mantelli
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Eleonora Gallerani
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Valentina Telatin
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Laura Squarzon
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Serena Masiero
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Riccardo Gavioli
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Luisa Barzon
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Antonella Caputo
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121, Ferrara, Italy.
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy.
| |
Collapse
|
7
|
Immunogenicity of a Two-Dose Human Papillomavirus Vaccine Schedule in HIV-Infected Adolescents with Immune Reconstitution. Vaccines (Basel) 2022; 10:vaccines10010118. [PMID: 35062779 PMCID: PMC8779595 DOI: 10.3390/vaccines10010118] [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: 12/19/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/10/2022] Open
Abstract
HIV-infected patients are at increased risk of human papillomavirus (HPV) acquisition and HPV-associated diseases. This study set out to determine whether a two-dose (2D) HPV vaccination schedule was sufficient in HIV-infected adolescents with immune reconstitution (IR) following antiretroviral treatment. Participants aged 9–15 years who had CD4 cell counts > 500 cells/mm3 and HIV-1 RNA < 40 copies/mL for at least one year were assigned to the 2D schedule, while older participants or those without IR received a three-dose (3D) schedule. Antibodies to HPV-16 and -18 were measured using a pseudovirion-based neutralization assay. A total of 96 subjects were enrolled; 31.3% and 68.7% received the 2D and 3D schedule, respectively. Of these, 66.7% and 57.6% of the 2D and 3D participants, respectively, were male. The seroconversion rates for HPV-16 and HPV-18 were 100% in all cases, except for HPV-18 in males who received the 3D schedule (97.4%). In males, the anti-HPV-16 geometric mean titers (GMTs) were 6859.3 (95% confidence interval, 4394.3–10,707.1) and 7011.1 (4648.8–10,573.9) in the 2D and 3D groups (p = 0.946), respectively, and the anti-HPV-18 GMTs were 2039.3 (1432.2–2903.8) and 2859.8 (1810.0–4518.4) in the 2D and 3D (p = 0.313) groups, respectively. In females, the anti-HPV-16 GMTs were 15,758.7 (8868.0–28,003.4) and 26,241.6 (16,972.7–40,572.3) in the 2D and 3D groups (p = 0.197), respectively, and the anti-HPV-18 GMTs were 5971.4 (3026.8–11,780.6) and 9993.1 (5950.8–16,781.1) in the 2D and 3D groups (p = 0.271), respectively. In summary, a 2D schedule is as immunogenic in young adolescents with IR as a 3D schedule in older subjects and those without IR.
Collapse
|
8
|
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: 13] [Impact Index Per Article: 3.3] [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
|
9
|
Huber B, Wang JW, Roden RBS, Kirnbauer R. RG1-VLP and Other L2-Based, Broad-Spectrum HPV Vaccine Candidates. J Clin Med 2021; 10:1044. [PMID: 33802456 PMCID: PMC7959455 DOI: 10.3390/jcm10051044] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 02/24/2021] [Accepted: 02/26/2021] [Indexed: 12/19/2022] Open
Abstract
Licensed human papillomavirus (HPV) vaccines contain virus-like particles (VLPs) self-assembled from L1 major-capsid proteins that are remarkably effective prophylactic immunogens. However, the induced type-restricted immune response limits coverage to the included vaccine types, and costly multiplex formulations, restrictive storage and distribution conditions drive the need for next generation HPV vaccines. Vaccine candidates based upon the minor structural protein L2 are particularly promising because conserved N-terminal epitopes induce broadly cross-type neutralizing and protective antibodies. Several strategies to increase the immunological potency of such epitopes are being investigated, including concatemeric multimers, fusion to toll-like receptors ligands or T cell epitopes, as well as immunodominant presentation by different nanoparticle or VLP structures. Several promising L2-based vaccine candidates have reached or will soon enter first-in-man clinical studies. RG1-VLP present the HPV16L2 amino-acid 17-36 conserved neutralization epitope "RG1" repetitively and closely spaced on an immunodominant surface loop of HPV16 L1-VLP and small animal immunizations provide cross-protection against challenge with all medically-significant high-risk and several low-risk HPV types. With a successful current good manufacturing practice (cGMP) campaign and this promising breadth of activity, even encompassing cross-neutralization of several cutaneous HPV types, RG1-VLP are ready for a first-in-human clinical study. This review aims to provide a general overview of these candidates with a special focus on the RG1-VLP vaccine and its road to the clinic.
Collapse
Affiliation(s)
- Bettina Huber
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Joshua Weiyuan Wang
- Department of Pathology, The Johns Hopkins University, Baltimore, MD 21218, USA; (J.W.W.); (R.B.S.R.)
- PathoVax LLC, Baltimore, MD 21205, USA
| | - Richard B. S. Roden
- Department of Pathology, The Johns Hopkins University, Baltimore, MD 21218, USA; (J.W.W.); (R.B.S.R.)
- Department of Gynecology and Obstetrics, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Reinhard Kirnbauer
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria;
| |
Collapse
|
10
|
Stanley M, Joura E, Yen GP, Kothari S, Luxembourg A, Saah A, Walia A, Perez G, Khoury H, Badgley D, Brown DR. Systematic literature review of neutralizing antibody immune responses to non-vaccine targeted high-risk HPV types induced by the bivalent and the quadrivalent vaccines. Vaccine 2021; 39:2214-2223. [PMID: 33658126 DOI: 10.1016/j.vaccine.2021.01.060] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Studies on the cross-protective effect of HPV bivalent and quadrivalent vaccines demonstrated inconsistent findings against additional HPV types covered by the nonavalent vaccine. The objective of this study was to conduct a systematic literature review to assess the consistency and durability of the cross-protective neutralizing antibody immune responses of the currently licensed bivalent and quadrivalent vaccines to non-vaccine HPV types targeted by the nonavalent vaccine (HPV 6, 11, 31, 33, 45, 52, and 58). METHODS PubMed and EMBASE databases were searched from 2008 to 2019 to identify studies reporting antibody/immune response after vaccination with either the bivalent, quadrivalent, or nonavalent vaccine. Key outcomes were seroconversion, seropositivity or geometric mean titers against HPV types 6, 11, 31, 33, 45, 52, and 58. RESULTS Eighteen publications met inclusion criteria, reporting on 14 interventional and five observational studies. Across all studies, immune responses to non-vaccine high-risk HPV types after bivalent vaccination were higher than baseline or quadrivalent vaccine. Nonavalent vaccine elicited near total seroconversion to HPV types 31, 33, 45, 52, and 58, with seropositivity remaining near 100% up to 24 months post-dose 1. In contrast, bivalent and quadrivalent vaccination resulted in lower seroconversion levels for non-vaccine types, which waned over time. CONCLUSIONS The cross-protection antibody/immune response among participants having received all three doses of bivalent or quadrivalent vaccine is not comparable to the specific response elicited by HPV vaccine types. Even in cases where a statistically significant cross-reactive immunological response is reported, long-term data on the duration of the response beyond two years are very limited. Further, the lack of a standard for assays limits comparability of results between studies.
Collapse
Affiliation(s)
| | - Elmar Joura
- Gynecologic Oncology, Medical University Vienna, Vienna, Austria
| | - Glorian P Yen
- Center for Observational and Real-World Evidence, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Smita Kothari
- Center for Observational and Real-World Evidence, Merck & Co., Inc., Kenilworth, NJ, USA.
| | - Alain Luxembourg
- Late Stage Development Vaccines, Merck & Co., Inc., Kenilworth, NJ USA
| | - Alfred Saah
- Global Center for Scientific Affairs, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Anuj Walia
- Global Vaccines Medical Affairs, Merck & Co., Inc., Kenilworth, NJ USA
| | | | - Hanane Khoury
- Certara Evidence and Access, Montreal, Quebec, Canada
| | | | - Darron R Brown
- Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
11
|
Kuan LY, Chua SH, Pan JY, Yew YW, Tan WP. The Quadrivalent Human Papillomavirus Vaccine in Recalcitrant Acral Warts:
A Retrospective Study. ANNALS OF THE ACADEMY OF MEDICINE, SINGAPORE 2020. [DOI: 10.47102/annals-acadmedsg.2020342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Introduction: The human papillomavirus (HPV) vaccine has been reported to lead to
clinical clearance of lesions when used as an off-label treatment for recalcitrant extragenital
warts. The aim of the study is to evaluate the therapeutic and adverse effects of HPV vaccine
as an adjunctive therapy for treatment-resistant acral warts.
Methods: Patients with persistent warts despite first and second line therapies, and
subsequently receiving the quadrivalent HPV vaccine between July 2013 and June
2016 as an adjunctive treatment for recalcitrant warts at the National Skin Centre,
were included.
Results: Twenty-six patients with a median age of 34 years (range 8 to 77 years) were treated
with the HPV vaccine. Nineteen (73.1%) patients completed 3 doses of the vaccine, of whom
5 (26%) achieved complete clearance, 8 (42%) had partial clearance and 6 (32%) did not
respond to the vaccine. Among the 4 patients who received 2 doses of the vaccine, 3 (75%)
had complete clearance whereas 1 (25%) had partial improvement of their warts. None of the
patients reported adverse reactions.
Conclusion: Our study suggests a potential adjunctive role of the HPV vaccine in the
treatment of acral warts recalcitrant to conventional therapy.
Keywords: Acral warts, HPV vaccine, quadrivalent vaccine, recalcitrant warts
Collapse
|
12
|
Mboumba Bouassa RS, Péré H, Jenabian MA, Veyer D, Meye JF, Touzé A, Bélec L. Natural and vaccine-induced B cell-derived systemic and mucosal humoral immunity to human papillomavirus. Expert Rev Anti Infect Ther 2020; 18:579-607. [PMID: 32242472 DOI: 10.1080/14787210.2020.1750950] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction: Human papillomavirus (HPV) are the causative agent of mucosal neoplasia. Both cervical, anal and oropharyngeal cancers incidence is constantly increasing, making the HPV infection, a significant worldwide concern. Together, the CD8+ T cytotoxic cell-mediated response and the HPV-specific antibody response control most of the HPV infections before the development of cancers.Areas covered: We searched the MEDLINE and EMBASE databases and identified 228 eligible studies from 1987 to 2019 which examines both naturally acquired and vaccine induced humoral immunity against HPV infection in female and male subjects from worldwide origin. Herein, we synthesize current knowledge on the features of systemic and mucosal humoral immunity against HPV. We discuss the issues of the balance between the viral clearance or the escape to the host immune response, the differences between natural and vaccine-induced HPV-specific antibodies and their neutralizing capability. We also discuss the protection afforded after natural infection or following prophylactic vaccination.Expert opinion: Understanding the antibody response induced by HPV infection has led to the design of first-generation prophylactic vaccines. Now, prophylactic vaccination induces protective and long-lasting antibody response which would also strengthened the natural moderate humoral response in people previously exposed to the virus.
Collapse
Affiliation(s)
- Ralph-Sydney Mboumba Bouassa
- Laboratoire De Virologie, Assistance Publique-Hôpitaux De Paris (AP-HP), Hôpital Européen Georges Pompidou, Paris, France.,Laboratoire de virologie, Ecole Doctorale Régionale En Infectiologie Tropicale, Franceville, Gabon.,INSERM UMR U970 (Immunothérapie Et Traitement Anti-angiogénique En cancérologie), Paris Centre De Recherche Cardiovasculaire (PARCC), Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Hélène Péré
- Laboratoire De Virologie, Assistance Publique-Hôpitaux De Paris (AP-HP), Hôpital Européen Georges Pompidou, Paris, France.,INSERM UMR U970 (Immunothérapie Et Traitement Anti-angiogénique En cancérologie), Paris Centre De Recherche Cardiovasculaire (PARCC), Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Faculté de Médecine, Université Paris Descartes, Paris, France
| | - Mohammad-Ali Jenabian
- Département Des Sciences Biologiques Et Centre De Recherche BioMed, Université Du Québec À Montréal (UQAM), Montreal, QC, Canada
| | - David Veyer
- Laboratoire De Virologie, Assistance Publique-Hôpitaux De Paris (AP-HP), Hôpital Européen Georges Pompidou, Paris, France
| | - Jean-François Meye
- Service De Gynécologie Obstétrique, Centre Hospitalo-Universitaire d'Agondjé Et Faculté De Médecine De Libreville, Université Des Sciences De La Santé, Libreville, Gabon
| | - Antoine Touzé
- UMRINRA ISP 1282, Equipe Biologie Des Infections À Polyomavirus, Université De Tours, Tours, France
| | - Laurent Bélec
- Laboratoire De Virologie, Assistance Publique-Hôpitaux De Paris (AP-HP), Hôpital Européen Georges Pompidou, Paris, France.,INSERM UMR U970 (Immunothérapie Et Traitement Anti-angiogénique En cancérologie), Paris Centre De Recherche Cardiovasculaire (PARCC), Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Faculté de Médecine, Université Paris Descartes, Paris, France
| |
Collapse
|
13
|
Nicoli F, Mantelli B, Gallerani E, Telatin V, Bonazzi I, Marconi P, Gavioli R, Gabrielli L, Lazzarotto T, Barzon L, Palù G, Caputo A. HPV-Specific Systemic Antibody Responses and Memory B Cells are Independently Maintained up to 6 Years and in a Vaccine-Specific Manner Following Immunization with Cervarix and Gardasil in Adolescent and Young Adult Women in Vaccination Programs in Italy. Vaccines (Basel) 2020; 8:vaccines8010026. [PMID: 31947611 PMCID: PMC7175219 DOI: 10.3390/vaccines8010026] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 12/26/2022] Open
Abstract
Human papillomavirus (HPV) persistent infections are associated with cervical cancer and other HPV-related diseases and tumors. Thus, the characterization of long lasting immunity to currently available HPV vaccines is important. A total of 149 female subjects vaccinated with Cervarix or Gardasil participated to the study and they were stratified according to age (10–12-year-old and 16–20-year-old). Humoral immune responses (IgG and neutralizing antibody titers, antibody avidity) and circulating memory B cells were analyzed after an average of 4–6 years from the third immunization. The humoral responses against HPV-16 and HPV-18 (and HPV-6 and HPV-11 for Gardasil) were high in both age groups and vaccines up to six years from the third dose. However, Cervarix induced significantly higher and more persistent antibody responses, while the two vaccines were rather equivalent in inducing memory B cells against HPV-16 and HPV-18. Moreover, the percentage of subjects with vaccine-specific memory B cells was even superior among Gardasil vaccinees and, conversely, Cervarix vaccinated individuals with circulating antibodies, but undetectable memory B cells were found. Finally, a higher proportion of Cervarix-vaccinated subjects displayed cross-neutralizing responses against non-vaccine types HPV-31 and HPV-45. Gardasil and Cervarix may, thus, differently affect long-lasting humoral immunity from both the quantitative and qualitative point of view.
Collapse
Affiliation(s)
- Francesco Nicoli
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, 44121 Ferrara, Italy; (F.N.); (E.G.); (P.M.); (R.G.)
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (B.M.); (V.T.); (I.B.); (L.B.); (G.P.)
| | - Barbara Mantelli
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (B.M.); (V.T.); (I.B.); (L.B.); (G.P.)
| | - Eleonora Gallerani
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, 44121 Ferrara, Italy; (F.N.); (E.G.); (P.M.); (R.G.)
| | - Valentina Telatin
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (B.M.); (V.T.); (I.B.); (L.B.); (G.P.)
| | - Irene Bonazzi
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (B.M.); (V.T.); (I.B.); (L.B.); (G.P.)
| | - Peggy Marconi
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, 44121 Ferrara, Italy; (F.N.); (E.G.); (P.M.); (R.G.)
| | - Riccardo Gavioli
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, 44121 Ferrara, Italy; (F.N.); (E.G.); (P.M.); (R.G.)
| | - Liliana Gabrielli
- Operative Unit of Clinical Microbiology, St Orsola-Malpighi University Hospital, 40138 Bologna, Italy;
| | - Tiziana Lazzarotto
- Department of Specialized, Experimental and Diagnostic Medicine, University of Bologna, 40138 Bologna, Italy;
| | - Luisa Barzon
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (B.M.); (V.T.); (I.B.); (L.B.); (G.P.)
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (B.M.); (V.T.); (I.B.); (L.B.); (G.P.)
| | - Antonella Caputo
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, 44121 Ferrara, Italy; (F.N.); (E.G.); (P.M.); (R.G.)
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (B.M.); (V.T.); (I.B.); (L.B.); (G.P.)
- Correspondence: ; Tel.: +39-0532-974410
| |
Collapse
|
14
|
Selective Persistence of HPV Cross-Neutralising Antibodies following Reduced-Dose HPV Vaccine Schedules. Vaccines (Basel) 2019; 7:vaccines7040200. [PMID: 31795211 PMCID: PMC6963583 DOI: 10.3390/vaccines7040200] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/20/2019] [Accepted: 11/26/2019] [Indexed: 12/21/2022] Open
Abstract
The duration of cross-neutralising antibody responses (cross-NAb) following HPV immunisation is unknown. We compared cross-NAb responses in cohort of girls who were either unimmunised or had received immunisation with one, two or three doses of 4vHPV (Gardasil®, Merck Inc., Kenilworth, NJ, USA) six years earlier, before and one month after a booster dose of 2vHPV (Cervarix®, GSK, Brentford, UK). NAb to potentially cross-reactive HPV genotypes 31, 33, 45, 52 and 58 were measured using a HPV pseudovirion-based neutralisation assay. Girls who had previously received at least one dose of 4vHPV had significantly higher NAb titres for HPV31 when compared with unimmunised girls, whereas no difference in NAb titre was observed for four other genotypes (33, 45, 52 and 58). Following a single further immunisation with 2vHPV, NAb titres to each of the five tested HPV genotypes were comparable for girls who previously received one, two or three doses of 4vHPV, and were significantly higher than for previously unimmunised girls. Immunisation with one, two or three doses of 4vHPV induced NAb to HPV31 that persisted for six years, but there was no persistence of NAb to HPV33, 45, 52 or 58. Our results suggest that one or two doses of 4vHPV may provide long-term protection against HPV31.
Collapse
|
15
|
Artemchuk H, Eriksson T, Poljak M, Surcel HM, Dillner J, Lehtinen M, Faust H. Long-term Antibody Response to Human Papillomavirus Vaccines: Up to 12 Years of Follow-up in the Finnish Maternity Cohort. J Infect Dis 2019; 219:582-589. [PMID: 30239832 DOI: 10.1093/infdis/jiy545] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 09/11/2018] [Indexed: 01/11/2023] Open
Abstract
Background Most cervical cancers are caused by vaccine-preventable infections with human papillomaviruses (HPV). The HPV prophylactic vaccines Gardasil and Cervarix have been widely used for >10 years and are reported to induce high antibody levels. A head-to-head comparison of the antibody responses induced by the 2 vaccines has been performed only up to 5 years. Methods Among 3300 Finnish females aged 16-17 years who got 1 of the 2 HPV vaccines in phase 3 licensure trials, virtually all consented to registry-based long-term follow-up. Linkage with the Finnish Maternity Cohort found that they donated >2500 serum samples up to 12 years later. Sera of 337 (38.6%) Gardasil and 730 (30.3%) Cervarix vaccine recipients were retrieved from the Finnish Maternity Cohort biobank and type-specific anti-HPV antibody levels were determined using in-house multiplexed heparin-HPV pseudovirion Luminex assay. Results Anti-HPV-16 and anti-HPV-18 antibody levels remained stable and above natural infection-related antibody levels for up to 12 years for most vaccine recipients. The median antibody levels were higher among Cervarix recipients 7-12 years post vaccination (P < .0001). Conclusions The stability of vaccine-induced antibody levels is in accordance with the high long-term protection reported previously. The differences in antibody levels induced by the 2 vaccines imply that continued follow-up to identify possible breakthrough cases and estimation of the minimal protective levels of serum antibodies is a research priority.
Collapse
Affiliation(s)
- Hanna Artemchuk
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Mario Poljak
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Slovenia
| | - Heljä-Marja Surcel
- Faculty of Medicine, University of Oulu, Oulu, Finland.,European Science Infrastructure Services, Oulu, Finland
| | - Joakim Dillner
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Clinical Pathology/Cytology, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Matti Lehtinen
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,University of Tampere, Finland
| | - Helena Faust
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
16
|
Sauvageau C, Panicker G, Unger ER, De Serres G, Schiller J, Ouakki M, Gilca V. Priming effect of bivalent and quadrivalent vaccine for HPV 31/33/45/52: an exploratory analysis from two clinical trials. Hum Vaccin Immunother 2019; 16:590-594. [PMID: 31545130 DOI: 10.1080/21645515.2019.1669413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
The main objective of this post hoc analysis is to compare the magnitude of the immune response to HPV31/33/45/52 and 58 after a dose of 9vHPV vaccine given to naïve (previously unvaccinated) subjects and subjects previously vaccinated with a dose of 2vHPV or 4vHPV vaccine. Results from two clinical trials conducted in the same region, in comparable populations and by the same research team were included in this analysis. In study A, a dose of 9vHPV was administered 6 months after a single dose of 2vHPV as well as to naïve subjects. In study B, a dose of 9vHPV was administered 36-96 months (mean 65 months) after a single dose of 4vHPV. Blood samples were collected just before and one month post-9vHPV vaccine administration. For both studies, antibody responses were measured using the same 9-plex virus-like particle based IgG ELISA (M9ELISA). One month after 9vHPV dose administration, all subjects were seropositive to HPV 31/33/45/52 and 58. Subjects who had previously received 2vHPV or 4vHPV had significantly higher (1.8-8.0-fold) GMTs than naive subjects for HPV31/33/45/52 types but not for HPV58. GMTs to HPV31/33/45/52 and 58 were not significantly different between subjects who received a 2vHPV or 4vHPV dose prior to 9vHPV. The strong anamnestic response to one dose of 9vHPV given as late as 3-8 years after a single dose of 2vHPV or 4vHPV vaccine indicates these vaccines induced priming to types only included in the 9vHPV vaccine.
Collapse
Affiliation(s)
- Chantal Sauvageau
- Division of Biological Risks, Quebec Public Health Institute, Quebec, Quebec, Canada.,Division of Infectious Disease and Immunity, Laval University Research Hospital Center, Quebec, Quebec, Canada
| | - Gitika Panicker
- Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Georgia, Atlanta, USA
| | - Elizabeth R Unger
- Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Georgia, Atlanta, USA
| | - Gaston De Serres
- Division of Biological Risks, Quebec Public Health Institute, Quebec, Quebec, Canada.,Division of Infectious Disease and Immunity, Laval University Research Hospital Center, Quebec, Quebec, Canada
| | - John Schiller
- Laboratory of Cellular Oncology, National Cancer Institute, Bethesda, MD, USA
| | - Manale Ouakki
- Division of Biological Risks, Quebec Public Health Institute, Quebec, Quebec, Canada
| | - Vladimir Gilca
- Division of Biological Risks, Quebec Public Health Institute, Quebec, Quebec, Canada.,Division of Infectious Disease and Immunity, Laval University Research Hospital Center, Quebec, Quebec, Canada
| |
Collapse
|
17
|
Wissing MD, Burchell AN, El-Zein M, Tellier PP, Coutlée F, Franco EL. Vaccination of Young Women Decreases Human Papillomavirus Transmission in Heterosexual Couples: Findings from the HITCH Cohort Study. Cancer Epidemiol Biomarkers Prev 2019; 28:1825-1834. [PMID: 31488413 DOI: 10.1158/1055-9965.epi-19-0618] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/13/2019] [Accepted: 08/27/2019] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Vaccination against human papillomaviruses (HPV) prevents HPV infections and, consequently, cervical lesions. However, the effect of vaccination on HPV transmission within couples is unknown. METHODS We used data from HITCH, a prospective cohort study of heterosexual couples (women ages 18-24 years) in Montreal, 2005 to 2013. Vaccination history was self-reported. Genital samples were tested for HPV DNA by PCR (linear array). Type-specific viral loads were quantified using real-time PCR. OR and HR were estimated using multilevel mixed-effects logistic regression and a parametric model for interval- censored survival-time data, respectively. Differences in viral loads were evaluated using the Friedman ANOVA test. RESULTS Among 497 couples, 12, 16, and 35 women received 1, 2, or 3 vaccination doses at baseline, respectively. Median age at vaccination was 18 years. Most women (92.1%) had their first coitus before vaccination. At baseline, partner concordance of persistent HPV6/11/16/18 infections was lower in vaccinated than unvaccinated women [adjusted OR = 0.10; 95% confidence interval (CI), 0.01-0.65] but not for non α7/α9/α10-HPV types (adjusted OR = 1.00; 95% CI, 0.44-2.29). Incidence of persistent α7/α9/α10 HPV types in women was inversely associated with vaccination status at baseline (adjusted HR = 0.12; 95% CI, 0.03-0.47). Likewise, male partners of vaccinated women had a lower incidence of α7/α9/α10 HPV infections (adjusted OR = 0.22; 95% CI, 0.05-0.95). Vaccinated women with HPV 6/11/16/18 infections had lower viral loads (P = 0.001) relative to unvaccinated women. CONCLUSIONS Vaccination of sexually active women significantly reduced transmission of α7/α9/α10 HPV types in heterosexual couples. IMPACT These results underscore and quantify the positive effect of HPV vaccination on HPV transmission within heterosexual couples.
Collapse
Affiliation(s)
- Michel D Wissing
- Division of Cancer Epidemiology, Gerald Bronfman Department of Oncology, McGill University, Montreal, Québec, Canada.
| | - Ann N Burchell
- Department of Family and Community Medicine and MAP Centre for Research on Inner City Health, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
- Department of Family and Community Medicine and Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Mariam El-Zein
- Division of Cancer Epidemiology, Gerald Bronfman Department of Oncology, McGill University, Montreal, Québec, Canada
| | | | - François Coutlée
- Departement de Biologie médicale et service de microbiologie, Centre Hospitalier de l'Université de Montréal, Montreal, Québec, Canada
- Departement de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Québec, Canada
| | - Eduardo L Franco
- Division of Cancer Epidemiology, Gerald Bronfman Department of Oncology, McGill University, Montreal, Québec, Canada.
| |
Collapse
|
18
|
Toh ZQ, Kosasih J, Russell FM, Garland SM, Mulholland EK, Licciardi PV. Recombinant human papillomavirus nonavalent vaccine in the prevention of cancers caused by human papillomavirus. Infect Drug Resist 2019; 12:1951-1967. [PMID: 31308715 PMCID: PMC6613616 DOI: 10.2147/idr.s178381] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/26/2019] [Indexed: 12/26/2022] Open
Abstract
Human papillomavirus (HPV) types 16 and 18 cause 70% of cervical cancer cases globally. The nonavalent HPV vaccine (9vHPV) was licensed in 2014 and protects against the next five most common cancer-causing HPV types (HPV 31/33/45/52/58) after HPV 16/18. Phase III clinical studies have demonstrated high vaccine efficacy (>90%) against cervical, vulvar, and vaginal precancers caused by these additional types, and have shown comparable immunogenicity to the shared genotypes to quadrivalent HPV vaccine (4vHPV). Vaccine efficacy and antibody responses for 9vHPV are found to persist for at least five years while longer-term observational studies are ongoing to monitor long-term vaccine effectiveness. The implementation of 9vHPV has the potential to prevent up to 93% of cervical cancer cases, as well as a significant proportion of other HPV-related anogenital cancers. This review article summarizes the current evidence for 9vHPV in terms of vaccine efficacy against HPV infection and related anogenital precancers, safety, and immunogenicity, as well as discussing the potential impact of this vaccine on the cervical cancer burden globally.
Collapse
Affiliation(s)
- Zheng Quan Toh
- Infection and Immunity, Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Jennie Kosasih
- Infection and Immunity, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Fiona M Russell
- Infection and Immunity, Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Centre for International Child Health, Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Suzanne M Garland
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, Victoria, Australia.,Regional WHO HPV Reference Laboratory, Centre for Women's Infectious Diseases Research, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Edward K Mulholland
- Infection and Immunity, Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, University of London, London WC1E7HT, UK
| | - Paul V Licciardi
- Infection and Immunity, Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
19
|
Leung TF, Liu APY, Lim FS, Thollot F, Oh HML, Lee BW, Rombo L, Tan NC, Rouzier R, De Simoni S, Suryakiran P, Hezareh M, Thomas F, Folschweiller N, Struyf F. Comparative immunogenicity and safety of human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine and 4vHPV vaccine administered according to two- or three-dose schedules in girls aged 9-14 years: Results to month 36 from a randomized trial. Vaccine 2018; 36:98-106. [PMID: 29174109 DOI: 10.1016/j.vaccine.2017.11.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/26/2017] [Accepted: 11/13/2017] [Indexed: 12/20/2022]
Abstract
This observer-blind study (clinicaltrials.gov NCT01462357) compared the immunogenicity and safety of two doses (2D) of the HPV-16/18 AS04-adjuvanted vaccine (2D of AS04-HPV-16/18) vs. two or three doses of the 4vHPV vaccine [2D or 3D of 4vHPV] in 1075 healthy girls aged 9-14 years. Girls were randomized (1:1:1) to receive 2D of AS04-HPV-16/18 at months (M) 0, 6 (N = 359), 2D of 4vHPV at M0, 6 (N = 358) or 3D of 4vHPV at M0, 2, 6 (N = 358). 351, 339 and 346 girls, respectively, returned for the concluding visit at M36. Superiority was demonstrated at M7 and M12; comparison of the immune response to both vaccine antigens was made between 2D of AS04-HPV-16/18 and 2D or 3D of 4vHPV at subsequent time points in the according-to-protocol immunogenicity cohort (ATP-I; N = 958 at M36) and the total vaccinated cohort (TVC: N = 1036 at M36). HPV-16/18-specific T-cell- and B-cell-mediated immune responses and safety were also investigated. At M36, anti-HPV-16/18 ELISA responses in the 2D AS04-HPV-16/18 group remained superior to those of the 2D and 3D 4vHPV groups. In the M36 TVC, geometric mean titers were 2.78-fold (HPV-16) and 6.84-fold (HPV-18) higher for 2D of AS04-HPV-16/18 vs. 2D of 4vHPV and 2.3-fold (HPV-16) and 4.14-fold (HPV-18) higher vs. 3D of 4vHPV. Results were confirmed by vaccine pseudovirion-based neutralisation assay. Numbers of circulating CD4+ T cells and B cells appeared similar across groups. Safety was in line with the known safety profiles of both vaccines. In conclusion, superior HPV-16/18 antibody responses were elicited by 2D of the AS04-HPV-16/18 compared with 2D or 3D of the 4vHPV vaccine in girls aged 9-14 years. CLINICAL TRIAL REGISTRATION NCT0146235.
Collapse
Affiliation(s)
- Ting Fan Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong.
| | - Anthony Pak-Yin Liu
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Fong Seng Lim
- National Healthcare Group Polyclinics and National University of Singapore, Singapore
| | - Franck Thollot
- Association Française de Pédiatrie Ambulatoire (AFPA), Essey Les Nancy, France
| | - Helen May Lin Oh
- Division of Infectious Disease, Changi General Hospital, Singapore
| | - Bee Wah Lee
- Mount Elizabeth Medical Centre and National University of Singapore, Singapore
| | - Lars Rombo
- Centre for Clinical Research, Sormland County Council, Uppsala University and Karolinska Institute, Stockholm, Sweden
| | - Ngiap Chuan Tan
- SingHealth Polyclinics and DUKE-NUS Graduate Medical School, Singapore, Singapore
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Bissett SL, Godi A, Jit M, Beddows S. Seropositivity to non-vaccine incorporated genotypes induced by the bivalent and quadrivalent HPV vaccines: A systematic review and meta-analysis. Vaccine 2017. [PMID: 28633892 DOI: 10.1016/j.vaccine.2017.06.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Human papillomavirus vaccines have demonstrated remarkable efficacy against persistent infection and disease associated with vaccine-incorporated genotypes and a degree of efficacy against some genetically related, non-vaccine-incorporated genotypes. The vaccines differ in the extent of cross-protection against these non-vaccine genotypes. Data supporting the role for neutralizing antibodies as a correlate or surrogate of cross-protection are lacking, as is a robust assessment of the seroconversion rates against these non-vaccine genotypes. METHODS We performed a systematic review and meta-analysis of available data on vaccine-induced neutralizing antibody seropositivity to non-vaccine incorporated HPV genotypes. RESULTS Of 304 articles screened, 9 were included in the analysis representing ca. 700 individuals. The pooled estimate for seropositivity against HPV31 for the bivalent vaccine (86%; 95%CI 78-91%) was higher than that for the quadrivalent vaccine (61%; 39-79%; p=0.011). The pooled estimate for seropositivity against HPV45 for the bivalent vaccine (50%; 37-64%) was also higher than that for the quadrivalent vaccine (16%; 6-36%; p=0.007). Seropositivity against HPV33, HPV52 and HPV58 were similar between the vaccines. Mean seropositivity rates across non-vaccine genotypes were positively associated with the corresponding vaccine efficacy data reported from vaccine trials. CONCLUSIONS These data improve our understanding of vaccine-induced functional antibody specificity against non-vaccine incorporated genotypes and may help to parameterize vaccine-impact models and improve patient management in a post-vaccine setting.
Collapse
Affiliation(s)
- Sara L Bissett
- Virus Reference Department, Public Health England, London, UK
| | - Anna Godi
- Virus Reference Department, Public Health England, London, UK
| | - Mark Jit
- Modelling and Economics Unit, Public Health England, London, UK; Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Simon Beddows
- Virus Reference Department, Public Health England, London, UK.
| |
Collapse
|
21
|
Vergara N, Espinoza G, Balanda M, Quiero A, Hidalgo W, San Martín H, Ramírez A, Ramírez E. Prevalence of Human Papillomavirus infection among Chilean women from 2012 to 2016. J Med Virol 2017; 89:1646-1653. [PMID: 28294358 DOI: 10.1002/jmv.24805] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 01/24/2017] [Indexed: 02/02/2023]
Abstract
Here, we evaluated the prevalence of Human Papillomavirus (HPV) in two groups of Chilean women. The first group consisted of 3235 women aged 18-64 years attended in six primary care centers of Santiago. The second group consisted of 456 women 18-85 aged who consulted the Gynaecology Department of the Reference Hospital of Santiago. Samples were collected from October 2012 to February 2016. Cervical swabs were analyzed both HPV genotyping by PCR and Reverse Line Blot, and cervical cytology by Pap testing. Results showed a prevalence of 12.0% HPV positive, 10.3% high-risk (HR) HPV types positive, 3.9% low-risk (LR) HPV types positive, and 1.0% Pap positive in group 1. The most frequent types were 16, 66, and 59, with a prevalence of 3.0%, 1.6%, and 1.5%, respectively. The prevalence were 71.9% HPV positive, 67.3% HR-HPV types positive, 13.6% LR-HPV types positive, and 62.5% Pap positive in group 2. The most frequent types were 16, 31, and 58, with prevalence of 33.6%, 10.5%, and 7.0%, respectively. Among infected women with HPV: 7.6% were infected with HPV16 or HPV18, 3.0% with HPV31, HPV33 or HPV45, and 6.7% with any other HR-HPV. These findings show great difference in HPV prevalence and types between primary care and reference center, and provide useful epidemiological information to assess the impact of HPV vaccination in the future.
Collapse
Affiliation(s)
- Nicolás Vergara
- Sección Virus Oncogénicos, Subdepto. de Enfermedades Virales, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Gloria Espinoza
- Dirección de Atención Primaria, Servicio de Salud Metropolitano Central, Santiago, Chile
| | - Monserrat Balanda
- Sección Virus Oncogénicos, Subdepto. de Enfermedades Virales, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Andrea Quiero
- Dirección de Atención Primaria, Servicio de Salud Metropolitano Central, Santiago, Chile
| | - Wilma Hidalgo
- Dirección de Salud, Municipalidad de Huechuraba, Santiago, Chile
| | - Héctor San Martín
- Sección Virus Oncogénicos, Subdepto. de Enfermedades Virales, Instituto de Salud Pública de Chile, Santiago, Chile
| | | | - Eugenio Ramírez
- Sección Virus Oncogénicos, Subdepto. de Enfermedades Virales, Instituto de Salud Pública de Chile, Santiago, Chile.,Programa de Virología, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
22
|
Bissett SL, Godi A, Beddows S. The DE and FG loops of the HPV major capsid protein contribute to the epitopes of vaccine-induced cross-neutralising antibodies. Sci Rep 2016; 6:39730. [PMID: 28004837 PMCID: PMC5177933 DOI: 10.1038/srep39730] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 11/28/2016] [Indexed: 01/05/2023] Open
Abstract
The human papillomavirus (HPV) vaccines consist of major capsid protein (L1) virus-like particles (VLP) and are highly efficacious against the development of cervical cancer precursors attributable to oncogenic genotypes, HPV16 and HPV18. A degree of vaccine-induced cross-protection has also been demonstrated against genetically-related genotypes in the Alpha-7 (HPV18-like) and Alpha-9 (HPV16-like) species groups which is coincident with the detection of L1 cross-neutralising antibodies. In this study the L1 domains recognised by inter-genotype cross-neutralising antibodies were delineated. L1 crystallographic homology models predicted a degree of structural diversity between the L1 loops of HPV16 and the non-vaccine Alpha-9 genotypes. These structural predictions informed the design of chimeric pseudovirions with inter-genotype loop swaps which demonstrated that the L1 domains recognised by inter-genotype cross-neutralising antibodies comprise residues within the DE loop and the late region of the FG loop. These data contribute to our understanding of the L1 domains recognised by vaccine-induced cross-neutralising antibodies. Such specificities may play a critical role in vaccine-induced cross-protection.
Collapse
Affiliation(s)
- Sara L Bissett
- Virus Reference Department, Public Health England, London, UK
| | - Anna Godi
- Virus Reference Department, Public Health England, London, UK
| | - Simon Beddows
- Virus Reference Department, Public Health England, London, UK
| |
Collapse
|
23
|
Abstract
SUMMARYHuman papillomavirus (HPV) is the necessary cause of cervical cancer, the fourth most common cancer and cause of cancer-related death in females worldwide. HPV also causes anal, vaginal, vulvar, penile, and oropharyngeal cancer. Prophylactic HPV vaccines based on recombinantly expressed virus-like particles have been developed. Two first-generation, U.S. Food and Drug Administration (FDA)-approved vaccines prevent infections and disease caused by HPV16 and HPV18, the two HPV genotypes that cause approximately 70% of cervical cancer, and one of these vaccines also prevents HPV6 and HPV11, the two HPV genotypes that cause 90% of genital warts. A next-generation vaccine, recently approved by the U.S. FDA, targets HPV16, HPV18, and five additional HPV genotypes that together causes approximately 90% of cervical cancer as well as HPV6 and HPV11. In clinical trials, these vaccines have shown high levels of efficacy against disease and infections caused by the targeted HPV genotypes in adolescent females and males and older females. Data indicate population effectiveness, and therefore cost effectiveness, is highest in HPV-naive young females prior to becoming sexually active. Countries that implemented HPV vaccination before 2010 have already experienced decreases in population prevalence of targeted HPV genotypes and related anogenital diseases in women and via herd protection in heterosexual men. Importantly, after more than 100 million doses given worldwide, HPV vaccination has demonstrated an excellent safety profile. With demonstrated efficacy, cost-effectiveness, and safety, universal HPV vaccination of all young, adolescent women, and with available resources at least high-risk groups of men, should be a global health priority. Failure to do so will result in millions of women dying from avertable cervical cancers, especially in low- and middle-income countries, and many thousands of women and men dying from other HPV-related cancers.
Collapse
|
24
|
Toh ZQ, Licciardi PV, Fong J, Garland SM, Tabrizi SN, Russell FM, Mulholland EK. Reduced dose human papillomavirus vaccination: An update of the current state-of-the-art. Vaccine 2015; 33:5042-50. [DOI: 10.1016/j.vaccine.2015.07.102] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 07/23/2015] [Accepted: 07/29/2015] [Indexed: 01/16/2023]
|
25
|
Di Mario S, Basevi V, Lopalco PL, Balduzzi S, D'Amico R, Magrini N. Are the Two Human Papillomavirus Vaccines Really Similar? A Systematic Review of Available Evidence: Efficacy of the Two Vaccines against HPV. J Immunol Res 2015; 2015:435141. [PMID: 26380321 PMCID: PMC4562171 DOI: 10.1155/2015/435141] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 03/30/2015] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND When the bivalent and the quadrivalent HPV vaccines were marketed they were presented as having comparable efficacy against cervical cancer. Differences between the vaccines are HPV types included and formulation of the adjuvant. METHOD A systematic review was conducted to assess the efficacy of the two vaccines against cervical cancer. Outcomes considered were CIN2+, CIN3+, and AIS. RESULTS Nine reports (38,419 women) were included. At enrollment mean age of women was 20 years, 90% had negative cytology, and 80% were seronegative and/or DNA negative for HPV 16 or 18 (naïve women). In the TVC-naïve, VE against CIN2+ was 58% (95% CI: 35, 72); heterogeneity was detected, VE being 65% (95% CI: 54, 74) for the bivalent and 43% (95% CI: 23, 57) for the quadrivalent. VE against CIN3+ was 78% (95% CI: <0, 97); heterogeneity was substantial, VE being 93% (95% CI: 77, 98) for the bivalent and 43% (95% CI: 12, 63) for the quadrivalent. VE in the TVC was much lower. No sufficient data were available on AIS. CONCLUSIONS In naïve girls bivalent vaccine shows higher efficacy, even if the number of events detected is low. In women already infected the benefit of the vaccination seems negligible.
Collapse
Affiliation(s)
- Simona Di Mario
- SaPeRiDoc Unit, Department of Primary Health Care, Regional Health Authority of Emilia-Romagna, Viale Aldo Moro 21, 40127 Bologna, Italy
| | - Vittorio Basevi
- SaPeRiDoc Unit, Department of Primary Health Care, Regional Health Authority of Emilia-Romagna, Viale Aldo Moro 21, 40127 Bologna, Italy
| | - Pier Luigi Lopalco
- Office of Chief Scientist, European Centre for Disease Prevention and Control (ECDC), 171 83 Stockholm, Sweden
| | - Sara Balduzzi
- Statistics Unit, Department of Diagnostic and Clinical Medicine and Public Health, University of Modena & Reggio Emilia, Via del Pozzo 71, 41100 Modena, Italy
| | - Roberto D'Amico
- Statistics Unit, Department of Diagnostic and Clinical Medicine and Public Health, University of Modena & Reggio Emilia, Via del Pozzo 71, 41100 Modena, Italy
| | - Nicola Magrini
- Drug Evaluation Unit, WHO Collaborating Centre for Evidence Based Research Synthesis and Guidelines Development, Regional Health and Social Agency of Emilia-Romagna, Viale Aldo Moro 21, 40127 Bologna, Italy
| |
Collapse
|
26
|
Leung TF, Liu APY, Lim FS, Thollot F, Oh HML, Lee BW, Rombo L, Tan NC, Rouzier R, Friel D, De Muynck B, De Simoni S, Suryakiran P, Hezareh M, Folschweiller N, Thomas F, Struyf F. Comparative immunogenicity and safety of human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine and HPV-6/11/16/18 vaccine administered according to 2- and 3-dose schedules in girls aged 9-14 years: Results to month 12 from a randomized trial. Hum Vaccin Immunother 2015; 11:1689-702. [PMID: 26062002 PMCID: PMC4514190 DOI: 10.1080/21645515.2015.1050570] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 04/23/2015] [Accepted: 05/08/2015] [Indexed: 11/10/2022] Open
Abstract
This observer-blind study (clinicaltrials.gov NCT01462357) compared the immunogenicity and safety of 2 doses of the HPV-16/18 AS04-adjuvanted vaccine (HPV-16/18(2D)) vs. 2 or 3 doses of the HPV-6/11/16/18 vaccine (HPV-6/11/16/18(2D) and HPV-6/11/16/18(3D)) in healthy girls aged 9-14 y. Girls were randomized (1:1:1) to receive HPV-16/18(2D) at months (M) 0,6 (N = 359), HPV-6/11/16/18(2D) at M0,6 (N = 358) or HPV-6/11/16/18(3D) at M0,2,6 (N = 358). The primary objective was non-inferiority/superiority of HPV-16/18 antibodies by ELISA for HPV-16/18(2D) vs. HPV-6/11/16/18(2D) at M7 in the according-to-protocol immunogenicity cohort (ATP-I) and total vaccinated cohort, respectively. Secondary objectives included non-inferiority/superiority of HPV-16/18(2D) vs. HPV-6/11/16/18(3D) at M7, non-inferiority/superiority at M12, HPV-16/18 neutralizing antibodies, frequencies of T-cells/B-cells, reactogenicity and safety. Antibody responses at M7 for HPV-16/18(2D) were superior to those for HPV-6/11/16/18(2D) and HPV-6/11/16/18(3D) (lower limit of 95% confidence interval for geometric mean titer ratio (GMR) was >1): HPV-16/18(2D)/HPV-6/11/16/18(2D) GMRs were 1.69 [1.49-1.91] for anti-HPV-16 and 4.52 [3.97-5.13] for anti-HPV-18; HPV-16/18(2D)/HPV-6/11/16/18(3D) GMRs were 1.72 [1.54-1.93] for anti-HPV-16 and 3.22 [2.82-3.68] for anti-HPV-18; p = 0.0001 for all comparisons. Non-inferiority/superiority was also demonstrated at M12. Among initially seronegative girls in the ATP-I, neutralizing antibody titers were at least 1.8-fold higher for HPV-16/18(2D) vs. HPV-6/11/16/18(2D) and HPV-6/11/16/18(3D) at M7 and M12. Frequencies of HPV-16/18-specific T-cells and B-cells were in similar ranges between groups. Reactogenicity and safety were in line with the known profile of each vaccine. In conclusion, superior HPV-16/18 antibody responses were elicited by 2 doses of the HPV-16/18 AS04-adjuvanted vaccine compared with 2 or 3 doses of the HPV-6/11/16/18 vaccine in girls (9-14 years).
Collapse
Key Words
- 2D, 2-dose
- 3D, 3-dose
- AAHS, aluminum hydroxyphosphate sulfate
- ANOVA, analysis of variance
- AS04, Adjuvant System containing 50 µg 3-O-desacyl-4 ′-monophosphoryl lipid A (MPL) adsorbed on aluminum salt (500 µg Al3+)
- ATP-I, according-to-protocol immunogenicity cohort
- CI, confidence interval
- CMI, cell-mediated immunity
- ED50, effective dose producing 50% response
- ELISA, enzyme-linked immunosorbent assay
- ELISPOT, enzyme-linked immunosorbent spot assay
- EU, ELISA unit
- GMR, geometric mean titer ratio
- GMT, geometric mean antibody titer
- HPV, human papillomavirus
- HPV-16/18(2D), 2-dose schedule of the HPV-16/18 vaccine
- HPV-6/11/16/18(2D), 2-dose schedule of the HPV-6/11/16/18 vaccine
- HPV-6/11/16/18(3D), 3-dose schedule of the HPV-6/11/16/18 vaccine
- IFNγ, interferon
- IgG, immunoglobulin G
- M, month(s)
- PBMC, peripheral blood mononuclear cells
- PBNA, pseudovirion-based neutralisation assay
- SAE, serious adverse event
- TVC, total vaccinated cohort
- VLP, virus-like particle
- administration schedule
- female adolescents
- human papillomavirus (HPV) vaccines
- immunogenicity
- pIMD, potential immune-mediated disease
- safety
- y, year(s)
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adolescent
- Alphapapillomavirus/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- Child
- Female
- Human Papillomavirus Recombinant Vaccine Quadrivalent, Types 6, 11, 16, 18/administration & dosage
- Human Papillomavirus Recombinant Vaccine Quadrivalent, Types 6, 11, 16, 18/adverse effects
- Human Papillomavirus Recombinant Vaccine Quadrivalent, Types 6, 11, 16, 18/immunology
- Humans
- Immunity, Cellular
- Immunity, Humoral
- Immunization Schedule
- Papillomavirus Infections/prevention & control
- Papillomavirus Vaccines/administration & dosage
- Papillomavirus Vaccines/adverse effects
- Papillomavirus Vaccines/immunology
- Time Factors
- Vaccine Potency
Collapse
Affiliation(s)
- Ting Fan Leung
- Department of Paediatrics; The Chinese University of Hong Kong; Shatin, Hong Kong
- These authors equally contributed to the work
| | - Anthony Pak-Yin Liu
- Department of Paediatrics and Adolescent Medicine; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong
- These authors equally contributed to the work
| | - Fong Seng Lim
- National Healthcare Group Polyclinics and National University of Singapore; Singapore
| | - Franck Thollot
- Association Française de Pédiatrie Ambulatoire (AFPA); Essey Les Nancy, France
| | - Helen May Lin Oh
- Division of Infectious Disease; Changi General Hospital; Singapore
| | - Bee Wah Lee
- Mount Elizabeth Medical Center and National University of Singapore; Singapore
| | - Lars Rombo
- Centre for Clinical Research; Sormland County Council; Uppsala University and Karolinska Institute; Stockholm, Sweden
| | - Ngiap Chuan Tan
- SingHealth Polyclinics and DUKE-NUS Graduate Medical School; Singapore
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Leyh-Bannurah SR, Prugger C, de Koning MN, Goette H, Lellé RJ. Cervical human papillomavirus prevalence and genotype distribution among hybrid capture 2 positive women 15 to 64 years of age in the Gurage zone, rural Ethiopia. Infect Agent Cancer 2014; 9:33. [PMID: 25320636 PMCID: PMC4197284 DOI: 10.1186/1750-9378-9-33] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 09/16/2014] [Indexed: 11/23/2022] Open
Abstract
Background Human papillomavirus (HPV) infection is a prerequisite of cervical cancer, the leading cause of cancer mortality in Ethiopian women today. Data on Ethiopian cervical HPV prevalence and genotype distribution are rare, but essential as pre-vaccine baseline data to monitor changes after initiating HPV vaccination. The objectives of this study were to assess the cervical HPV prevalence, genotype distribution and associated correlates among female hospital outpatients in rural Ethiopia. Methods We examined a consecutive sample of 537 women 15–64 years of age in rural Ethiopia between November and December 2006. Screening for low risk (LR) and high-risk (HR) cervical HPV infection was performed and HR positive samples were genotyped with a GP5+/6 + − and SPF10-primer based system. Results The age-standardized prevalence of HPV, HPV HR and HPV LR infection was 17.3% (95% CI 14.1-20.5), 15.8% (95% CI 12.7-18.9) and 3.9% (95% CI 2.3-5.6), respectively. Among HC2 HPV HR positive infections (n = 86), the most common genotype was HPV 16 (24.4%), followed by 52 (11.6%), 56 (10.5%) and 31 (10.5%). Non-married relationship and widowhood, increasing number of lifetime sexual partners, human immunodeficiency virus infection and non-traditional housing type, but not age, were significantly associated with HR HPV infection. Conclusions These results on cervical HPV prevalence and genotype distribution may serve as baseline data in evaluating the impact of future HPV vaccination programmes in rural Ethiopia. Electronic supplementary material The online version of this article (doi:10.1186/1750-9378-9-33) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Christof Prugger
- Institute of Epidemiology and Social Medicine, University of Muenster, Muenster, Germany ; INSERM, U970, Paris Cardiovascular Research Centre, University Paris Descartes, Sorbonne Paris Cité, Paris, France
| | | | - Hartmut Goette
- Department of Molecular Diagnostics Europe, QIAGEN GmbH, Hilden, Germany
| | - Ralph J Lellé
- Department of Gynecology and Obstetrics, University Hospital of Muenster, Muenster, Germany
| |
Collapse
|