1
|
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
|
2
|
Wang R, Huang H, Yu C, Li X, Wang Y, Xie L. Current status and future directions for the development of human papillomavirus vaccines. Front Immunol 2024; 15:1362770. [PMID: 38983849 PMCID: PMC11231394 DOI: 10.3389/fimmu.2024.1362770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 06/11/2024] [Indexed: 07/11/2024] Open
Abstract
The development of human papillomavirus (HPV) vaccines has made substantive progress, as represented by the approval of five prophylactic vaccines since 2006. Generally, the deployment of prophylactic HPV vaccines is effective in preventing newly acquired infections and incidences of HPV-related malignancies. However, there is still a long way to go regarding the prevention of all HPV infections and the eradication of established HPV infections, as well as the subsequent progression to cancer. Optimizing prophylactic HPV vaccines by incorporating L1 proteins from more HPV subtypes, exploring adjuvants that reinforce cellular immune responses to eradicate HPV-infected cells, and developing therapeutic HPV vaccines used either alone or in combination with other cancer therapeutic modalities might bring about a new era getting closer to the vision to get rid of HPV infection and related diseases. Herein, we summarize strategies for the development of HPV vaccines, both prophylactic and therapeutic, with an emphasis on the selection of antigens and adjuvants, as well as implications for vaccine efficacy based on preclinical studies and clinical trials. Additionally, we outline current cutting-edge insights on formulation strategies, dosing schedules, and age expansion among HPV vaccine recipients, which might play important roles in addressing barriers to vaccine uptake, such as vaccine hesitancy and vaccine availability.
Collapse
Affiliation(s)
- Rui Wang
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Hongpeng Huang
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Chulin Yu
- 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
- Cell Culture Engineering Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
3
|
Lee HY, Xiong S, Sur A, Khang T, Vue B, Culhane-Pera KA, Pergament S, Torres MB, Koopmeiners JS, Desai J. Evaluating Human Papillomavirus eHealth in Hmong Adolescents to Promote Vaccinations: Pilot Feasibility Study. JMIR Form Res 2023; 7:e38388. [PMID: 37338961 PMCID: PMC10337404 DOI: 10.2196/38388] [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: 03/30/2022] [Revised: 12/13/2022] [Accepted: 12/13/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Human papillomavirus (HPV) is a common sexually transmitted infection, causing multiple cancers, including cervical, penile, and anal. Infection and subsequent health risks caused by HPV can be diminished by HPV vaccination. Unfortunately, vaccination rates among Hmong Americans are substantially lower than those among other racial and ethnic groups, despite having higher cervical cancer rates than non-Hispanic White women. Such disparities and sparse literature highlight the need for innovative and culturally appropriate educational interventions to improve HPV vaccine rates in Hmong Americans. OBJECTIVE We aimed to develop and evaluate the effectiveness and usability of an innovative web-based eHealth educational website, the Hmong Promoting Vaccines website (HmongHPV website), for Hmong-American parents and adolescents to improve their knowledge, self-efficacy, and decision-making capacities to obtain HPV vaccinations. METHODS Through social cognitive theory and community-based participatory action research process, we created a theory-driven and culturally and linguistically appropriate website for Hmong parents and adolescents. We conducted a pre-post intervention pilot study to assess the website's effectiveness and usability. Overall, 30 Hmong-American parent and adolescent dyads responded to questions about HPV and HPV vaccine knowledge, self-efficacy, and decision-making at preintervention, 1 week after intervention, and at the 5-week follow-up. Participants responded to survey questions about website content and processes at 1 and 5 weeks, and a subset of 20 dyad participants participated in telephone interviews 6 weeks later. We used paired t tests (2-tailed) to measure the change in knowledge, self-efficacy, and decision-making processes, and used template analysis to identify a priori themes for website usability. RESULTS Participants' HPV and HPV vaccine knowledge improved significantly from pre- to postintervention stage and follow-up. Knowledge scores increased from preintervention to 1 week after intervention for both parents (HPV knowledge, P=.01; vaccine knowledge, P=.01) and children (HPV knowledge, P=.01; vaccine knowledge, P<.001), which were sustained at the 5-week follow-up. Parents' average self-efficacy score increased from 21.6 at baseline to 23.9 (P=.007) at post intervention and 23.5 (P=.054) at follow-up. Similar improvements were observed in the teenagers' self-efficacy scores (from 30.3 at baseline to 35.6, P=.009, at post intervention and 35.9, P=.006, at follow-up). Collaborative decision-making between parents and adolescents improved immediately after using the website (P=.002) and at follow-up (P=.02). The interview data also revealed that the website's content was informative and engaging; in particular, participants enjoyed the web-based quizzes and vaccine reminders. CONCLUSIONS This theory-driven, community-based participatory action research-designed and culturally and linguistically appropriate educational website was well received. It improved Hmong parents' and adolescents' knowledge, self-efficacy, and decision-making processes regarding HPV vaccination. Future studies should examine the website's impact on HPV vaccine uptake and its potential for broader use across various settings (eg, clinics and schools).
Collapse
Affiliation(s)
- Hee Yun Lee
- School of Social Work, The University of Alabama, Tuscaloosa, AL, United States
| | - Serena Xiong
- School of Medicine, Washington University in St Louis, St Louis, MO, United States
| | - Aparajita Sur
- School of Public Health, University of Minnesota, Twin Cities, MN, United States
| | - Tounhia Khang
- SoLaHmo Partnership for Health & Wellness, Community University Health Care Center, Minneapolis, MN, United States
| | - Bai Vue
- SoLaHmo Partnership for Health & Wellness, Community University Health Care Center, Minneapolis, MN, United States
| | - Kathleen A Culhane-Pera
- SoLaHmo Partnership for Health & Wellness, Community University Health Care Center, Minneapolis, MN, United States
| | - Shannon Pergament
- SoLaHmo Partnership for Health & Wellness, Community University Health Care Center, Minneapolis, MN, United States
| | - M Beatriz Torres
- Department of Public Health, Mercyhurst University, Erie, PA, United States
| | - Joseph S Koopmeiners
- School of Public Health, University of Minnesota, Twin Cities, MN, United States
| | - Jay Desai
- Minnesota Department of Health, Saint Paul, MN, United States
| |
Collapse
|
4
|
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
|
5
|
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: 5.0] [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
|
6
|
Bhattacharya D. Instructing durable humoral immunity for COVID-19 and other vaccinable diseases. Immunity 2022; 55:945-964. [PMID: 35637104 PMCID: PMC9085459 DOI: 10.1016/j.immuni.2022.05.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/02/2022] [Accepted: 05/05/2022] [Indexed: 11/29/2022]
Abstract
Many aspects of SARS-CoV-2 have fully conformed with the principles established by decades of viral immunology research, ultimately leading to the crowning achievement of highly effective COVID-19 vaccines. Nonetheless, the pandemic has also exposed areas where our fundamental knowledge is thinner. Some key unknowns are the duration of humoral immunity post-primary infection or vaccination and how long booster shots confer protection. As a corollary, if protection does not last as long as desired, what are some ways it can be improved? Here, I discuss lessons from other infections and vaccines that point to several key features that influence durable antibody production and the perseverance of immunity. These include (1) the specific innate sensors that are initially triggered, (2) the kinetics of antigen delivery and persistence, (3) the starting B cell receptor (BCR) avidity and antigen valency, and (4) the memory B cell subsets that are recalled by boosters. I further highlight the fundamental B cell-intrinsic and B cell-extrinsic pathways that, if understood better, would provide a rational framework for vaccines to reliably provide durable immunity.
Collapse
Affiliation(s)
- Deepta Bhattacharya
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ 85724, USA.
| |
Collapse
|
7
|
Prabhu PR, Carter JJ, Galloway DA. B Cell Responses upon Human Papillomavirus (HPV) Infection and Vaccination. Vaccines (Basel) 2022; 10:vaccines10060837. [PMID: 35746445 PMCID: PMC9229470 DOI: 10.3390/vaccines10060837] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 02/01/2023] Open
Abstract
Infection with human papillomavirus (HPV) is the necessary cause of cervical cancer. Availability of vaccines against HPV makes it a highly preventable disease. HPV vaccines act through type-specific neutralizing antibodies produced by antigen-specific plasma cells known as long-lived plasma cells (LLPC). However, just as any other vaccine, success of HPV vaccine is attributed to the immunologic memory that it builds, which is largely attained through generation and maintenance of a class of B cells named memory B cells (Bmem). Both LLPCs and Bmems are important in inducing and maintaining immune memory and it is therefore necessary to understand their role after HPV vaccination to better predict outcomes. This review summarizes current knowledge of B-cell responses following HPV vaccination and natural infection, including molecular signatures associated with these responses.
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: 7] [Impact Index Per Article: 2.3] [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
|
Case Report: Posterior Uveitis after Divalent Human Papillomavirus Vaccination in an Asian Female. Optom Vis Sci 2021; 97:390-394. [PMID: 32511159 DOI: 10.1097/opx.0000000000001523] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
SIGNIFICANCE Vaccine-associated uveitis has appeared in recent years because of various vaccines, whereas cases for human papillomavirus (HPV) vaccination were rarely reported. With more and more females becoming aware of its importance and choosing HPV vaccination, much more attention should be paid on the adverse effects of it. PURPOSE The purpose of this study was to report a rare case of posterior uveitis after divalent HPV vaccination in an Asian female. CASE REPORT A 29-year-old woman presented with acute vision loss accompanied by symptoms of headache, tinnitus, and myalgia after the third injection of HPV vaccination. The best-corrected visual acuity dropped to 20/500 for both eyes, and optical coherence tomography revealed bilateral multifocal submacular fluid. A short course of oral prednisone as well as Ozurdex intravitreal injection resulted in the reversal of all signs and symptoms. CONCLUSIONS Although this case resembled Harada disease, we diagnosed it as vaccine-induced uveitis rather than coincidental autoimmune disease based on the rapid response to a short course of systemic corticosteroids. Because vaccine-induced uveitis is rare and difficult to distinguish from coincidental autoimmune disease, our case reminds eye care providers to be aware of the possible association between vaccination and a Harada-like reaction and to ask appropriately directed questions when obtaining history from young patients with uveitis. Based on this case, we also suggest Ozurdex intravitreal injection as a potential therapeutic choice, especially for patients with contraindication or personal concern to systemic corticosteroid.
Collapse
|
10
|
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
|
11
|
Pattyn J, Panicker G, Willhauck‐Fleckenstein M, Van Keer S, Téblick L, Pieters Z, Tjalma WAA, Matheeussen V, Van Damme P, Waterboer T, Unger ER, Vorsters A. Comparison of a VLP-based and GST-L1-based multiplex immunoassay to detect vaccine-induced HPV-specific antibodies in first-void urine. J Med Virol 2020; 92:3774-3783. [PMID: 32266996 PMCID: PMC7687076 DOI: 10.1002/jmv.25841] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/27/2020] [Accepted: 03/28/2020] [Indexed: 12/22/2022]
Abstract
Vaccine-induced human papillomavirus (HPV) antibodies originating from cervicovaginal secretions were recently shown to be detectable in first-void (FV) urine. This presents a novel opportunity for noninvasive sampling to monitor HPV antibody status in women participating in large epidemiological studies and HPV vaccine trials. With a view towards method optimization, this study compared the measurement of HPV antibodies in FV urine using a multiplex L1/L2 virus-like particles (VLP)-based ELISA (M4ELISA) with previously reported results using a glutathione S-transferase (GST)-L1-based immunoassay (GST-L1-MIA). We tested 53 paired FV urine and serum samples from 19- to 26-year-old healthy women, unvaccinated (n = 17) or vaccinated with either the bivalent or quadrivalent HPV-vaccine during adolescence (n = 36). HPV6/11/16/18 antibodies were measured using M4ELISA and compared with GST-L1-MIA results. Inter-assay and inter-specimen correlations were examined using the Spearman's rank test (rs). As expected, lower HPV antibody concentrations were found in FV urine than in serum. Vaccinated women had significantly higher HPV6/11/16/18 antibody levels in both FV urine and serum compared with those unvaccinated (M4ELISA; FV urine P = .0003; serum P ≤ .0001). HPV antibody levels in FV urine and serum showed a significant positive correlation (M4ELISA anti-HPV6/11/16/18, rs = 0.85/0.86/0.91/0.79, P ≤ .001). Despite assay differences, there was moderate to good correlation between M4ELISA and GST-L1-MIA (FV urine anti-HPV6/11/16/18, rs = 0.86/0.83/0.89/0.53, P ≤ .0001; serum anti-HPV6/11/16/18, rs = 0.93/0.89/0.94/0.75, P ≤ .0001). FV urine HPV antibody detection is comparable with both assays, further supporting this noninvasive sampling method as a possible option for HPV vaccine assessment. Approaches to improve the sensitivity and larger studies are warranted to determine the feasibility of FV urine for vaccine-induced HPV antibody detection.
Collapse
Affiliation(s)
- Jade Pattyn
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of AntwerpAntwerpBelgium
| | - Gitika Panicker
- Division of High‐Consequence Pathogens and PathologyNational Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention (CDC)AtlantaGeorgia
| | - Martina Willhauck‐Fleckenstein
- Infections and Cancer Epidemiology Group, Infections, Inflammation and Cancer Research Program, German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Severien Van Keer
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of AntwerpAntwerpBelgium
| | - Laura Téblick
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of AntwerpAntwerpBelgium
| | - Zoë Pieters
- Centre for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of AntwerpAntwerpBelgium
- Centre for Statistics, I‐Biostat, Hasselt UniversityHasseltBelgium
| | - Wiebren A. A. Tjalma
- Multidisciplinary Breast Clinic, Unit Gynaecologic Oncology, Department of Obstetrics and GynaecologyAntwerp University HospitalAntwerpBelgium
- Molecular Imaging, Pathology, Radiotherapy, Oncology (MIPRO), Faculty of Medicine and Health Sciences, University of AntwerpAntwerpBelgium
| | - Veerle Matheeussen
- Department of MicrobiologyAntwerp University Hospital (UZA)AntwerpBelgium
- Department of Medical Microbiology (LMM)Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of AntwerpAntwerpBelgium
- Department of Medical BiochemistryFaculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of AntwerpAntwerpBelgium
| | - Pierre Van Damme
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of AntwerpAntwerpBelgium
| | - Tim Waterboer
- Infections and Cancer Epidemiology Group, Infections, Inflammation and Cancer Research Program, German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Elizabeth R. Unger
- Division of High‐Consequence Pathogens and PathologyNational Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention (CDC)AtlantaGeorgia
| | - Alex Vorsters
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of AntwerpAntwerpBelgium
| |
Collapse
|
12
|
Hu Y, Zhang X, He Y, Ma Z, Xie Y, Lu X, Xu Y, Zhang Y, Jiang Y, Xiao H, Struyf F, Folschweiller N, Jiang J, Poncelet S, Karkada N, Jastorff A, Borys D. Long‐term persistence of immune response to the AS04‐adjuvanted HPV‐16/18 vaccine in Chinese girls aged 9‐17 years: Results from an 8‐9‐year follow‐up phase III open‐label study. Asia Pac J Clin Oncol 2020; 16:392-399. [PMID: 32780946 PMCID: PMC7754390 DOI: 10.1111/ajco.13398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 05/21/2020] [Indexed: 11/29/2022]
Abstract
Aim In 9‐17‐year‐old Chinese girls, the AS04‐adjuvanted HPV‐16/18 vaccine (AS04‐HPV‐16/18) given as three‐dose schedule induced high antibody levels, which were noninferior 1 month after the third dose to those observed in 18‐25‐year‐old Chinese women in a large efficacy study. We assessed the persistence of antibodies 8‐9 years after vaccination in the same subjects. Methods This follow‐up phase III, open‐label study (NCT03355820) included subjects who had received three doses of AS04‐HPV‐16/18 in the initial trial (NCT00996125). Serum antibody concentrations were assessed by ELISA and compared to antibody persistence observed in 18‐25‐year‐old Chinese women 6 years after first vaccination in the efficacy study (NCT00779766). Results Out of the 227 enrolled subjects, 223 were included in the per‐protocol immunogenicity analysis. Mean interval from first AS04‐HPV‐16/18 dose to blood sampling was 101.4 months (8.5 years). For antibodies against HPV‐16 and ‐18, 8.5 years after first vaccine dose all subjects remained seropositive and antibody. Geometric mean concentrations (GMCs) were 1236.3 (95% confidence interval [CI]: 1121.8; 1362.4) and 535.6 (95% CI: 478.6; 599.4) ELISA Units/mL, respectively. These seropositivity rates and antibody GMCs were higher than those observed 6 years after first vaccination of 18‐25‐year‐old women. Conclusion Sustained anti‐HPV‐16 and ‐18 immune responses were observed 8‐9 years after AS04‐HPV‐16/18 vaccination of 9‐17 year‐old Chinese girls that were higher than the ones observed 6 years after first vaccination in Chinese adult women in whom AS04‐HPV‐16/18 efficacy against cervical intraepithelial neoplasia of grade ≥2 was demonstrated.
Collapse
Affiliation(s)
- Yuemei Hu
- Jiangsu Province Center for Disease Control and Prevention Jiangsu China
| | - Xiang Zhang
- Taizhou City Center for Disease Control and Prevention Taizhou China
| | - Yilin He
- Taizhou City Center for Disease Control and Prevention Taizhou China
| | - Zhilong Ma
- Taizhou City Center for Disease Control and Prevention Taizhou China
| | - Yan Xie
- Taizhou City Center for Disease Control and Prevention Taizhou China
| | - Xiangbin Lu
- Taizhou City Center for Disease Control and Prevention Taizhou China
| | - Yabin Xu
- Taizhou City Center for Disease Control and Prevention Taizhou China
| | - Yanqiu Zhang
- Taizhou City Center for Disease Control and Prevention Taizhou China
| | - Yunyu Jiang
- Taizhou City Center for Disease Control and Prevention Taizhou China
| | - Hui Xiao
- Taizhou City Center for Disease Control and Prevention Taizhou China
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Bruce MG, Meites E, Bulkow L, Panicker G, Hurlburt D, Lecy D, Thompson G, Rudolph K, Unger ER, Hennessy T, Markowitz LE. A prospective cohort study of immunogenicity of quadrivalent human papillomavirus vaccination among Alaska Native Children, Alaska, United States. Vaccine 2020; 38:6585-6591. [PMID: 32814639 DOI: 10.1016/j.vaccine.2020.08.005] [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] [Received: 04/27/2020] [Revised: 07/31/2020] [Accepted: 08/02/2020] [Indexed: 11/26/2022]
Abstract
OBJECTIVE In the United States, HPV vaccination is routinely recommended at age 11 or 12 years; the series can be started at age 9. We conducted a cohort study to assess long-term immunogenicity of quadrivalent HPV vaccine (4vHPV) in an American Indian/Alaska Native (AI/AN) Indigenous population. METHODS During 2011-2014, we enrolled AI/AN girls and boys aged 9-14 years, who were vaccinated with a 3-dose series of 4vHPV. Serum specimens were collected at five time points: immediately prior to doses 2 and 3, and at one month, one year, and two years after series completion. Antibody testing was performed using a multiplex virus-like-particle-IgG ELISA for 4vHPV types (HPV 6/11/16/18). RESULTS Among 477 children (405 girls/72 boys) completing the 3-dose series, median age at enrollment was 11.2 years. Of the 477, 72 (15%) were tested before dose 2 and 70 (15%) before dose 3. Following series completion, 435 (91%) were tested at one month, 382 (80%) at one year, and 351 (74%) at two years. All tested participants had detectable antibody to 4vHPV types at all time points measured. Geometric mean concentrations (GMCs) for 4vHPV types at one month and two years post-series completion were 269.9 and 32.7 AU/ml for HPV6, 349.3 and 42.9 AU/ml for HPV11, 1240.2 and 168.3 IU/ml HPV16, and 493.2 and 52.2 IU/ml for HPV18. Among children tested after each dose, GMCs after doses 1 and 2 were 3.9 and 32.2 AU/ml for HPV6, 5.3 and 45.6 AU/ml for HPV11, 20.8 and 187.9 IU/ml for HPV16; and 6.6 and 49.7 IU/ml for HPV18. No serious adverse events were reported. CONCLUSION All AI/AN children developed antibodies to all 4vHPV types after vaccination. GMCs rose after each dose, then decreased to a plateau over the subsequent two years. This cohort will continue to be followed to determine duration of antibody response.
Collapse
Affiliation(s)
- Michael G Bruce
- Arctic Investigations Program, Division of Preparedness and Emerging Infections, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Anchorage, AK, USA.
| | - Elissa Meites
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Lisa Bulkow
- Arctic Investigations Program, Division of Preparedness and Emerging Infections, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Anchorage, AK, USA
| | - Gitika Panicker
- Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Debby Hurlburt
- Arctic Investigations Program, Division of Preparedness and Emerging Infections, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Anchorage, AK, USA
| | - Danielle Lecy
- Arctic Investigations Program, Division of Preparedness and Emerging Infections, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Anchorage, AK, USA
| | - Gail Thompson
- Arctic Investigations Program, Division of Preparedness and Emerging Infections, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Anchorage, AK, USA
| | - Karen Rudolph
- Arctic Investigations Program, Division of Preparedness and Emerging Infections, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Anchorage, AK, USA
| | - Elizabeth R Unger
- Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Thomas Hennessy
- Arctic Investigations Program, Division of Preparedness and Emerging Infections, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Anchorage, AK, USA
| | - Lauri E Markowitz
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| |
Collapse
|
14
|
Pattyn J, Van Keer S, Téblick L, Van Damme P, Vorsters A. Non-invasive Assessment of Vaccine-Induced HPV Antibodies via First-Void Urine. Front Immunol 2020; 11:1657. [PMID: 32849573 PMCID: PMC7419594 DOI: 10.3389/fimmu.2020.01657] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/22/2020] [Indexed: 12/26/2022] Open
Abstract
The potential of first-void (FV) urine as a non-invasive method to monitor human papillomavirus (HPV) vaccination has been reported, mainly focusing on urine as a sample to assess HPV DNA. Besides HPV DNA, vaccine-induced HPV antibodies originating from cervicovaginal secretions were recently shown to be detectable in FV urine as well. This presents a novel opportunity for non-invasive sampling to monitor HPV antibody status in women participating in large epidemiological studies and HPV vaccine trials. The simultaneous assessment of both HPV infection and immunogenicity on a non-invasive, readily obtained sample is particularly attractive.
Collapse
Affiliation(s)
- Jade Pattyn
- Faculty of Medicine and Health Sciences, Centre for the Evaluation of Vaccination, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Severien Van Keer
- Faculty of Medicine and Health Sciences, Centre for the Evaluation of Vaccination, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Laura Téblick
- Faculty of Medicine and Health Sciences, Centre for the Evaluation of Vaccination, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Pierre Van Damme
- Faculty of Medicine and Health Sciences, Centre for the Evaluation of Vaccination, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Alex Vorsters
- Faculty of Medicine and Health Sciences, Centre for the Evaluation of Vaccination, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
15
|
Lai L, Ault K, Rouphael N, Beck A, Domjahn B, Xu Y, Anderson EJ, Cheng A, Nakamura A, Hoagland RJ, Kelley C, Edupuganti S, Mask K, Nesin M, Unger ER, Panicker G, David H, Mulligan MJ. Duration of Cellular and Humoral Responses after Quadrivalent Human Papillomavirus Vaccination in Healthy Female Adults with or without Prior Type 16 and/or 18 Exposure. Vaccines (Basel) 2020; 8:vaccines8030348. [PMID: 32629943 PMCID: PMC7563427 DOI: 10.3390/vaccines8030348] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/22/2020] [Accepted: 06/24/2020] [Indexed: 12/14/2022] Open
Abstract
Human papillomavirus virus (HPV) vaccines aim to provide durable protection and are ideal to study the association of cellular with humoral responses. We assessed the duration and characteristics of immune responses provided by the quadrivalent HPV (4vHPV) vaccine in healthy female adults with or without prior exposure with type 16 and 18 HPV. In a prospective cohort, vaccine naïve females received three doses of 4vHPV vaccine and were followed for two years to assess cellular (intracellular cytokine staining, proliferation and B cell ELISpot assays) and humoral (multiplex L1/L2 viral-like particles (VLP) and M4 ELISAs) responses. Frequencies of vaccine-specific CD4+ T cells correlated with antibody responses. Higher HPV antibody titers were found at all time points in participants previously exposed to HPV, except for anti-HPV-18 at Day 187 (one week post the third vaccination). Retrospective cohorts enrolled females who had previously received two or three 4vHPV doses and tested antibody titers by M4 ELISA and pseudovirion neutralization assay along with memory B cells (MBCs). Almost all women enrolled in a retrospective cohort with two prior doses and all women enrolled in a retrospective cohort with three prior doses had sustained antibody and memory responses. Our findings indicate that HPV vaccination induces a long-lasting, robust cellular and humoral immune responses.
Collapse
Affiliation(s)
- Lilin Lai
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, 500 Irvin Court Suite 200, Decatur, GA 30030, USA; (L.L.); (A.B.); (B.D.); (Y.X.); (A.C.); (C.K.); (S.E.); (K.M.); (M.J.M.)
| | - Kevin Ault
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA;
| | - Nadine Rouphael
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, 500 Irvin Court Suite 200, Decatur, GA 30030, USA; (L.L.); (A.B.); (B.D.); (Y.X.); (A.C.); (C.K.); (S.E.); (K.M.); (M.J.M.)
- Correspondence: ; Tel.: +1-404-712-1435
| | - Allison Beck
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, 500 Irvin Court Suite 200, Decatur, GA 30030, USA; (L.L.); (A.B.); (B.D.); (Y.X.); (A.C.); (C.K.); (S.E.); (K.M.); (M.J.M.)
| | - Briyana Domjahn
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, 500 Irvin Court Suite 200, Decatur, GA 30030, USA; (L.L.); (A.B.); (B.D.); (Y.X.); (A.C.); (C.K.); (S.E.); (K.M.); (M.J.M.)
| | - Yongxian Xu
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, 500 Irvin Court Suite 200, Decatur, GA 30030, USA; (L.L.); (A.B.); (B.D.); (Y.X.); (A.C.); (C.K.); (S.E.); (K.M.); (M.J.M.)
| | - Evan J. Anderson
- Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Drive NE, Atlanta, GA 30322, USA;
| | - Andrew Cheng
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, 500 Irvin Court Suite 200, Decatur, GA 30030, USA; (L.L.); (A.B.); (B.D.); (Y.X.); (A.C.); (C.K.); (S.E.); (K.M.); (M.J.M.)
| | - Aya Nakamura
- The EMMES Company, LLC, 401 N. Washington St., Suite 700, Rockville, MD 20850, USA;
| | | | - Colleen Kelley
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, 500 Irvin Court Suite 200, Decatur, GA 30030, USA; (L.L.); (A.B.); (B.D.); (Y.X.); (A.C.); (C.K.); (S.E.); (K.M.); (M.J.M.)
| | - Srilatha Edupuganti
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, 500 Irvin Court Suite 200, Decatur, GA 30030, USA; (L.L.); (A.B.); (B.D.); (Y.X.); (A.C.); (C.K.); (S.E.); (K.M.); (M.J.M.)
| | - Karen Mask
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, 500 Irvin Court Suite 200, Decatur, GA 30030, USA; (L.L.); (A.B.); (B.D.); (Y.X.); (A.C.); (C.K.); (S.E.); (K.M.); (M.J.M.)
| | - Mirjana Nesin
- Division of Microbiology and Infectious Diseases, NIAID, NIH, 5601 Fishers Lane, Rockville, MD 20892-9825, USA; (M.N.); (H.D.)
| | - Elizabeth R. Unger
- Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, USA; (E.R.U.); (G.P.)
| | - Gitika Panicker
- Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, USA; (E.R.U.); (G.P.)
| | - Hagit David
- Division of Microbiology and Infectious Diseases, NIAID, NIH, 5601 Fishers Lane, Rockville, MD 20892-9825, USA; (M.N.); (H.D.)
| | - Mark J. Mulligan
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, 500 Irvin Court Suite 200, Decatur, GA 30030, USA; (L.L.); (A.B.); (B.D.); (Y.X.); (A.C.); (C.K.); (S.E.); (K.M.); (M.J.M.)
- New York University Langone Vaccine Center, Alexandria Center for Life Sciences (West Tower), 430 E 29th St, Room 304, New York, NY 10016, USA
| |
Collapse
|
16
|
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: 1.8] [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
|
17
|
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: 3.0] [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
|
18
|
Abstract
Vaccines are considered one of the most important advances in modern medicine and have greatly improved our quality of life by reducing or eliminating many serious infectious diseases. Successful vaccines have been developed against many of the most common human pathogens, and this success has not been dependent upon any one specific class of vaccine since subunit vaccines, non-replicating whole-virus or whole-bacteria vaccines, and attenuated live vaccines have all been effective for particular vaccine targets. After completing the initial immunization series, one common aspect of successful vaccines is that they induce long-term protective immunity. In contrast, several partially successful vaccines appear to induce protection that is relatively short-lived and it is likely that long-term protective immunity will be critical for making effective vaccines against our most challenging diseases such as AIDS and malaria.
Collapse
Affiliation(s)
- Ian J Amanna
- Najít Technologies, Inc, Beaverton, OR, 97006, USA
| | - Mark K Slifka
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA.
| |
Collapse
|
19
|
Yusupov A, Popovsky D, Mahmood L, Kim AS, Akman AE, Yuan H. The nonavalent vaccine: a review of high-risk HPVs and a plea to the CDC. AMERICAN JOURNAL OF STEM CELLS 2019; 8:52-64. [PMID: 31976155 PMCID: PMC6971474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 11/25/2019] [Indexed: 06/10/2023]
Abstract
Two of the leading strategies to prevent cervical cancer are prophylactic human papillomavirus (HPV) vaccination and routine Papanicolaou (Pap) testing. However, regardless of being vaccinated with first-generation (bivalent and quadrivalent) HPV vaccines at the recommended dosing schedule, many women are still found to have low- and high-grade cervical intraepithelial lesions. Studies have shown that this is largely due to: (1) first-generation vaccines only protecting against 70% of high-risk HPV types that cause cervical cancer (HPVs 16/18) and (2) vaccinated women being more prone to infection with non-protected high-risk HPV types than unvaccinated women. Fortunately, the FDA recently approved a nonavalent vaccine that protects against 5 additional high-risk HPV types that cause 20% of cervical cancers (HPVs 31/33/45/52/58), which is the only HPV vaccine currently available in the United States. Although the Advisory Committee on Immunization Practices (ACIP) recommends the nonavalent vaccine in men and women up to the age of 45 years, it does not recommend the nonavalent vaccine in those previously vaccinated with 3 doses of bivalent or quadrivalent vaccine, deeming them "adequately vaccinated". As this population is most at risk, this review serves to provide background and argue for a change in their recommendation.
Collapse
Affiliation(s)
- Ariel Yusupov
- Georgetown University School of MedicineWashington, DC, USA
| | | | - Lyaba Mahmood
- Georgetown University School of MedicineWashington, DC, USA
| | - Andrew S Kim
- Georgetown University School of MedicineWashington, DC, USA
| | - Alex E Akman
- Georgetown University School of MedicineWashington, DC, USA
| | - Hang Yuan
- Department of Pathology, Georgetown University Medical CenterWashington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical CenterWashington, DC, USA
| |
Collapse
|
20
|
Long-term HPV-specific immune response after one versus two and three doses of bivalent HPV vaccination in Dutch girls. Vaccine 2019; 37:7280-7288. [DOI: 10.1016/j.vaccine.2019.09.066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/04/2019] [Accepted: 09/19/2019] [Indexed: 02/06/2023]
|
21
|
Pattyn J, Van Keer S, Tjalma W, Matheeussen V, Van Damme P, Vorsters A. Infection and vaccine-induced HPV-specific antibodies in cervicovaginal secretions. A review of the literature. PAPILLOMAVIRUS RESEARCH 2019; 8:100185. [PMID: 31494291 PMCID: PMC6804463 DOI: 10.1016/j.pvr.2019.100185] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/19/2019] [Accepted: 09/04/2019] [Indexed: 02/02/2023]
Abstract
Background Human papillomavirus (HPV) infects and propagates in the cervical mucosal epithelium. Hence, in addition to assessing systemic immunity, the accurate measurement of cervical immunity is important to evaluate local immune responses to HPV infection and vaccination. This review discusses studies that investigated the presence of infection and vaccine-induced HPV-specific antibodies in cervicovaginal secretions (CVS). Methods We searched the two main health sciences databases, PubMed and the ISI Web of Science, from the earliest dates available to March 2019. From the eligible publications, information was extracted regarding: (i) study design, (ii) the reported HPV-specific antibody concentrations in CVS (and the associated serum levels, when provided), (iii) the CVS collection method, and (iv) the immunoassays used. Results The systematic search and selection process yielded 44 articles. The evidence of HPV-specific antibodies in CVS after natural infection (26/44) and HPV vaccination (18/44) is discussed. Many studies indicate that HPV-specific antibody detection in CVS is variable but feasible with a variety of collection methods and immunoassays. Most CVS samples were collected by cervicovaginal washing or wicks, and antibody presence was mostly determined by VLP-based ELISAs. The moderate to strong correlation between vaccine-induced antibody levels in serum and in CVS indicates that HPV vaccines generate antibodies that transudate through the cervical mucosal epithelium. Conclusion Although HPV-specific antibodies have lower titres in CVS than in serum samples, studies have shown that their detection in CVS is feasible. Nevertheless, the high variability of published observations and the lack of a strictly uniform, well-validated method for the collection, isolation and quantification of antibodies indicates a need for specific methods to improve and standardize the detection of HPV-specific antibodies in CVS.
Collapse
Affiliation(s)
- Jade Pattyn
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Belgium.
| | - Severien Van Keer
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Belgium
| | - Wiebren Tjalma
- Multidisciplinary Breast Clinic, Gynaecological Oncology Unit, Department of Obstetrics and Gynaecology, Antwerp University Hospital (UZA) (Belgium), Molecular Imaging, Pathology, Radiotherapy, and Oncology (MIPRO), Faculty of Medicine and Health Sciences, University of Antwerp, Belgium
| | - Veerle Matheeussen
- Department of Microbiology, Antwerp University Hospital (UZA) (Belgium); Department of Medical Microbiology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp (Belgium); Department of Medical Biochemistry, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Belgium
| | - Pierre Van Damme
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Belgium
| | - Alex Vorsters
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Belgium
| |
Collapse
|
22
|
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: 7.4] [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
|
23
|
Lacey CJ. HPV vaccination in HIV infection. ACTA ACUST UNITED AC 2019; 8:100174. [PMID: 31252073 PMCID: PMC6603434 DOI: 10.1016/j.pvr.2019.100174] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/23/2019] [Accepted: 06/24/2019] [Indexed: 02/07/2023]
Abstract
Persons with HIV are at increased risk of HPV infection, HPV disease, and HPV-related cancers compared to HIV negative persons. In persons with HIV, immune responses to vaccination are often sub-optimal, and while these improve with ART, they often remain lower and decline more rapidly than in HIV-negative individuals. Although the evidence base to support the immunogenicity of HPV vaccines in HIV + ve persons is reasonable, the evidence base to support the efficacy of HPV vaccines in HIV + ve individuals is inconsistent. There is one study in HIV + ve men who have sex with men (MSM) which showed no effect, and two other studies, one in HIV + ve women and one in HIV + ve adolescents that showed reduced effectiveness. All these effectiveness studies used Gardasil 4 (G4). Two studies in HIV + ve persons have shown superior immunogenicity of Cervarix (which uses a TLR4 agonist adjuvant) compared to G4. Studies of Hepatitis B vaccines in HIV + ve persons have shown that either (i) increased number of doses (ii) increased vaccine dose, or (iii) TLR agonist adjuvanted vaccines, all produce increased immunogenicity compared to standard vaccine regimes. Therefore, questions remain as to optimal HPV vaccine regimes in HIV and further clinical trials with different HPV vaccine regimes are needed.
Collapse
|
24
|
Schellenbacher C, Huber B, Skoll M, Shafti-Keramat S, Roden RBS, Kirnbauer R. Incorporation of RG1 epitope into HPV16L1-VLP does not compromise L1-specific immunity. Vaccine 2019; 37:3529-3534. [PMID: 31147274 DOI: 10.1016/j.vaccine.2019.05.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 04/28/2019] [Accepted: 05/05/2019] [Indexed: 12/13/2022]
Abstract
The candidate pan-Human Papillomavirus (HPV) vaccine RG1-VLP are HPV16 major capsid protein L1 virus-like-particles (VLP) comprising a type-common epitope of HPV16 minor capsid protein L2 (RG1; aa17-36). Vaccinations have previously demonstrated efficacy against genital high-risk (hr), low-risk (lr) and cutaneous HPV. To compare RG1-VLP to licensed vaccines, rabbits (n = 3) were immunized thrice with 1 µg, 5 µg, 25 µg, or 125 µg of RG1-VLP or a 1/4 dose of Cervarix®. 5 µg of RG1-VLP or 16L1-VLP (Cervarix) induced comparable HPV16 capsid-reactive and neutralizing antibodies titers (62,500/12,500-62,500 or 1000/10,000). 25 µg RG1-VLP induced robust cross-neutralization titers (50-1000) against hrHPV18/31/33/45/52/58/26/70. To mimic reduced immunization schedules in adolescents, mice (n = 10) were immunized twice with RG1-VLP (5 µg) plus 18L1-VLP (5 µg). HPV16 neutralization (titers of 10,000) similar to Cervarix and Gardasil and cross-protection against hrHPV58 vaginal challenge was observed. RG1-VLP vaccination induces hrHPV16 neutralization comparable to similar doses of licensed vaccines, plus cross-neutralization to heterologous hrHPV even when combined with HPV18L1-VLP.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- Capsid Proteins/genetics
- Capsid Proteins/immunology
- Epitopes/genetics
- Epitopes/immunology
- Immunization Schedule
- Oncogene Proteins, Viral/genetics
- Oncogene Proteins, Viral/immunology
- Papillomavirus Vaccines/administration & dosage
- Papillomavirus Vaccines/genetics
- Papillomavirus Vaccines/immunology
- Rabbits
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Treatment Outcome
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/immunology
Collapse
Affiliation(s)
- C Schellenbacher
- Department of Dermatology, Medical University Vienna (MUW), Austria.
| | - B Huber
- Department of Dermatology, Medical University Vienna (MUW), Austria
| | - M Skoll
- Department of Dermatology, Medical University Vienna (MUW), Austria
| | - S Shafti-Keramat
- Department of Dermatology, Medical University Vienna (MUW), Austria
| | - R B S Roden
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - R Kirnbauer
- Department of Dermatology, Medical University Vienna (MUW), Austria
| |
Collapse
|
25
|
Van Keer S, Willhauck-Fleckenstein M, Pattyn J, Butt J, Tjalma WAA, Van Ostade X, Hens N, Van Damme P, Waterboer T, Vorsters A. First-void urine as a non-invasive liquid biopsy source to detect vaccine-induced human papillomavirus antibodies originating from cervicovaginal secretions. J Clin Virol 2019; 117:11-18. [PMID: 31129514 DOI: 10.1016/j.jcv.2019.05.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/04/2019] [Accepted: 05/10/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Monitoring HPV antibodies non-invasively would be a major advantage for large epidemiological studies and follow-up of vaccinees. OBJECTIVES This study investigated the presence of HPV-specific antibody transudates from systemic circulation in first-void urine of (un)vaccinated subjects and the agreement with paired sera. STUDY DESIGN In this case-control study, 55 paired first-void urine and serum samples were included from 19- to 26-year-old women, unvaccinated (n = 19) or vaccinated (n = 36) with the bi- or quadrivalent HPV vaccine during adolescence (NCT02714114). Human IgA, total human IgG, and HPV6/11/16/18-Ig(M/G/A) were measured in paired samples. RESULTS Significant positive Spearman rank correlations (rs) were found in HPV-specific antibody levels between paired samples (HPV6: rs = 0.777; HPV11: rs = 0.757; HPV16: rs = 0.876; HPV18: rs = 0.636 (p < 0.001)). In both first-void urine and serum, significantly higher HPV6/11/16/18 antibody levels were observed in vaccinated compared with unvaccinated women (p ≤ 0.017). CONCLUSIONS The present study provides the first proof that vaccine-induced HPV antibodies are detectable in the first-void urine of young women. Moreover, significant positive correlations were observed between HPV6/11/16/18-antibodies in first-void urine and paired sera. Further optimization and validation are required to demonstrate its potential use in epidemiological studies and follow-up of HPV vaccination.
Collapse
Affiliation(s)
- Severien Van Keer
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium.
| | - Martina Willhauck-Fleckenstein
- Infections and Cancer Epidemiology Group, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Jade Pattyn
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Julia Butt
- Infections and Cancer Epidemiology Group, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Wiebren A A Tjalma
- Multidisciplinary Breast Clinic, Unit Gynaecologic Oncology, Department of Obstetrics and Gynaecology, Antwerp University Hospital (UZA), Wilrijkstraat 10, 2650, Edegem, Belgium; Molecular Imaging, Pathology, Radiotherapy, Oncology (MIPRO), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Xaveer Van Ostade
- Laboratory of Protein Science, Proteomics & Epigenetic Signalling (PPES), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Niel Hens
- CHERMID, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium; Centre for Statistics, I-Biostat, Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium
| | - Pierre Van Damme
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Tim Waterboer
- Infections and Cancer Epidemiology Group, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Alex Vorsters
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| |
Collapse
|
26
|
Slifka MK, Amanna IJ. Role of Multivalency and Antigenic Threshold in Generating Protective Antibody Responses. Front Immunol 2019; 10:956. [PMID: 31118935 PMCID: PMC6504826 DOI: 10.3389/fimmu.2019.00956] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 04/15/2019] [Indexed: 12/03/2022] Open
Abstract
Vaccines play a vital role in protecting our communities against infectious disease. Unfortunately, some vaccines provide only partial protection or in some cases vaccine-mediated immunity may wane rapidly, resulting in either increased susceptibility to that disease or a requirement for more booster vaccinations in order to maintain immunity above a protective level. The durability of antibody responses after infection or vaccination appears to be intrinsically determined by the structural biology of the antigen, with multivalent protein antigens often providing more long-lived immunity than monovalent antigens. This forms the basis for the Imprinted Lifespan model describing the differential survival of long-lived plasma cell populations. There are, however, exceptions to this rule with examples of highly attenuated live virus vaccines that are rapidly cleared and elicit only short-lived immunity despite the expression of multivalent surface epitopes. These exceptions have led to the concept that multivalency alone may not reliably determine the duration of protective humoral immune responses unless a minimum number of long-lived plasma cells are generated by reaching an appropriate antigenic threshold of B cell stimulation. Examples of long-term and in some cases, potentially lifelong antibody responses following immunization against human papilloma virus (HPV), Japanese encephalitis virus (JEV), Hepatitis B virus (HBV), and Hepatitis A virus (HAV) provide several lessons in understanding durable serological memory in human subjects. Moreover, studies involving influenza vaccination provide the unique opportunity to compare the durability of hemagglutinin (HA)-specific antibody titers mounted in response to antigenically repetitive whole virus (i.e., multivalent HA), or detergent-disrupted “split” virus, in comparison to the long-term immune responses induced by natural influenza infection. Here, we discuss the underlying mechanisms that may be associated with the induction of protective immunity by long-lived plasma cells and their importance in future vaccine design.
Collapse
Affiliation(s)
- Mark K Slifka
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Ian J Amanna
- Najít Technologies, Inc., Beaverton, OR, United States
| |
Collapse
|
27
|
Effect of Multiple Vaccinations with Tumor Cell-Based Vaccine with Codon-Modified GM-CSF on Tumor Growth in a Mouse Model. Cancers (Basel) 2019; 11:cancers11030368. [PMID: 30875953 PMCID: PMC6468346 DOI: 10.3390/cancers11030368] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/07/2019] [Accepted: 03/11/2019] [Indexed: 12/11/2022] Open
Abstract
Ectopic expression of codon-modified granulocyte-macrophage colony-stimulating factor (cGM-CSF) in TC-1 cells (TC-1/cGM-CSF), a model cell line for human papillomavirus (HPV)-infected cervical cancer cells, increased the expression level of GM-CSF and improved the efficacy of tumor cell-based vaccines in a cervical cancer mouse model. The number of vaccine doses required to induce a long-term immune response in a cervical cancer mouse model is poorly understood. Here, we investigated one, three, and five doses of the irradiated TC-1/cGM-CSF vaccine to determine which dose was effective in inducing a greater immune response and the suppression of tumors. Our findings showed that three doses of irradiated TC-1/cGM-CSF vaccine elicited slower tumor growth rates and enhanced survival rates compared with one dose or five doses of irradiated TC-1/cGM-CSF vaccine. Consistently, mice vaccinated with three doses of irradiated TC-1/cGM-CSF vaccine exhibited stronger interferon gamma (IFN-γ) production in HPV E7-specific CD8⁺ T cells and CD4⁺ T cells. A higher percentage of natural killer cells and interferon-producing killer dendritic cells (IKDCs) appeared in the splenocytes of the mice vaccinated with three doses of irradiated TC-1/cGM-CSF vaccine compared with those of the mice vaccinated with one dose or five doses of irradiated TC-1/cGM-CSF vaccine. Our findings demonstrate that single or multiple vaccinations, such as five doses, with irradiated TC-1/cGM-CSF vaccine suppressed the immune response, whereas three doses of irradiated TC-1/cGM-CSF vaccine elicited a greater immune response and subsequent tumor suppression.
Collapse
|
28
|
van der Weele P, Breeuwsma M, Donken R, van Logchem E, van Marm-Wattimena N, de Melker H, Meijer CJLM, King AJ. Effect of the bivalent HPV vaccine on viral load of vaccine and non-vaccine HPV types in incident clearing and persistent infections in young Dutch females. PLoS One 2019; 14:e0212927. [PMID: 30830913 PMCID: PMC6398842 DOI: 10.1371/journal.pone.0212927] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 02/12/2019] [Indexed: 12/19/2022] Open
Abstract
Background HPV vaccination with the bivalent vaccine is efficacious against HPV16 and 18 infections and cross-protection against non-vaccine HPV types has been demonstrated. Here, we assessed (cross-) protective effects of the bivalent HPV16/18 vaccine on incident and persistent infections and viral load (VL) of fifteen HPV types in an observational cohort study monitoring HPV vaccine effects. Methods Vaginal samples were obtained annually. Type-specific VL assays were developed for HPV6,11,31 33,35,39,45,51,52,56,58,59 and 66 and used in addition to existing HPV16 and 18 assays. Rate differences of incident clearing and persistent infections were correlated with differences in VL and vaccination status. Results HPV16/18 vaccination resulted in significantly lower incidence of HPV16/18 infections and significantly lower VL in breakthrough HPV16 (p<0.01) and 18 infections (p<0.01). The effects of vaccination on non-vaccine type VL were ambiguous. Incidence and/or persistence rates of HPV31, 33, 35 and 45 were reduced in the vaccinated group. However, no significant type specific VL effects were found against HPV31, 33, 45, 52 in the vaccinated group. For HPV 6, 59 and 66 no significant reductions in numbers of incident and persistent infections were found, however borderline) VL reductions following vaccination were observed for HPV6 (p = 0.01), 59 (p = 0.10) and 66 (p = 0.03), suggesting a minor effect of the vaccine on the VL level of these HPV types. Overall, vaccination resulted in infections with slightly lower VL, irrespective of HPV type. Conclusions In conclusion, vaccination with the bivalent HPV16/18 vaccine results in significantly reduced numbers of HPV16 and 18 incidence rates and reduced VL in breakthrough infections. Significant reductions in incident and/or persistent HPV31, 33, 35 and 45 infections were found, but no significant effect was observed on the VL for infections with these types. For the other non-vaccine HPV types no reduction in incident and/or persistent infections were found, but overall the VL tended to be somewhat lower in vaccinated women.
Collapse
Affiliation(s)
- Pascal van der Weele
- National Institute for Public Health and the Environment (RIVM), Centre for Infectious Disease Control, Bilthoven, the Netherlands.,Vrije Universiteit - University Medical Center (VUmc), Department of Pathology, Amsterdam, the Netherlands
| | - Martijn Breeuwsma
- National Institute for Public Health and the Environment (RIVM), Centre for Infectious Disease Control, Bilthoven, the Netherlands
| | - Robine Donken
- National Institute for Public Health and the Environment (RIVM), Centre for Infectious Disease Control, Bilthoven, the Netherlands.,Vrije Universiteit - University Medical Center (VUmc), Department of Pathology, Amsterdam, the Netherlands
| | - Elske van Logchem
- National Institute for Public Health and the Environment (RIVM), Centre for Infectious Disease Control, Bilthoven, the Netherlands
| | - Naomi van Marm-Wattimena
- National Institute for Public Health and the Environment (RIVM), Centre for Infectious Disease Control, Bilthoven, the Netherlands
| | - Hester de Melker
- National Institute for Public Health and the Environment (RIVM), Centre for Infectious Disease Control, Bilthoven, the Netherlands
| | - Chris J L M Meijer
- Vrije Universiteit - University Medical Center (VUmc), Department of Pathology, Amsterdam, the Netherlands
| | - Audrey J King
- National Institute for Public Health and the Environment (RIVM), Centre for Infectious Disease Control, Bilthoven, the Netherlands
| |
Collapse
|
29
|
Racial differences in human papilloma virus types amongst United States women with HIV and cervical precancer. AIDS 2018; 32:2821-2826. [PMID: 30234608 DOI: 10.1097/qad.0000000000002005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Recent studies reported a lower human papillomavirus 16 (HPV16) prevalence in cervical precancer among African American than Caucasian women in the general population. We assessed this relationship in women with HIV. DESIGN Women living with or at risk for HIV in the Women's Interagency HIV Study were followed semi-annually with Pap tests, colposcopy/histology (if indicated), and collection of cervicovaginal lavage samples for HPV testing by PCR. Racial and ethnic groups were defined using genomic Ancestry Informative Markers (AIMs). RESULTS Among 175 cases of cervical intraepithelial neoplasia 3 or worse (CIN-3+), 154 were diagnosed in women with HIV. African American (27%) and Hispanic (37%) cases were significantly less likely than Caucasian (62%) women to test positive for HPV16 (P = 0.01). In multivariate logistic regression models, these associations remained significant for African Americans (odds ratio = 0.13; 95% confidence interval (CI) 0.04-0.44; P = 0.001) but not Hispanics, after controlling for HIV status, CD4 count, history of AIDS, age, smoking, and sexual behavior. Limiting the analysis to women with HIV did not change the findings. CONCLUSION HPV16 prevalence is lower in African American compared with Caucasian women with HIV and cervical precancer, independent of immune status. Future studies to determine why these racial differences exist are warranted, and whether there are similar associations between race and invasive cervical cancer in women with HIV. Further, HPV types not covered by quadrivalent and bivalent vaccines may play an especially important role in cervical precancer among HIV-positive African American women, a possible advantage to using nonavalent HPV vaccine in this population.
Collapse
|
30
|
Phillips A, Patel C, Pillsbury A, Brotherton J, Macartney K. Safety of Human Papillomavirus Vaccines: An Updated Review. Drug Saf 2018; 41:329-346. [PMID: 29280070 DOI: 10.1007/s40264-017-0625-z] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Human papillomavirus (HPV) vaccines are now included in immunisation programmes in 71 countries. Unfortunately, uptake has been impacted in some countries by reduced confidence in the safety of the HPV vaccine. In 2013, we published an extensive review demonstrating a reassuring safety profile for bivalent (2vHPV) and quadrivalent (4vHPV) vaccines. A nonavalent (9vHPV) vaccine is now available and HPV immunisation programmes have been extended to males in 11 countries. The aim of this updated narrative review was to examine the evidence on HPV vaccine safety, focusing on the 9vHPV vaccine, special populations and adverse events of special interest (AESI). The previous searches were replicated to identify studies to August 2016, including additional search terms for AESI. We identified 109 studies, including 15 population-based studies in over 2.5 million vaccinated individuals across six countries. All vaccines demonstrated an acceptable safety profile; injection-site reactions were slightly more common for 9vHPV vaccine than for 4vHPV vaccine. There was no consistent evidence of an increased risk of any AESI, including demyelinating syndromes or neurological conditions such as complex regional pain or postural orthostatic tachycardia syndromes. The risk-benefit profile for HPV vaccines remains highly favourable.
Collapse
Affiliation(s)
| | - Cyra Patel
- National Centre for Immunisation Research and Surveillance, Kids Research Institute, The Sydney Children's Hospitals Network, Cnr Hawkesbury Road and Hainsworth Street, Westmead, NSW, 2145, Australia
| | - Alexis Pillsbury
- National Centre for Immunisation Research and Surveillance, Kids Research Institute, The Sydney Children's Hospitals Network, Cnr Hawkesbury Road and Hainsworth Street, Westmead, NSW, 2145, Australia
| | - Julia Brotherton
- National HPV Vaccination Program Register, Victorian Cytology Service, Level 6, 176 Wellington Parade, East Melbourne, Carlton, VIC, 3002, Australia
- The University of Melbourne, Melbourne, VIC, Australia
| | - Kristine Macartney
- The University of Sydney, Sydney, NSW, Australia.
- National Centre for Immunisation Research and Surveillance, Kids Research Institute, The Sydney Children's Hospitals Network, Cnr Hawkesbury Road and Hainsworth Street, Westmead, NSW, 2145, Australia.
| |
Collapse
|
31
|
Pinto LA, Dillner J, Beddows S, Unger ER. Immunogenicity of HPV prophylactic vaccines: Serology assays and their use in HPV vaccine evaluation and development. Vaccine 2018; 36:4792-4799. [PMID: 29361344 PMCID: PMC6050153 DOI: 10.1016/j.vaccine.2017.11.089] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 11/17/2017] [Indexed: 11/23/2022]
Abstract
When administered as standard three-dose schedules, the licensed HPV prophylactic vaccines have demonstrated extraordinary immunogenicity and efficacy. We summarize the immunogenicity of these licensed vaccines and the most commonly used serology assays, with a focus on key considerations for one-dose vaccine schedules. Although immune correlates of protection against infection are not entirely clear, both preclinical and clinical evidence point to neutralizing antibodies as the principal mechanism of protection. Thus, immunogenicity assessments in vaccine trials have focused on measurements of antibody responses to the vaccine. Non-inferiority of antibody responses after two doses of HPV vaccines separated by 6 months has been demonstrated and this evidence supported the recent WHO recommendations for two-dose vaccination schedules in both boys and girls 9-14 years of age. There is also some evidence suggesting that one dose of HPV vaccines may provide protection similar to the currently recommended two-dose regimens but robust data on efficacy and immunogenicity of one-dose vaccine schedules are lacking. In addition, immunogenicity has been assessed and reported using different methods, precluding direct comparison of results between different studies and vaccines. New head-to-head vaccine trials evaluating one-dose immunogenicity and efficacy have been initiated and an increase in the number of trials relying on immunobridging is anticipated. Therefore, standardized measurement and reporting of immunogenicity for the up to nine HPV types targeted by the current vaccines is now critical. Building on previous HPV serology assay standardization and harmonization efforts initiated by the WHO HPV LabNet in 2006, new secondary standards, critical reference reagents and testing guidelines will be generated as part of a new partnership to facilitate harmonization of the immunogenicity testing in new HPV vaccine trials.
Collapse
MESH Headings
- Adolescent
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Child
- Clinical Trials as Topic
- Female
- Human Papillomavirus Recombinant Vaccine Quadrivalent, Types 6, 11, 16, 18/administration & dosage
- Human Papillomavirus Recombinant Vaccine Quadrivalent, Types 6, 11, 16, 18/immunology
- Humans
- Immunization Schedule
- Immunogenicity, Vaccine
- Male
- Mass Vaccination/standards
- Neutralization Tests/standards
- Papillomavirus Infections/prevention & control
- Papillomavirus Vaccines/administration & dosage
- Papillomavirus Vaccines/immunology
- Treatment Outcome
- Uterine Cervical Neoplasms/prevention & control
- World Health Organization
Collapse
Affiliation(s)
- Ligia A Pinto
- Vaccine, Cancer and Immunity Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, USA.
| | - Joakim Dillner
- Department of Laboratory Medicine, Karolinska Institutet, 141 86 Stockholm, Sweden.
| | - Simon Beddows
- Virus Reference Department, Public Health England, London, UK.
| | - Elizabeth R Unger
- Chronic Viral Diseases Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| |
Collapse
|
32
|
Toh ZQ, Cheow KWB, Russell FM, Hoe E, Reyburn R, Fong J, Tuivaga E, Ratu FT, Nguyen CD, Matanitobua S, Reitsma A, Tabrizi SN, Garland SM, Mulholland EK, Licciardi PV. Cellular Immune Responses 6 Years Following 1, 2, or 3 Doses of Quadrivalent HPV Vaccine in Fijian Girls and Subsequent Responses to a Dose of Bivalent HPV Vaccine. Open Forum Infect Dis 2018; 5:ofy147. [PMID: 30019002 PMCID: PMC6041981 DOI: 10.1093/ofid/ofy147] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 06/18/2018] [Indexed: 12/19/2022] Open
Abstract
Background This study examined the cellular immunity of 0, 1, 2, and 3 doses of Gardasil vaccine (4vHPV) in girls after 6 years and their responses to a subsequent dose of Cervarix vaccine (2vHPV). Methods A subset of girls (n = 59) who previously received 0, 1, 2, or 3 doses of 4vHPV 6 years earlier were randomly selected from a cohort study of Fijian girls (age 15-19 years). Blood was collected before and 28 days after a dose of 2vHPV. The HPV16- and HPV18-specific cellular immune response was determined by IFNγ-ELISPOT and by measurement of cytokines in peripheral blood mononuclear cell supernatants. Results Six years after 4vHPV vaccination, HPV18-specific responses were significantly lower in the 1- (1D) or 2-dose (2D) recipients compared with 3-dose recipients (2D: IFNγ-ELISPOT: P = .008; cytokines, IFNγ: P = .002; IL-2: P = .022; TNFα: P = .016; IL-10: P = .018; 1D: IL-2: P = .031; IL-10: P = .014). These differences were no longer significant post-2vHPV. No significant differences in HPV16 responses (except IL-2, P < .05) were observed between the 2- or 1-dose recipients and 3-dose recipients. Conclusions These data suggest that cellular immunity following reduced-dose schedules was detectable after 6 years, although the responses were variable between HPV types and dosage groups. The clinical significance of this is unknown. Further studies on the impact of reduced dose schedules are needed, particularly in high-disease burden settings.
Collapse
Affiliation(s)
- Zheng Quan Toh
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | | | - Fiona M Russell
- Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Centre for International Child Health, Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Edwin Hoe
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Rita Reyburn
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - James Fong
- Ministry of Health and Medical Services, Suva, Fiji
| | | | | | - Cattram D Nguyen
- Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Centre for International Child Health, Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | | | - Andrea Reitsma
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Sepehr N Tabrizi
- Department of Obstetrics and Gynecology, The University of Melbourne, The Royal Women's Hospital and Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Regional HPV Labnet Reference Laboratory, Department of Microbiology and Infectious Disease, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Suzanne M Garland
- Department of Obstetrics and Gynecology, The University of Melbourne, The Royal Women's Hospital and Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Regional HPV Labnet Reference Laboratory, Department of Microbiology and Infectious Disease, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Edward K Mulholland
- Murdoch Children's Research Institute, Parkville, Victoria, Australia.,London School of Hygiene and Tropical Medicine, University of London, London, United Kingdom.,Department of Child Health, Menzies School of Health Research, Darwin, Northern Territory, Australia
| | - Paul V Licciardi
- Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Centre for International Child Health, Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
33
|
Chen X, Zhang T, Liu H, Hao Y, Liao G, Xu X. Displaying 31RG-1 peptide on the surface of HPV16 L1 by use of a human papillomavirus chimeric virus-like particle induces cross-neutralizing antibody responses in mice. Hum Vaccin Immunother 2018; 14:2025-2033. [PMID: 29683766 DOI: 10.1080/21645515.2018.1464355] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Current available human papillomavirus (HPV) vaccines are based on the major capsid protein L1 virus-like particles (VLPs), which mainly induce type-specific neutralizing antibodies against vaccine types. Continuing to add more types of VLPs in a vaccine raises the complexity and cost of production which remains the principal impediment to achieve broad implementation of HPV vaccines, particularly in developing regions. In this study, we constructed 16L1-31L2 chimeric VLP (cVLP) by displaying HPV31 L2 aa.17-38 on the h4 coil surface region of HPV16 L1, and assessed its immunogenicity in mouse model. We found that the cVLP adjuvanted with alum plus monophosphoryl lipid A could induce cross-neutralizing antibody responses against 16 out of 17 tested HPV pseudoviruses, and the titer against HPV16 was as high as that was induced by HPV16 L1VLP (titer > 105), more importantly, titers over 103 were observed against two HR-HPVs including HPV31 (titer, 2,200) and -59 (titer, 1,013), among which HPV59 was not covered by Gardasil-9, and medium or low titers of cross-neutralizing antibodies against other 13 tested HPV pseudoviruses were also observed. Our data demonstrate that 16L1-31L2 cVLP is a promising candidate for the formulation of broader spectrum HPV vaccines.
Collapse
Affiliation(s)
- Xue Chen
- a Department of Biophysics and Structural Biology , Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College , Beijing , China
| | - Ting Zhang
- a Department of Biophysics and Structural Biology , Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College , Beijing , China
| | - Hongyang Liu
- a Department of Biophysics and Structural Biology , Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College , Beijing , China
| | - Yaru Hao
- a Department of Biophysics and Structural Biology , Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College , Beijing , China
| | - Guoyang Liao
- b The Fifth Department of Biological Products , Institute of Medical Biology, Chinese Academy of Medical Science, Peking Union Medical College , Yunnan , China
| | - Xuemei Xu
- a Department of Biophysics and Structural Biology , Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College , Beijing , China
| |
Collapse
|
34
|
Van Kriekinge G, Sohn WY, Aljunid SM, Soon R, Yong CM, Chen J, Lee IH. Comparative Cost-Effectiveness Analysis of Two Different Two-Dose Human Papillomavirus Vaccines in Malaysia. Asian Pac J Cancer Prev 2018; 19:933-940. [PMID: 29693347 PMCID: PMC6031794 DOI: 10.22034/apjcp.2018.19.4.933] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Purpose: To comparatively evaluate the results of a 2-dose human papillomavirus (HPV) vaccination programme with the AS04-adjuvanted HPV16/18 vaccine (AS04-HPV-16/18v) or HPV-6/11/16/18 vaccine (4vHPVv), in addition to cervical cancer (CC) screening, in Malaysia. Methods: A lifetime Markov model replicating the natural history of HPV in 13-year-old girls was adapted to Malaysia to assess the impact of vaccination on pre-cancerous lesions, genital warts and CC cases, CC deaths, quality-adjusted life years (QALYs), and costs from the perspective of the Malaysian Ministry of Health. Vaccine effectiveness was based on efficacy and HPV type distribution. Both vaccines were assumed to have equal efficacy against vaccine-type HPV but differed for protection against non-vaccine types. Vaccine price parity was used and health and cost outcomes were discounted at 3%/annum. Sensitivity analyses tested the robustness of the results. Results: The model predicted that AS04-HPV-16/18v would result in 361 fewer CC cases and 115 fewer CC deaths than 4vHPVv, whereas 4vHPVv averted 4,241 cases of genital warts over the cohort’s lifetime. Discounted total costs showed savings of 18.50 million Malaysian Ringgits and 246 QALYs in favour of AS04-HPV-16/18v. In one-way sensitivity analyses, the discount rate was the most influential variable for costs and QALYs, but AS04-HPV-16/18v remained dominant throughout. A two-way sensitivity analysis to assess the longevity of cross-protection for both vaccines confirmed the base-case. Conclusions: In Malaysia, the use of AS04-HPV-16/18v, in addition to screening, was modelled to be dominant over 4vHPVv, with greater estimated CC benefits and lower costs.
Collapse
|
35
|
Weinberg A, Huang S, Moscicki AB, Saah A, Levin MJ. Persistence of memory B-cell and T-cell responses to the quadrivalent HPV vaccine in HIV-infected children. AIDS 2018; 32:851-860. [PMID: 29424778 PMCID: PMC5869173 DOI: 10.1097/qad.0000000000001773] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To determine the magnitude and persistence of quadrivalent human papillomavirus (HPV)16 and HPV18 B-cell and T-cell memory after three or four doses of quadrivalent HPV vaccine (QHPV) in HIV-infected children. METHODS Seventy-four HIV-infected children immunized with four doses and 23 with three doses of QHPV had HPV16 and HPV18 IgG B-cell and IFNγ and IL2 T-cell ELISPOT performed at 2, 3.5 and 4-5 years after the last dose. RESULTS HPV16 and HPV18 T-cell responses were similar in both treatment groups, with higher responses to HPV16 vs. HPV18. These HPV T-cell responses correlated with HIV disease characteristics at the study visits. Global T-cell function declined over time as measured by nonspecific mitogenic stimulation. B-cell memory was similar across treatment groups and HPV genotypes. There was a decline in HPV-specific B-cell memory over time that reached statistical significance for HPV16 in the four-dose group. CONCLUSION B-cell and T-cell memory did not significantly differ after either three or four doses of QHPV in HIV-infected children. The clinical consequences of decreasing global T-cell function and HPV B-cell memory over time in HIV-infected children requires further investigation.
Collapse
Affiliation(s)
- Adriana Weinberg
- Section of Pediatric Infectious Diseases, Departments of Pediatrics and Medicine, and Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sharon Huang
- Center for Biostatistics in AIDS Research, Harvard School of Public Health, Boston, Massachusetts
| | - Anna-Barbara Moscicki
- Department of Pediatrics, University of California Los Angeles, Los Angeles, California
| | | | - Myron J Levin
- Section of Pediatric Infectious Diseases, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
36
|
Safety and Immunogenicity of the HPV-16/18 AS04-adjuvanted Vaccine in 4-6-year-old Girls: Results to Month 12 From a Randomized Trial. Pediatr Infect Dis J 2018; 37:e93-e102. [PMID: 29424799 DOI: 10.1097/inf.0000000000001871] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The burden of cervical cancer caused by human papillomavirus (HPV) is high in Latin America. The suboptimal HPV vaccination coverage in adolescents could be improved by pediatric immunization. HPV vaccination has not yet been reported in girls <9 years of age. METHODS This ongoing phase III, controlled, randomized, single-blind, multicenter study conducted in Colombia, Mexico and Panama (NCT01627561) evaluated the safety and immunogenicity of AS04-HPV-16/18 vaccine in 4-6-year-old girls. Healthy girls (randomized 1:1) received either 2 doses of AS04-HPV-16/18 vaccine (HPV group, N=74) or 1 dose of each measles-mumps-rubella and diphtheria-tetanus-acellular-pertussis vaccines (control group, N=74) 6 months apart. We report the safety and serum anti-HPV-16 and anti-HPV-18 antibodies (measured by enzyme-linked immunosorbent assay) up to 6 months postvaccination, that is, month (M) 12. RESULTS Injection site pain was the most frequently reported solicited local symptom in HPV vaccinees. The incidence of other solicited and unsolicited symptoms after each vaccination was similar between the HPV and control group. Until M12, 1 girl in the HPV group and 2 in the control group reported serious adverse events; all serious adverse events were assessed as unrelated to study vaccines. No potential immune-mediated diseases were identified. All girls seroconverted for both antigens after 2 doses of AS04-HPV-16/18. In initially seronegative girls, anti-HPV-16 geometric mean concentrations were 20080.0 enzyme-linked immunosorbent assay units (EU)/mL at M7 and 3246.5 EU/mL at M12; anti-HPV-18 geometric mean concentrations were 10621.8 EU/mL at M7 and 1216.6 EU/mL at M12. CONCLUSIONS Two-dose vaccination with AS04-HPV-16/18 was well tolerated and induced adequate antibody responses in 4-6-year-old girls.
Collapse
|
37
|
Zurek Munk-Madsen M, Toft L, Kube T, Richter R, Ostergaard L, Søgaard OS, Tolstrup M, Kaufmann AM. Cellular immunogenicity of human papillomavirus vaccines Cervarix and Gardasil in adults with HIV infection. Hum Vaccin Immunother 2017; 14:909-916. [PMID: 29172992 PMCID: PMC5893199 DOI: 10.1080/21645515.2017.1407896] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Human papillomavirus (HPV) infection is a frequent cause of malignant and non-malignant disease, in particular among persons with HIV. HPV serotype-specific anti L1 antibodies protect against HPV infection but little is known about prophylactic HPV vaccine-induced cell-mediated immunity against HPV in high-risk individuals. We recently showed that both HPV vaccines (Gardasil® and Cervarix®) induce solid, serological immune responses in HIV-infected persons. This study aimed to characterize HPV-specific CD4 T cells in HIV-infected HPV-vaccine recipients, T cell responses being critical for B cell activation and antibody-isotype switching. Thirty HIV-infected patients on long-term antiretroviral treatment (ART) received 3 doses of either Cervarix (n = 15) or Gardasil (n = 15) vaccine at month 0, 1.5 and 6. Cryopreserved peripheral blood mononuclear cells (PBMC) from baseline, 7 and 12 months were subjected to 24-hour stimulation with specific pools of HPV L1-peptides (HPV6, 11, 16, 18, 31 and 45) and HPV E6/E7-peptide pools (HPV6/11 and HPV16/18). Fluorescence-activated cell sorting with intracellular staining (IC-FACS) against CD4, CD154, IL-2, and IFNγ was performed. Frequencies (%) of HPV-antigen specific CD4+ T cells (CD154+/IL-2+ or CD154+/ IFNγ+) were determined. Both HPV-vaccines significantly and comparably enhanced cell-mediated vaccine L1 antigen-specific immunity in HIV-positive adults receiving ART therapy at month 7 and 12 after first vaccine dose. This suggests that the vaccines induce CD4 T cellular memory despite HIV-induced immune compromisation.
Collapse
Affiliation(s)
- Maria Zurek Munk-Madsen
- a Clinic for Gynecology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health , Berlin , Germany.,b Department of Infectious Diseases , Aarhus University Hospital , Aarhus , Denmark
| | - Lars Toft
- b Department of Infectious Diseases , Aarhus University Hospital , Aarhus , Denmark
| | - Tina Kube
- a Clinic for Gynecology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health , Berlin , Germany
| | - Rolf Richter
- a Clinic for Gynecology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health , Berlin , Germany
| | - Lars Ostergaard
- b Department of Infectious Diseases , Aarhus University Hospital , Aarhus , Denmark
| | - Ole S Søgaard
- b Department of Infectious Diseases , Aarhus University Hospital , Aarhus , Denmark
| | - Martin Tolstrup
- b Department of Infectious Diseases , Aarhus University Hospital , Aarhus , Denmark
| | - Andreas M Kaufmann
- a Clinic for Gynecology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health , Berlin , Germany
| |
Collapse
|
38
|
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 2017; 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] [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
|
39
|
Mitchell TC, Casella CR. No pain no gain? Adjuvant effects of alum and monophosphoryl lipid A in pertussis and HPV vaccines. Curr Opin Immunol 2017; 47:17-25. [PMID: 28728074 DOI: 10.1016/j.coi.2017.06.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 06/28/2017] [Indexed: 10/19/2022]
Abstract
Development of non-infectious subunit vaccines is hampered by a slow pipeline of new adjuvants to replace or enhance alum in part because expectations of safety are high. Transient vaccine side effects are not clinical priorities because they cause no lasting harm and vaccine development has appropriately been focused on avoidance of serious adverse events. As a result, surprisingly little is known about the extent to which side effects caused by a vaccine's reactogencicity are predictive of successful immunization outcomes. Recent clinical studies of pertussis and human papillomavirus vaccines adjuvanted with alum or the TLR4 agonist monophosphoryl lipid A can be used to advance understanding of the relationship between vaccine side effects and immunization outcomes.
Collapse
Affiliation(s)
- Thomas C Mitchell
- The University of Louisville School of Medicine, Department of Microbiology and Immunology, Institute for Cellular Therapeutics, 570 S. Preston St., Donald Baxter Bldg., Louisville, KY 40202, USA.
| | - Carolyn R Casella
- The University of Louisville School of Medicine, Department of Microbiology and Immunology, Institute for Cellular Therapeutics, 570 S. Preston St., Donald Baxter Bldg., Louisville, KY 40202, USA
| |
Collapse
|
40
|
Harper DM, DeMars LR. HPV vaccines - A review of the first decade. Gynecol Oncol 2017; 146:196-204. [PMID: 28442134 DOI: 10.1016/j.ygyno.2017.04.004] [Citation(s) in RCA: 268] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 04/03/2017] [Accepted: 04/06/2017] [Indexed: 02/01/2023]
Abstract
Pre-adolescent girls (9-15years) have the option of receiving a two dose HPV vaccine series at either a six month or one year interval to provide protection from HPV 16, the most prevalent type associated with cervical cancers, as well as several other less prevalent types. This series of vaccinations is highly likely to protect her from HPV infection until she enters the routine screening program, whether that be primary HPV testing or a combination of HPV testing and cytology. The two dose program has been recommended by the World Health Organization (WHO) since 2015. For women 15years and older, the three dose vaccine schedule is still recommended. The past ten years of Gardasil use has provided evidence of reduced HPV 16/18 infections in countries where there has been high coverage. Gardasil9 has replaced Gardasil. Gardasil9 has the same rapid anti-HPV 18 and HPV45 titer loss as Gardasil did. Cervarix remains equivalent to Gardasil9 in the prevention of HPV infections and precancers of any HPV type; Cervarix also has demonstrated sustained high antibody titers for at least 10years. One dose of Cervarix provides protection against HPV 16/18 infection with robust antibody titers well above natural infection titers. This may offer the easiest and most cost effective vaccination program over time, especially in low and lower middle income countries. Cervical cancer screening must continue to control cancer incidence over the upcoming decades. Future studies of prophylactic HPV vaccines, as defined by the WHO, must demonstrate protection against six month type specific persistent infections, not actual cervical cancer precursor disease endpoints, such as cervical intraepithelial neoplasia grade 3 (CIN 3) or adenocarcinoma in situ (AIS). This simplifies and makes less expensive future comparative studies between existing and new generic vaccines.
Collapse
Affiliation(s)
- Diane M Harper
- School of Medicine, Departments of Family and Geriatric Medicine and Obstetrics and Gynecology, Speed School of Engineering, School of Public Health, Epidemiology and Population Health, Health Promotion and Behavioral Sciences, University of Louisville, Louisville, KY, United States.
| | - Leslie R DeMars
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| |
Collapse
|
41
|
Endo F, Tabata T, Sadato D, Kawamura M, Ando N, Oboki K, Ukaji M, Kobayashi K, Kobayashi Y, Ikeda T, Shibasaki F. Development of a simple and quick immunochromatography method for detection of anti-HPV-16/-18 antibodies. PLoS One 2017; 12:e0171314. [PMID: 28158224 PMCID: PMC5291722 DOI: 10.1371/journal.pone.0171314] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 01/19/2017] [Indexed: 12/20/2022] Open
Abstract
Immunochromatography (IC) is widely used to detect target molecules in biological fluids. Since this method can be performed without a special technique or device, IC is a convenient way to assess the existence of antibodies or pathogens such as viruses and bacteria, simply and quickly. In this study, we established an IC method to detect serum antibodies against oncogenic human papillomavirus (HPV)-16 and HPV-18 L1 proteins using recombinant L1 proteins produced by silkworms as antigens. Infection of oncogenic HPVs is a major risk factor of cervical cancer, which is one of the most common cancers in women worldwide. We first measured blood sera of two groups by magnetic beads enzyme-linked immunosorbent assay (MB-ELISA). For the first group, sera were collected prospectively from young women who planned to receive HPV vaccination. The second group consisted of children under 20 years of age, non-vaccinated healthy women, vaccinated healthy women, dysplasia, cervical intraepithelial neoplasia III, and cervical cancer patients. We confirmed that standard vaccination doses significantly increased serum HPV antibody concentrations, and the level was sustained at least more than 30 months after vaccination. In contrast, an increase in antibody concentration was not observed in patients with precancerous cervical changes and cervical cancer. We next measured the samples in both groups using the IC method we originally developed, and found that the measurement values of IC highly correlated with those of MB-ELISA. The simple and quick IC method would be a useful tool for rapid monitoring of L1 specific antibody levels in a non-laboratory environment. With less than one drop of serum, our IC can easily detect serum HPV-16/-18 antibodies within 15 minutes, without the need for electronic devices or techniques.
Collapse
Affiliation(s)
- Fumiko Endo
- Department of Molecular Medical Research, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kami-Kitazawa, Setagaya-ku, Tokyo, Japan
| | - Tsutomu Tabata
- Department of Obstetrics and Gynecology, Faculty of Medicine, Mie University, 2–174 Edobashi, Tsu-city, Mie, Japan
| | - Daichi Sadato
- Department of Molecular Medical Research, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kami-Kitazawa, Setagaya-ku, Tokyo, Japan
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, Japan
| | - Machiko Kawamura
- Department of Pediatrics, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, Japan
| | - Noriyuki Ando
- Josei-Kokoro-Clinic, 1-1-9 Machiya, Arakawa-ku, Tokyo, Japan
| | - Keisuke Oboki
- Department of Molecular Medical Research, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kami-Kitazawa, Setagaya-ku, Tokyo, Japan
- * E-mail: (FS); (KO)
| | - Masako Ukaji
- Department of Molecular Medical Research, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kami-Kitazawa, Setagaya-ku, Tokyo, Japan
| | | | | | - Tomoaki Ikeda
- Department of Obstetrics and Gynecology, Faculty of Medicine, Mie University, 2–174 Edobashi, Tsu-city, Mie, Japan
| | - Futoshi Shibasaki
- Department of Molecular Medical Research, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kami-Kitazawa, Setagaya-ku, Tokyo, Japan
- * E-mail: (FS); (KO)
| |
Collapse
|
42
|
Developments in L2-based human papillomavirus (HPV) vaccines. Virus Res 2016; 231:166-175. [PMID: 27889616 DOI: 10.1016/j.virusres.2016.11.020] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 11/17/2016] [Accepted: 11/18/2016] [Indexed: 11/21/2022]
Abstract
Infections with sexually transmitted high-risk Human Papillomavirus (hrHPV), of which there are at least 15 genotypes, are responsible for a tremendous disease burden by causing cervical, and subsets of other ano-genital and oro-pharyngeal carcinomas, together representing 5% of all cancer cases worldwide. HPV subunit vaccines consisting of virus-like particles (VLP) self-assembled from major capsid protein L1 plus adjuvant have been licensed. Prophylactic vaccinations with the 2-valent (HPV16/18), 4-valent (HPV6/11/16/18), or 9-valent (HPV6/11/16/18/31/33/45/52/58) vaccine induce high-titer neutralizing antibodies restricted to the vaccine types that cause up to 90% of cervical carcinomas, a subset of other ano-genital and oro-pharyngeal cancers and 90% of benign ano-genital warts (condylomata). The complexity of manufacturing multivalent L1-VLP vaccines limits the number of included VLP types and thus the vaccines' spectrum of protection, leaving a panel of oncogenic mucosal HPV unaddressed. In addition, current vaccines do not protect against cutaneous HPV types causing benign skin warts, or against beta-papillomavirus (betaPV) types implicated in the development of non-melanoma skin cancer (NMSC) in immunosuppressed patients. In contrast with L1-VLP, the minor capsid protein L2 contains type-common epitopes that induce low-titer yet broadly cross-neutralizing antibodies to heterologous PV types and provide cross-protection in animal challenge models. Efforts to increase the low immunogenicity of L2 (poly)-peptides and thereby to develop broader-spectrum HPV vaccines are the focus of this review.
Collapse
|
43
|
Sun S, Gao F, Mao Q, Shao J, Jiang L, Liu D, Wang Y, Yao X, Wu X, Sun B, Zhao D, Ma Y, Lu J, Kong W, Jiang C, Liang Z. Immunogenicity and protective efficacy of an EV71 virus-like particle vaccine against lethal challenge in newborn mice. Hum Vaccin Immunother 2016; 11:2406-13. [PMID: 26036916 DOI: 10.1080/21645515.2015.1053675] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Enterovirus 71(EV71) has caused severe epidemics of hand, foot and mouth disease (HFMD) in the Asia Pacific in recent years, particularly in infants and pre-school children. It has become a serious public health threat, as currently there are no approved vaccines or antiviral drugs for EV71 infection. Many EV71 vaccines have been under development worldwide, however the main focus is inactivated EV71 vaccines. For example, the inactivated EV71 vaccine has recently finished phase III clinical trial in Mainland China. There have been very few studies on EV71 virus like particles (VLPs). In this study, the immunogenicity and protective potency of the EV71 VLPs produced in insect cells were evaluated in mice with different dosages. Our results showed that EV71 VLPs could elicit high titers of neutralizing antibodies (NTAbs) in a dose-dependent manner and NTAbs were sustained after the second injection with an average GMT (geometric mean titer) level from 19 to 2960 in immunized mice. Survival rates were 100%, 100%, 85%, and 40% after challenge with 15 LD50 (median lethal dose) of EV71 in these newborn mice, respectively. ED50 (50% effective dose) of VLPs was 0.20 μg/dose in newborn mice, while NTAb titer under this dosage was about 50. Passive protection was determined with 2 methods and demonstrated that the survival rates were positively correlated with NTAb titers, which at 24 and 54 induced 50% survival rates in experimental animals. The ED50 of VLP vaccines and the passive NTAb titers were also analyzed. The maternal NTAb titer was similar as the passive NTAb titer in the mouse model challenged with our lethal mouse EV71 strain. Hence, our work has provided preliminary data on the protection potency of VLPs as a vaccine candidate and would facilitate future VLP vaccine development.
Collapse
Affiliation(s)
- Shiyang Sun
- a School of Life Sciences; Jilin University ; Changchun , PR China.,b National Engineering Laboratory for AIDS Vaccine; Jilin University ; Changchun , PR China
| | - Fan Gao
- c National Institutes for Food and Drug Control ; Beijing , PR China
| | - Qunying Mao
- c National Institutes for Food and Drug Control ; Beijing , PR China
| | - Jie Shao
- a School of Life Sciences; Jilin University ; Changchun , PR China.,b National Engineering Laboratory for AIDS Vaccine; Jilin University ; Changchun , PR China
| | - Liping Jiang
- a School of Life Sciences; Jilin University ; Changchun , PR China.,b National Engineering Laboratory for AIDS Vaccine; Jilin University ; Changchun , PR China
| | - Dawei Liu
- d Changchun BCHT Biotechnology Co. ; Changchun , PR China
| | - Yiping Wang
- c National Institutes for Food and Drug Control ; Beijing , PR China
| | - Xin Yao
- c National Institutes for Food and Drug Control ; Beijing , PR China
| | - Xing Wu
- c National Institutes for Food and Drug Control ; Beijing , PR China
| | - Bo Sun
- d Changchun BCHT Biotechnology Co. ; Changchun , PR China
| | - Dandan Zhao
- a School of Life Sciences; Jilin University ; Changchun , PR China.,b National Engineering Laboratory for AIDS Vaccine; Jilin University ; Changchun , PR China
| | - Youlei Ma
- d Changchun BCHT Biotechnology Co. ; Changchun , PR China
| | - Jingcai Lu
- a School of Life Sciences; Jilin University ; Changchun , PR China.,b National Engineering Laboratory for AIDS Vaccine; Jilin University ; Changchun , PR China
| | - Wei Kong
- a School of Life Sciences; Jilin University ; Changchun , PR China.,b National Engineering Laboratory for AIDS Vaccine; Jilin University ; Changchun , PR China.,e Key Laboratory for Molecular Enzymology & Engineering; The Ministry of Education; Jilin University ; Changchun , PR China
| | - Chunlai Jiang
- a School of Life Sciences; Jilin University ; Changchun , PR China.,b National Engineering Laboratory for AIDS Vaccine; Jilin University ; Changchun , PR China.,e Key Laboratory for Molecular Enzymology & Engineering; The Ministry of Education; Jilin University ; Changchun , PR China
| | - Zhenglun Liang
- c National Institutes for Food and Drug Control ; Beijing , PR China
| |
Collapse
|
44
|
Zhang X, Li S, Modis Y, Li Z, Zhang J, Xia N, Zhao Q. Functional assessment and structural basis of antibody binding to human papillomavirus capsid. Rev Med Virol 2015; 26:115-28. [PMID: 26676802 DOI: 10.1002/rmv.1867] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 11/19/2015] [Accepted: 11/20/2015] [Indexed: 02/05/2023]
Abstract
Persistent high-risk human papillomavirus (HPV) infection is linked to cervical cancer. Two prophylactic virus-like particle (VLP)-based vaccines have been marketed globally for nearly a decade. Here, we review the HPV pseudovirion (PsV)-based assays for the functional assessment of the HPV neutralizing antibodies and the structural basis for these clinically relevant epitopes. The PsV-based neutralization assay was developed to evaluate the efficacy of neutralization antibodies in sera elicited by vaccination or natural infection or to assess the functional characteristics of monoclonal antibodies. Different antibody binding modes were observed when an antibody was complexed with virions, PsVs or VLPs. The neutralizing epitopes are localized on surface loops of the L1 capsid protein, at various locations on the capsomere. Different neutralization antibodies exert their neutralizing function via different mechanisms. Some antibodies neutralize the virions by inducing conformational changes in the viral capsid, which can result in concealing the binding site for a cellular receptor like 1A1D-2 against dengue virus, or inducing premature genome release like E18 against enterovirus 71. Higher-resolution details on the epitope composition of HPV neutralizing antibodies would shed light on the structural basis of the highly efficacious vaccines and aid the design of next generation vaccines. In-depth understanding of epitope composition would ensure the development of function-indicating assays for the comparability exercise to support process improvement or process scale up. Elucidation of the structural elements of the type-specific epitopes would enable rational design of cross-type neutralization via epitope re-engineering or epitope grafting in hybrid VLPs.
Collapse
Affiliation(s)
- Xiao Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, China.,School of Public Health, Xiamen University, Xiamen, Fujian, 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, China.,School of Public Health, Xiamen University, Xiamen, Fujian, China.,School of Life Science, Xiamen University, Xiamen, Fujian, China
| | - Yorgo Modis
- Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Zhihai Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, China.,School of Life Science, Xiamen University, Xiamen, Fujian, China
| | - Jun Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, China.,School of Public Health, Xiamen University, Xiamen, Fujian, 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, China.,School of Public Health, Xiamen University, Xiamen, Fujian, China.,School of Life Science, Xiamen University, Xiamen, Fujian, China
| | - Qinjian Zhao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, China.,School of Public Health, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
45
|
Martins KAO, Cooper CL, Stronsky SM, Norris SLW, Kwilas SA, Steffens JT, Benko JG, van Tongeren SA, Bavari S. Adjuvant-enhanced CD4 T Cell Responses are Critical to Durable Vaccine Immunity. EBioMedicine 2015; 3:67-78. [PMID: 26870818 PMCID: PMC4739439 DOI: 10.1016/j.ebiom.2015.11.041] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/23/2015] [Accepted: 11/24/2015] [Indexed: 01/08/2023] Open
Abstract
Protein-based vaccines offer a safer alternative to live-attenuated or inactivated vaccines but have limited immunogenicity. The identification of adjuvants that augment immunogenicity, specifically in a manner that is durable and antigen-specific, is therefore critical for advanced development. In this study, we use the filovirus virus-like particle (VLP) as a model protein-based vaccine in order to evaluate the impact of four candidate vaccine adjuvants on enhancing long term protection from Ebola virus challenge. Adjuvants tested include poly-ICLC (Hiltonol), MPLA, CpG 2395, and alhydrogel. We compared and contrasted antibody responses, neutralizing antibody responses, effector T cell responses, and T follicular helper (Tfh) cell frequencies with each adjuvant's impact on durable protection. We demonstrate that in this system, the most effective adjuvant elicits a Th1-skewed antibody response and strong CD4 T cell responses, including an increase in Tfh frequency. Using immune-deficient animals and adoptive transfer of serum and cells from vaccinated animals into naïve animals, we further demonstrate that serum and CD4 T cells play a critical role in conferring protection within effective vaccination regimens. These studies inform on the requirements of long term immune protection, which can potentially be used to guide screening of clinical-grade adjuvants for vaccine clinical development. Adjuvants can prolong the protection afforded by protein-based vaccines and impact adaptive immune responses Enhanced CD4 T cell responses, helper and effector, correlate with duration of protection Durable protection from ma-EBOV is associated with Tfh frequency, Th1 antibody titers, and effector CD4 T cells
Protein-based vaccines are extremely safe, but they sometimes require the addition of adjuvants to enhance immunogenicity. In this study, we compared the impact of multiple adjuvants on immunogenicity, focusing on the duration of vaccine-mediated protection in mice. We then looked at how each adjuvant impacted the immune response in order to identify correlates of that long lasting immunity. The most effective adjuvant/vaccine combinations elicited multifunctional CD4 T cell responses and a Th1-skewed antibody response. By transferring antigen-experienced CD4 T cells and serum into naïve animals, we demonstrated that both CD4 T cells and serum were critical for durable vaccine-mediated protection.
Collapse
Key Words
- Adjuvant
- BME, beta mercaptoethanol
- CD, cluster of differentiation
- DSCF, Dwass, Steel, Critchlow-Fligner
- Durable protection
- ELISA, Enzyme linked immunosorbent assay
- ELISPOT, enzyme-linked immunospot assay
- Ebola virus
- FACS, fluorescence activated cell sorting
- FBS, fetal bovine serum
- GP, glycoprotein
- IACUC, Institutional Animal Care and Use Committee
- IM, intramuscular
- IP, intraperitoneal
- IQR, interquartile range
- Immune correlates
- LN, lymph node
- MPLA, monophosphoryl lipid A
- NAb, neutralizing antibody
- Ns, not significant
- PBS, phosphate buffered saline
- PRR, pattern recognition receptor
- Pfu, plaque forming unit
- PsVNA, pseudovirion neutralization assay
- TLR, Toll-like receptor
- USAMRIID, United States Army Medical Research Institute of Infectious Diseases
- VLP, virus-like particle
- Vaccine
- ma-EBOV, mouse-adapted Ebola virus
Collapse
Affiliation(s)
- Karen A O Martins
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Christopher L Cooper
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Sabrina M Stronsky
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Sarah L W Norris
- Research Support Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Steven A Kwilas
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Jesse T Steffens
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Jacqueline G Benko
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Sean A van Tongeren
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Sina Bavari
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA.
| |
Collapse
|
46
|
Godi A, Bissett SL, Miller E, Beddows S. Relationship between Humoral Immune Responses against HPV16, HPV18, HPV31 and HPV45 in 12-15 Year Old Girls Receiving Cervarix® or Gardasil® Vaccine. PLoS One 2015; 10:e0140926. [PMID: 26495976 PMCID: PMC4619723 DOI: 10.1371/journal.pone.0140926] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 10/01/2015] [Indexed: 12/22/2022] Open
Abstract
Background Human papillomavirus (HPV) vaccines confer protection against the oncogenic genotypes HPV16 and HPV18 through the generation of type-specific neutralizing antibodies raised against virus-like particles (VLP) representing these genotypes. The vaccines also confer a degree of cross-protection against HPV31 and HPV45, which are genetically-related to the vaccine types HPV16 and HPV18, respectively, although the mechanism is less certain. There are a number of humoral immune measures that have been examined in relation to the HPV vaccines, including VLP binding, pseudovirus neutralization and the enumeration of memory B cells. While the specificity of responses generated against the vaccine genotypes are fairly well studied, the relationship between these measures in relation to non-vaccine genotypes is less certain. Methods We carried out a comparative study of these immune measures against vaccine and non-vaccine genotypes using samples collected from 12–15 year old girls following immunization with three doses of either Cervarix® or Gardasil® HPV vaccine. Results The relationship between neutralizing and binding antibody titers and HPV-specific memory B cell levels for the vaccine genotypes, HPV16 and HPV18, were very good. The proportion of responders approached 100% for both vaccines while the magnitude of these responses induced by Cervarix® were generally higher than those following Gardasil® immunization. A similar pattern was found for the non-vaccine genotype HPV31, albeit at a lower magnitude compared to its genetically-related vaccine genotype, HPV16. However, both the enumeration of memory B cells and VLP binding responses against HPV45 were poorly related to its neutralizing antibody responses. Purified IgG derived from memory B cells demonstrated specificities similar to those found in the serum, including the capacity to neutralize HPV pseudoviruses. Conclusions These data suggest that pseudovirus neutralization should be used as the preferred humoral immune measure for studying HPV vaccine responses, particularly for non-vaccine genotypes.
Collapse
Affiliation(s)
- Anna Godi
- Virus Reference Department, Public Health England, London, United Kingdom
| | - Sara L. Bissett
- Virus Reference Department, Public Health England, London, United Kingdom
| | - Elizabeth Miller
- National Vaccine Evaluation Consortium, Public Health England, London, United Kingdom
| | - Simon Beddows
- Virus Reference Department, Public Health England, London, United Kingdom
- * E-mail:
| |
Collapse
|
47
|
Einstein MH, Takacs P, Chatterjee A, Sperling RS, Chakhtoura N, Blatter MM, Lalezari J, David MP, Lin L, Struyf F, Dubin G. Comparison of long-term immunogenicity and safety of human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine and HPV-6/11/16/18 vaccine in healthy women aged 18-45 years: end-of-study analysis of a Phase III randomized trial. Hum Vaccin Immunother 2015; 10:3435-45. [PMID: 25483701 PMCID: PMC4514070 DOI: 10.4161/hv.36121] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The observer-blind, randomized, age-stratified, head-to-head study (NCT00423046) comparing immunogenicity and safety of HPV-16/18 and HPV-6/11/16/18 vaccines in healthy women aged 18-45 y was completed. Five y after vaccination, in subjects from the Month 60 according-to-protocol cohort (seronegative and DNA negative for HPV type analyzed at baseline), serum neutralizing antibody (nAb) responses induced by HPV-16/18 vaccine remained 7.8-fold (18-26-y stratum), 5.6-fold (27-35-y stratum) and 2.3-fold (36-45-y stratum) higher than those induced by HPV-6/11/16/18 vaccine for HPV-16. For HPV-18, the fold differences were 12.1, 13.0 and 7.8, respectively. At Month 60, all (100%) subjects in HPV-16/18 vaccine group and the majority (95.7%-97.5%) in HPV-6/11/16/18 vaccine group were seropositive for HPV-16. For HPV-18, the majority (98.1%-100%) of subjects in HPV-16/18 vaccine group were seropositive; however, seropositivity rates in HPV-6/11/16/18 vaccine group decreased considerably (61.1%-76.9%) across the 3 age strata. In the total vaccinated cohort (received ≥1 dose regardless of baseline HPV serostatus and DNA status), geometric mean titers for anti-HPV-16 and anti-HPV-18 nAb were higher in HPV-16/18 vaccine group than in HPV-6/11/16/18 vaccine group. Based on the 5-y data, piece-wise and modified power-law models predicted a longer durability of nAb response for HPV-16/18 vaccine compared to HPV-6/11/16/18 vaccine. Beyond the differences apparent between the vaccines in terms of immunogenicity and modeled persistence of antibody responses, comparative studies including clinical endpoints would be needed to determine whether differences exist in duration of vaccine-induced protection.
Collapse
Key Words
- 50 μg) adsorbed on aluminum salt (500 μg Al3+)
- AAHS, amorphous aluminum hydroxyphosphate sulfate
- ANOVA, analysis of variance
- AS04, Adjuvant System containing 3-O-desacyl-4’-monophosphoryl lipid A (MPL
- ATP, according-to-protocol
- CI, confidence interval
- Cervarix®
- ED50, effective dose producing 50% response
- ELISA, enzyme-linked immunosorbent assay
- GMT, geometric mean titer
- Gardasil®
- HPV, human papillomavirus
- MSC, medically significant condition
- NOAD, new onset autoimmune disease
- NOCD, new onset chronic disease
- PBNA, pseudovirion-based neutralization assay
- SAE, serious adverse event
- SP, seropositivity
- TVC, total vaccinated cohort
- antibodies
- human papillomavirus
- immunogenicity
- models
- nAb, neutralizing antibodies
- neutralizing
- safety
- statistical
Collapse
Affiliation(s)
- Mark H Einstein
- a Montefiore Medical Center and Albert Einstein College of Medicine; Obstetrics & Gynecology and Women's Health ; Bronx , NY USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Zhang L, Wang W, Wang S. Effect of vaccine administration modality on immunogenicity and efficacy. Expert Rev Vaccines 2015; 14:1509-23. [PMID: 26313239 DOI: 10.1586/14760584.2015.1081067] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The many factors impacting the efficacy of a vaccine can be broadly divided into three categories: features of the vaccine itself, including immunogen design, vaccine type, formulation, adjuvant and dosing; individual variations among vaccine recipients and vaccine administration-related parameters. While much literature exists related to vaccines, and recently systems biology has started to dissect the impact of individual subject variation on vaccine efficacy, few studies have focused on the role of vaccine administration-related parameters on vaccine efficacy. Parenteral and mucosal vaccinations are traditional approaches for licensed vaccines; novel vaccine delivery approaches, including needless injection and adjuvant formulations, are being developed to further improve vaccine safety and efficacy. This review provides a brief summary of vaccine administration-related factors, including vaccination approach, delivery route and method of administration, to gain a better understanding of their potential impact on the safety and immunogenicity of candidate vaccines.
Collapse
Affiliation(s)
- Lu Zhang
- a 1 Department of Infectious Diseases, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China.,b 2 China-US Vaccine Research Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Wei Wang
- c 3 Wang Biologics, LLC, Chesterfield, MO 63017, USA ; Current affiliation: Bayer HealthCare, Berkeley, CA 94710, USA
| | - Shixia Wang
- d 4 Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|