1
|
Bahojb Mahdavi SZ, Jebelli A, Aghbash PS, Baradaran B, Amini M, Oroojalian F, Pouladi N, Baghi HB, de la Guardia M, Mokhtarzadeh AA. A comprehensive overview on the crosstalk between microRNAs and viral pathogenesis and infection. Med Res Rev 2024. [PMID: 39185567 DOI: 10.1002/med.22073] [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: 11/06/2021] [Revised: 04/11/2023] [Accepted: 08/04/2024] [Indexed: 08/27/2024]
Abstract
Infections caused by viruses as the smallest infectious agents, pose a major threat to global public health. Viral infections utilize different host mechanisms to facilitate their own propagation and pathogenesis. MicroRNAs (miRNAs), as small noncoding RNA molecules, play important regulatory roles in different diseases, including viral infections. They can promote or inhibit viral infection and have a pro-viral or antiviral role. Also, viral infections can modulate the expression of host miRNAs. Furthermore, viruses from different families evade the host immune response by producing their own miRNAs called viral miRNAs (v-miRNAs). Understanding the replication cycle of viruses and their relation with host miRNAs and v-miRNAs can help to find new treatments against viral infections. In this review, we aim to outline the structure, genome, and replication cycle of various viruses including hepatitis B, hepatitis C, influenza A virus, coronavirus, human immunodeficiency virus, human papillomavirus, herpes simplex virus, Epstein-Barr virus, Dengue virus, Zika virus, and Ebola virus. We also discuss the role of different host miRNAs and v-miRNAs and their role in the pathogenesis of these viral infections.
Collapse
Affiliation(s)
- Seyedeh Zahra Bahojb Mahdavi
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asiyeh Jebelli
- Department of Biological Science, Faculty of Basic Science, Higher Education Institute of Rab-Rashid, Tabriz, Iran
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Nasser Pouladi
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Hossein Bannazadeh Baghi
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Miguel de la Guardia
- Department of Analytical Chemistry, University of Valencia, Burjassot, Valencia, Spain
| | | |
Collapse
|
2
|
Catalán-Castorena O, Garibay-Cerdenares OL, Illades-Aguiar B, Rodríguez-Ruiz HA, Zubillaga-Guerrero MI, Leyva-Vázquez MA, Encarnación-Guevara S, Alarcón-Romero LDC. The role of HR-HPV integration in the progression of premalignant lesions into different cancer types. Heliyon 2024; 10:e34999. [PMID: 39170128 PMCID: PMC11336306 DOI: 10.1016/j.heliyon.2024.e34999] [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] [Received: 12/15/2023] [Revised: 07/19/2024] [Accepted: 07/21/2024] [Indexed: 08/23/2024] Open
Abstract
High-risk human papillomavirus (HR-HPV) is associated with the development of different types of cancer, such as cervical, head and neck (including oral, laryngeal, and oropharyngeal), vulvar, vaginal, penile, and anal cancers. The progression of premalignant lesions to cancer depends on factors associated with the host cell and the different epithelia infected by HPV, such as basal cells of the flat epithelium and the cells of the squamocolumnar transformation zone (STZ) found in the uterine cervix and the anal canal, which is rich in heparan sulfate proteoglycans and integrin-like receptors. On the other hand, factors associated with the viral genotype, infection with multiple viruses, viral load, viral persistence, and type of integration determine the viral breakage pattern and the sites at which the virus integrates into the host cell genome (introns, exons, intergenic regions), inducing the loss of function of tumor suppressor genes and increasing oncogene expression. This review describes the role of viral integration and the molecular mechanisms induced by HR-HPV in different types of tissues. The purpose of this review is to identify the common factors associated with the role of integration events in the progression of premalignant lesions in different types of cancer.
Collapse
Affiliation(s)
- Oscar Catalán-Castorena
- Research in Cytopathology and Histochemical Laboratory, Faculty of Chemical and Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero, 39089, Mexico
| | - Olga Lilia Garibay-Cerdenares
- Molecular Biomedicine Laboratory, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero, 39089, Mexico
- CONAHCyT-Autonomous University of Guerrero, Chilpancingo, Guerrero, 39089, Mexico
| | - Berenice Illades-Aguiar
- Molecular Biomedicine Laboratory, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero, 39089, Mexico
| | - Hugo Alberto Rodríguez-Ruiz
- Molecular Biomedicine Laboratory, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero, 39089, Mexico
| | - Ma. Isabel Zubillaga-Guerrero
- Research in Cytopathology and Histochemical Laboratory, Faculty of Chemical and Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero, 39089, Mexico
| | - Marco Antonio Leyva-Vázquez
- Molecular Biomedicine Laboratory, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero, 39089, Mexico
| | | | - Luz del Carmen Alarcón-Romero
- Research in Cytopathology and Histochemical Laboratory, Faculty of Chemical and Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero, 39089, Mexico
| |
Collapse
|
3
|
Gray P, Mariz FC, Eklund C, Eriksson T, Faust H, Kann H, Müller M, Paavonen J, Pimenoff VN, Sehr P, Surcel HM, Dillner J, Waterboer T, Lehtinen M. Lack of detectable HPV18 antibodies in 14% of quadrivalent vaccinees in a longitudinal cohort study. NPJ Vaccines 2024; 9:146. [PMID: 39138224 PMCID: PMC11322158 DOI: 10.1038/s41541-024-00941-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 07/30/2024] [Indexed: 08/15/2024] Open
Abstract
Although HPV vaccines are highly efficacious, a notable proportion of quadrivalent vaccinees are HPV18 seronegative post-vaccination. We have investigated this findings' validity by comparing vaccine-induced antibody responses using two different immunoassays. 6558 16-17-year-old females participated in the FUTURE II (NCT00092534) and PATRICIA (NCT00122681) trials in 2002-2004. Both the quadrivalent and bivalent vaccine recipients (QVR and BVR) received three doses. Twelve-year follow-up for 648 vaccinees was conducted by the Finnish Maternity Cohort. The presence of neutralising and binding HPV antibodies was analysed via HPV pseudovirion-based neutralisation and pseudovirion-binding assays. Four percent and 14.3% of the QVRs were seronegative for neutralising and binding antibodies to HPV16 and HPV18, respectively. No BVRs were HPV16/18 seronegative post-vaccination. The antibody titres were strongly correlated between the assays, Pearson's correlation coefficient, r[HPV16] = 0.92 and 0.85, and r[HPV18] = 0.91 and 0.86 among the QVRs and BVRs respectively. Fourteen percent of QVRs lacked detectable HPV18 antibodies in long-term follow-up.
Collapse
Affiliation(s)
- Penelope Gray
- Center for Cervical Cancer Elimination, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
| | - Filipe Colaço Mariz
- Tumorvirus-Specific Vaccination Strategies, Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer Feld 242, 69120, Heidelberg, Germany
| | - Carina Eklund
- Center for Cervical Cancer Elimination, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Tiina Eriksson
- Tampere University, Faculty of Medicine and Health Technology, Tampere, Finland
- Wellbeing services county of Pirkanmaa, PIRHA, Tays Research Services, Tampere, Finland
| | - Helena Faust
- Medical Products Agency Läkemedelsverket, Uppsala, Sweden
| | - Hanna Kann
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Martin Müller
- Tumorvirus-Specific Vaccination Strategies, Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer Feld 242, 69120, Heidelberg, Germany
| | - Jorma Paavonen
- Medical Faculty, University of Helsinki, Helsinki, Finland
| | - Ville N Pimenoff
- Center for Cervical Cancer Elimination, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biobank Borealis of Northern Finland, University of Oulu, Oulu, Finland
| | - Peter Sehr
- EMBL-DKFZ Chemical Biology Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Heljä-Marja Surcel
- Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biobank Borealis of Northern Finland, University of Oulu, Oulu, Finland
| | - Joakim Dillner
- Center for Cervical Cancer Elimination, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Tim Waterboer
- Infections and Cancer Epidemiology, Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer Feld 242, 69120, Heidelberg, Germany
| | - Matti Lehtinen
- Center for Cervical Cancer Elimination, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Tampere University, Faculty of Medicine and Health Technology, Tampere, Finland
| |
Collapse
|
4
|
Mariz FC, Putzker K, Sehr P, Müller M. Advances on two serological assays for human papillomavirus provide insights on the reactivity of antibodies against a cross-neutralization epitope of the minor capsid protein L2. Front Immunol 2023; 14:1272018. [PMID: 38022617 PMCID: PMC10663238 DOI: 10.3389/fimmu.2023.1272018] [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: 08/03/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction A second generation of prophylactic human papillomavirus (HPV) vaccines based on the minor capsid protein L2 has entered clinical trials as promising alternative to meet the gaps left out by the current vaccines concerning type-restricted protection, high costs and low penetrance in immunization programs of lowand middle-income countries. Most of the serological assays available to assess anti-HPV humoral responses are, however, not well suited for measuring vaccine-induced anti-L2 antibody responses. Methods In this work, we have advanced our automated, purely add-on High-Throughput Pseudovirion-Based Neutralization Assay (HT-PBNA) in an L2-oriented approach for measuring antibody-mediated neutralization of HPV types 6/16/18/31/33/52/58. Results and discussion With the optimized settings, we observed 24- to 120-fold higher sensitivity for detection of neutralizing Ab to the L2 protein of HPV6, HPV16, HPV18, and HPV31, compared to the standard HT-PBNA. Alternatively, we have also developed a highly sensitive, cell-free, colorimetric L2-peptide capture ELISA for which the results were strongly concordant with those of the advanced neutralization assay, named HT-fc-PBNA. These two high-throughput scalable assays represent attractive approaches to determine antibody-based correlates of protection for the HPV L2 vaccines that are to come.
Collapse
Affiliation(s)
- Filipe Colaco Mariz
- Tumorvirus-Specific Vaccination Strategies (F035), Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Kerstin Putzker
- EMBL-DKFZ Chemical Biology Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Peter Sehr
- EMBL-DKFZ Chemical Biology Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Martin Müller
- Tumorvirus-Specific Vaccination Strategies (F035), Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| |
Collapse
|
5
|
Abstract
Neutralizing antibodies (nAbs) are being increasingly used as passive antiviral reagents in prophylactic and therapeutic modalities and to guide viral vaccine design. In vivo, nAbs can mediate antiviral functions through several mechanisms, including neutralization, which is defined by in vitro assays in which nAbs block viral entry to target cells, and antibody effector functions, which are defined by in vitro assays that evaluate nAbs against viruses and infected cells in the presence of effector systems. Interpreting in vivo results in terms of these in vitro assays is challenging but important in choosing optimal passive antibody and vaccine strategies. Here, I review findings from many different viruses and conclude that, although some generalizations are possible, deciphering the relative contributions of different antiviral mechanisms to the in vivo efficacy of antibodies currently requires consideration of individual antibody-virus interactions.
Collapse
Affiliation(s)
- Dennis R Burton
- Department of Immunology and Microbiology, Consortium for HIV/AIDS Vaccine Development, International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA.
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
| |
Collapse
|
6
|
Purcell RA, Theisen RM, Arnold KB, Chung AW, Selva KJ. Polyfunctional antibodies: a path towards precision vaccines for vulnerable populations. Front Immunol 2023; 14:1183727. [PMID: 37600816 PMCID: PMC10433199 DOI: 10.3389/fimmu.2023.1183727] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/30/2023] [Indexed: 08/22/2023] Open
Abstract
Vaccine efficacy determined within the controlled environment of a clinical trial is usually substantially greater than real-world vaccine effectiveness. Typically, this results from reduced protection of immunologically vulnerable populations, such as children, elderly individuals and people with chronic comorbidities. Consequently, these high-risk groups are frequently recommended tailored immunisation schedules to boost responses. In addition, diverse groups of healthy adults may also be variably protected by the same vaccine regimen. Current population-based vaccination strategies that consider basic clinical parameters offer a glimpse into what may be achievable if more nuanced aspects of the immune response are considered in vaccine design. To date, vaccine development has been largely empirical. However, next-generation approaches require more rational strategies. We foresee a generation of precision vaccines that consider the mechanistic basis of vaccine response variations associated with both immunogenetic and baseline health differences. Recent efforts have highlighted the importance of balanced and diverse extra-neutralising antibody functions for vaccine-induced protection. However, in immunologically vulnerable populations, significant modulation of polyfunctional antibody responses that mediate both neutralisation and effector functions has been observed. Here, we review the current understanding of key genetic and inflammatory modulators of antibody polyfunctionality that affect vaccination outcomes and consider how this knowledge may be harnessed to tailor vaccine design for improved public health.
Collapse
Affiliation(s)
- Ruth A. Purcell
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Robert M. Theisen
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Kelly B. Arnold
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Amy W. Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Kevin J. Selva
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
7
|
Joshi S, Anantharaman D, Muwonge R, Bhatla N, Panicker G, Butt J, Rani Reddy Poli U, Malvi SG, Esmy PO, Lucas E, Verma Y, Shah A, Zomawia E, Pimple S, Jayant K, Hingmire S, Chiwate A, Divate U, Vashist S, Mishra G, Jadhav R, Siddiqi M, Sankaran S, Pillai Rameshwari Ammal Kannan T, Kartha P, Shastri SS, Sauvaget C, Radhakrishna Pillai M, Waterboer T, Müller M, Sehr P, Unger ER, Sankaranarayanan R, Basu P. Evaluation of immune response to single dose of quadrivalent HPV vaccine at 10-year post-vaccination. Vaccine 2023; 41:236-245. [PMID: 36446654 PMCID: PMC9792650 DOI: 10.1016/j.vaccine.2022.11.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/02/2022] [Accepted: 11/19/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND The recent World Health Organization recommendation supporting single-dose of HPV vaccine will significantly reduce programmatic cost, mitigate the supply shortage, and simplify logistics, thus allowing more low- and middle-income countries to introduce the vaccine. From a programmatic perspective the durability of protection offered by a single-dose will be a key consideration. The primary objectives of the present study were to determine whether recipients of a single-dose of quadrivalent HPV vaccine had sustained immune response against targeted HPV types (HPV 6,11,16,18) at 10 years post-vaccination and whether this response was superior to the natural antibody titres observed in unvaccinated women. METHODS Participants received at age 10-18 years either one, two or three doses of the quadrivalent HPV vaccine. Serology samples were obtained at different timepoints up to 10 years after vaccination from a convenience sample of vaccinated participants and from age-matched unvaccinated women at one timepoint. The evolution of the binding and neutralizing antibody response was presented by dose received. 10-year durability of immune responses induced by a single-dose was compared to that after three doses of the vaccine and in unvaccinated married women. RESULTS The dynamics of antibody response among the single-dose recipients observed over 120 months show stabilized levels 18 months after vaccination for all four HPV types. Although the HPV type-specific (binding or neutralizing) antibody titres after a single-dose were significantly inferior to those after three doses of the vaccine (lower bounds of GMT ratios < 0.5), they were all significantly higher than those observed in unvaccinated women following natural infections (GMT ratios: 2.05 to 4.04-fold higher). The results correlate well with the high vaccine efficacy of single-dose against persistent HPV 16/18 infections reported by us earlier at 10-years post-vaccination. CONCLUSION Our study demonstrates the high and durable immune response in single-dose recipients of HPV vaccine at 10-years post vaccination.
Collapse
Affiliation(s)
- Smita Joshi
- Jehangir Clinical Development Centre, Jehangir Hospital Premises, Pune 411 001, India
| | - Devasena Anantharaman
- Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram 695 014, Kerala, India
| | - Richard Muwonge
- Early Detection, Prevention & Infections Branch, International Agency for Research on Cancer, Lyon, France
| | - Neerja Bhatla
- Department of Obstetrics & Gynaecology, All India Institute of Medical Sciences, New Delhi 110 029, India
| | - Gitika Panicker
- National Center for Emerging and Zoonotic Infectious Diseases, CDC, USA
| | - Julia Butt
- Infection, Inflammation and Cancer Program, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, D-69120 Heidelberg, Germany
| | | | - Sylla G Malvi
- Tata Memorial Centre Rural Cancer Project, Nargis Dutt Memorial Cancer Hospital, Barshi District Solapur, Maharashtra 413 401, India
| | - Pulikkottil O Esmy
- Christian Fellowship Community Health Centre, Ambillikai (near Oddanchathram), Dindigul District, Tamil Nadu 624 612, India
| | - Eric Lucas
- Early Detection, Prevention & Infections Branch, International Agency for Research on Cancer, Lyon, France
| | - Yogesh Verma
- Sikkim Manipal Institute of Medical Sciences, Sikkim Manipal University, Gangtok, Sikkim 737102, India
| | - Anand Shah
- Gujarat Cancer & Research Institute (GCRI), M.P. Shah Cancer Hospital, Civil Hospital Campus, Asarwa, Ahmedabad 380 016, India
| | | | - Sharmila Pimple
- Department of Preventive Oncology, Centre for Cancer Epidemiology (CCE), Homi Bhabha National Institute, Tata Memorial Centre, Mumbai, India
| | - Kasturi Jayant
- Tata Memorial Centre Rural Cancer Project, Nargis Dutt Memorial Cancer Hospital, Barshi District Solapur, Maharashtra 413 401, India
| | - Sanjay Hingmire
- Tata Memorial Centre Rural Cancer Project, Nargis Dutt Memorial Cancer Hospital, Barshi District Solapur, Maharashtra 413 401, India
| | - Aruna Chiwate
- Tata Memorial Centre Rural Cancer Project, Nargis Dutt Memorial Cancer Hospital, Barshi District Solapur, Maharashtra 413 401, India
| | - Uma Divate
- Jehangir Clinical Development Centre, Jehangir Hospital Premises, Pune 411 001, India
| | - Shachi Vashist
- Department of Obstetrics & Gynaecology, All India Institute of Medical Sciences, New Delhi 110 029, India
| | - Gauravi Mishra
- Department of Preventive Oncology, Centre for Cancer Epidemiology (CCE), Homi Bhabha National Institute, Tata Memorial Centre, Mumbai, India
| | - Radhika Jadhav
- Jehangir Clinical Development Centre, Jehangir Hospital Premises, Pune 411 001, India
| | - Maqsood Siddiqi
- Cancer Foundation of India, Kolkata, West Bengal 700 039, India
| | - Subha Sankaran
- Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram 695 014, Kerala, India
| | | | - Purnima Kartha
- Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram 695 014, Kerala, India
| | - Surendra S Shastri
- Department of Health Disparities Research, Division of Cancer Prevention and Population Sciences, University of Texas M.D. Anderson Cancer Centre, 1400 Pressler St, Houston, TX 77030-3906, United States
| | - Catherine Sauvaget
- Early Detection, Prevention & Infections Branch, International Agency for Research on Cancer, Lyon, France
| | - M Radhakrishna Pillai
- Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram 695 014, Kerala, India
| | - Tim Waterboer
- Infection, Inflammation and Cancer Program, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, D-69120 Heidelberg, Germany
| | - Martin Müller
- Infection, Inflammation and Cancer Program, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, D-69120 Heidelberg, Germany
| | - Peter Sehr
- EMBL-DKFZ Chemical Biology Core Facility, European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - Elizabeth R Unger
- National Center for Emerging and Zoonotic Infectious Diseases, CDC, USA
| | - Rengaswamy Sankaranarayanan
- Early Detection, Prevention & Infections Branch, International Agency for Research on Cancer, Lyon, France; Karkinos Healthcare, Kerala Operations, Ernakulam, India
| | - Partha Basu
- Early Detection, Prevention & Infections Branch, International Agency for Research on Cancer, Lyon, France.
| |
Collapse
|
8
|
Pseudotyped Virus for Papillomavirus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1407:85-103. [PMID: 36920693 DOI: 10.1007/978-981-99-0113-5_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Papillomavirus is difficult to culture in vitro, which limits its related research. The development of pseudotyped virus technology provides a valuable research tool for virus infectivity research, vaccine evaluation, infection inhibitor evaluation, and so on. Depending on the application fields, different measures have been developed to generate various kinds of pseudotyped papillomavirus. L1-based and L2-based HPV vaccines should be evaluated using different pseudotyped virus system. Pseudotyped papillomavirus animal models need high-titer pseudotyped virus and unique handling procedure to generate robust results. This paper reviewed the development, optimization, standardization, and application of various pseudotyped papillomavirus methods.
Collapse
|
9
|
Ahmels M, Mariz FC, Braspenning-Wesch I, Stephan S, Huber B, Schmidt G, Cao R, Müller M, Kirnbauer R, Rösl F, Hasche D. Next generation L2-based HPV vaccines cross-protect against cutaneous papillomavirus infection and tumor development. Front Immunol 2022; 13:1010790. [PMID: 36263027 PMCID: PMC9574214 DOI: 10.3389/fimmu.2022.1010790] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/16/2022] [Indexed: 11/25/2022] Open
Abstract
Licensed L1-VLP-based immunizations against high-risk mucosal human papillomavirus (HPV) types have been a great success in reducing anogenital cancers, although they are limited in their cross-protection against HPV types not covered by the vaccine. Further, their utility in protection against cutaneous HPV types, of which some contribute to non-melanoma skin cancer (NMSC) development, is rather low. Next generation vaccines achieve broadly cross-protective immunity against highly conserved sequences of L2. In this exploratory study, we tested two novel HPV vaccine candidates, HPV16 RG1-VLP and CUT-PANHPVAX, in the preclinical natural infection model Mastomys coucha. After immunization with either vaccines, a mock control or MnPV L1-VLPs, the animals were experimentally infected and monitored. Besides vaccine-specific seroconversion against HPV L2 peptides, the animals also developed cross-reactive antibodies against the cutaneous Mastomys natalensis papillomavirus (MnPV) L2, which were cross-neutralizing MnPV pseudovirions in vitro. Further, both L2-based vaccines also conferred in vivo protection as the viral loads in plucked hair after experimental infection were lower compared to mock-vaccinated control animals. Importantly, the formation of neutralizing antibodies, whether directed against L1-VLPs or L2, was able to prevent skin tumor formation and even microscopical signs of MnPV infection in the skin. For the first time, our study shows the proof-of-principle of next generation L2-based vaccines even across different PV genera in an infection animal model with its genuine PV. It provides fundamental insights into the humoral immunity elicited by L2-based vaccines against PV-induced skin tumors, with important implications to the design of next generation HPV vaccines.
Collapse
Affiliation(s)
- Melinda Ahmels
- Division of Viral Transformation Mechanisms, Research Program “Infection, Inflammation and Cancer”, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Filipe C. Mariz
- Research Group Tumorvirus-specific Vaccination Strategies, Research Program “Infection, Inflammation and Cancer”, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ilona Braspenning-Wesch
- Division of Viral Transformation Mechanisms, Research Program “Infection, Inflammation and Cancer”, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sonja Stephan
- Division of Viral Transformation Mechanisms, Research Program “Infection, Inflammation and Cancer”, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bettina Huber
- Laboratory of Viral Oncology, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Gabriele Schmidt
- Core Facility Unit Light Microscopy, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rui Cao
- Division of Viral Transformation Mechanisms, Research Program “Infection, Inflammation and Cancer”, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Müller
- Research Group Tumorvirus-specific Vaccination Strategies, Research Program “Infection, Inflammation and Cancer”, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Reinhard Kirnbauer
- Laboratory of Viral Oncology, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Frank Rösl
- Division of Viral Transformation Mechanisms, Research Program “Infection, Inflammation and Cancer”, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel Hasche
- Division of Viral Transformation Mechanisms, Research Program “Infection, Inflammation and Cancer”, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
10
|
Olczak P, Wong M, Tsai HL, Wang H, Kirnbauer R, Griffith AJ, Lambert PF, Roden R. Vaccination with human alphapapillomavirus-derived L2 multimer protects against human betapapillomavirus challenge, including in epidermodysplasia verruciformis model mice. Virology 2022; 575:63-73. [PMID: 36070626 PMCID: PMC9710205 DOI: 10.1016/j.virol.2022.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/18/2022] [Accepted: 08/18/2022] [Indexed: 11/22/2022]
Abstract
Human alphapapillomaviruses (αHPV) infect genital mucosa, and a high-risk subset is a necessary cause of cervical cancer. Licensed L1 virus-like particle (VLP) vaccines offer immunity against the nine most common αHPV associated with cervical cancer and genital warts. However, vaccination with an αHPV L2-based multimer vaccine, α11-88x5, protected mice and rabbits from vaginal and skin challenge with diverse αHPV types. While generally clinically inapparent, human betapapillomaviruses (βHPV) are possibly associated with cutaneous squamous cell carcinoma (CSCC) in epidermodysplasia verruciformis (EV) and immunocompromised patients. Here we show that α11-88x5 vaccination protected wild type and EV model mice against HPV5 challenge. Passive transfer of antiserum conferred protection independently of Fc receptors (FcR) or Gr-1+ phagocytes. Antisera demonstrated robust antibody titers against ten βHPV by L1/L2 VLP ELISA and neutralized and protected against challenge by 3 additional βHPV (HPV49/76/96). Thus, unlike the licensed vaccines, α11-88x5 vaccination elicits broad immunity against αHPV and βHPV.
Collapse
Affiliation(s)
- Pola Olczak
- Department of Pathology, Johns Hopkins University, 1550 Orleans St, Baltimore, MD, 21287, United States
| | - Margaret Wong
- Department of Pathology, Johns Hopkins University, 1550 Orleans St, Baltimore, MD, 21287, United States
| | - Hua-Ling Tsai
- Department of Biostatistics, Johns Hopkins University, 550 N Broadway, Baltimore, MD, 21205, United States
| | - Hao Wang
- Department of Biostatistics, Johns Hopkins University, 550 N Broadway, Baltimore, MD, 21205, United States
| | - Reinhard Kirnbauer
- Department of Dermatology, Medical University of Vienna, 1090, Vienna, Austria
| | - Andrew J Griffith
- Department of Otolaryngology Head-Neck Surgery, College of Medicine-Memphis, University of Tennessee Health Sciences Center, 910 Madison Ave, Memphis, TN, 38163, United States
| | - Paul F Lambert
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, 53705, United States
| | - Richard Roden
- Department of Pathology, Johns Hopkins University, 1550 Orleans St, Baltimore, MD, 21287, United States.
| |
Collapse
|
11
|
Morales CG, Jimenez NR, Herbst-Kralovetz MM, Lee NR. Novel Vaccine Strategies and Factors to Consider in Addressing Health Disparities of HPV Infection and Cervical Cancer Development among Native American Women. Med Sci (Basel) 2022; 10:52. [PMID: 36135837 PMCID: PMC9503187 DOI: 10.3390/medsci10030052] [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] [Received: 07/15/2022] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 12/24/2022] Open
Abstract
Cervical cancer is the 4th most common type of cancer in women world-wide. Many factors play a role in cervical cancer development/progression that include genetics, social behaviors, social determinants of health, and even the microbiome. The prevalence of HPV infections and cervical cancer is high and often understudied among Native American communities. While effective HPV vaccines exist, less than 60% of 13- to 17-year-olds in the general population are up to date on their HPV vaccination as of 2020. Vaccination rates are higher among Native American adolescents, approximately 85% for females and 60% for males in the same age group. Unfortunately, the burden of cervical cancer remains high in many Native American populations. In this paper, we will discuss HPV infection, vaccination and the cervicovaginal microbiome with a Native American perspective. We will also provide insight into new strategies for developing novel methods and therapeutics to prevent HPV infections and limit HPV persistence and progression to cervical cancer in all populations.
Collapse
Affiliation(s)
- Crystal G. Morales
- Department of Biology, Northern Arizona University, Flagstaff, AZ 86011, USA
| | - Nicole R. Jimenez
- Department of Obstetrics and Gynecology, College of Medicine, University of Arizona, Phoenix, AZ 85004, USA
| | - Melissa M. Herbst-Kralovetz
- Department of Obstetrics and Gynecology, College of Medicine, University of Arizona, Phoenix, AZ 85004, USA
- Department of Basic Medical Sciences, College of Medicine, University of Arizona, Phoenix, AZ 85004, USA
| | - Naomi R. Lee
- Department of Chemistry and Biochemistry, Northern Arizona University, Flagstaff, AZ 86011, USA
| |
Collapse
|
12
|
Passive Immunization with a Single Monoclonal Neutralizing Antibody Protects against Cutaneous and Mucosal Mouse Papillomavirus Infections. J Virol 2022; 96:e0070322. [PMID: 35920658 PMCID: PMC9400481 DOI: 10.1128/jvi.00703-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
We have established a mouse papillomavirus (MmuPV1) model that induces both cutaneous and mucosal infections and cancers. In the current study, we use this model to test our hypothesis that passive immunization using a single neutralizing monoclonal antibody can protect both cutaneous and mucosal sites at different time points after viral inoculation. We conducted a series of experiments involving the administration of either a neutralizing monoclonal antibody, MPV.A4, or control monoclonal antibodies to both outbred and inbred athymic mice. Three clinically relevant mucosal sites (lower genital tract for females and anus and tongue for both males and females) and two cutaneous sites (muzzle and tail) were tested. At the termination of the experiments, all tested tissues were harvested for virological analyses. Significantly lower levels of viral signals were detected in the MPV.A4-treated female mice up to 6 h post-viral inoculation compared to those in the isotype control. Interestingly, males displayed partial protection when they received MPV.A4 at the time of viral inoculation, even though they were completely protected when receiving MPV.A4 at 24 h before viral inoculation. We detected MPV.A4 in the blood starting at 1 h and up to 8 weeks postadministration in some mice. Parallel to these in vivo studies, we conducted in vitro neutralization using a mouse keratinocyte cell line and observed complete neutralization up to 8 h post-viral inoculation. Thus, passive immunization with a monoclonal neutralizing antibody can protect against papillomavirus infection at both cutaneous and mucosal sites and is time dependent. IMPORTANCE This is the first study testing a single monoclonal neutralizing antibody (MPV.A4) by passive immunization against papillomavirus infections at both cutaneous and mucosal sites in the same host in the mouse papillomavirus model. We demonstrated that MPV.A4 administered before viral inoculation can protect both male and female athymic mice against MmuPV1 infections at cutaneous and mucosal sites. MPV.A4 also offers partial protection at 6 h post-viral inoculation in female mice. MPV.A4 can be detected in the blood from 1 h to 8 weeks after intraperitoneal (i.p.) injection. Interestingly, males were only partially protected when they received MPV.A4 at the time of viral inoculation. The failed protection in males was due to the absence of neutralizing MPV.A4 at the infected sites. Our findings suggest passive immunization with a single monoclonal neutralizing antibody can protect against diverse papillomavirus infections in a time-dependent manner in mice.
Collapse
|
13
|
Single-dose HPV vaccine immunity: is there a role for non-neutralizing antibodies? Trends Immunol 2022; 43:815-825. [PMID: 35995705 DOI: 10.1016/j.it.2022.07.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 07/28/2022] [Accepted: 07/30/2022] [Indexed: 11/22/2022]
Abstract
A single dose of human papillomavirus (HPV) vaccine against HPV infection (prerequisite for cervical cancer) appears to be as efficacious as two or three doses, despite inducing lower antibody titers. Neutralizing antibodies are thought to be the primary mediator of protection, but the threshold for protection is unknown. Antibody functions beyond neutralization have not been explored for HPV vaccines. Here, we discuss the immune mechanisms of HPV vaccines, with a focus on non-neutralizing antibody effector functions. In the context of single-dose HPV vaccination where antibody is limiting, we propose that non-neutralizing antibody functions may contribute to preventing HPV infection. Understanding the immunological basis of protection for single-dose HPV vaccination will provide a rationale for implementing single-dose HPV vaccine regimens.
Collapse
|
14
|
Olczak P, Matsui K, Wong M, Alvarez J, Lambert P, Christensen ND, Hu J, Huber B, Kirnbauer R, Wang JW, Roden RBS. RG2-VLP: a Vaccine Designed to Broadly Protect against Anogenital and Skin Human Papillomaviruses Causing Human Cancer. J Virol 2022; 96:e0056622. [PMID: 35703545 PMCID: PMC9278150 DOI: 10.1128/jvi.00566-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/20/2022] [Indexed: 12/20/2022] Open
Abstract
The family of human papillomaviruses (HPV) includes over 400 genotypes. Genus α genotypes generally infect the anogenital mucosa, and a subset of these HPV are a necessary, but not sufficient, cause of cervical cancer. Of the 13 high-risk (HR) and 11 intermediate-risk (IR) HPV associated with cervical cancer, genotypes 16 and 18 cause 50% and 20% of cases, respectively, whereas HPV16 dominates in other anogenital and oropharyngeal cancers. A plethora of βHPVs are associated with cutaneous squamous cell carcinoma (CSCC), especially in sun-exposed skin sites of epidermodysplasia verruciformis (EV), AIDS, and immunosuppressed patients. Licensed L1 virus-like particle (VLP) vaccines, such as Gardasil 9, target a subset of αHPV but no βHPV. To comprehensively target both α- and βHPVs, we developed a two-component VLP vaccine, RG2-VLP, in which L2 protective epitopes derived from a conserved αHPV epitope (amino acids 17 to 36 of HPV16 L2) and a consensus βHPV sequence in the same region are displayed within the DE loop of HPV16 and HPV18 L1 VLP, respectively. Unlike vaccination with Gardasil 9, vaccination of wild-type and EV model mice (Tmc6Δ/Δ or Tmc8Δ/Δ) with RG2-VLP induced robust L2-specific antibody titers and protected against β-type HPV5. RG2-VLP protected rabbits against 17 αHPV, including those not covered by Gardasil 9. HPV16- and HPV18-specific neutralizing antibody responses were similar between RG2-VLP- and Gardasil 9-vaccinated animals. However, only transfer of RG2-VLP antiserum effectively protected naive mice from challenge with all βHPVs tested. Taken together, these observations suggest RG2-VLP's potential as a broad-spectrum vaccine to prevent αHPV-driven anogenital, oropharyngeal, and βHPV-associated cutaneous cancers. IMPORTANCE Licensed preventive HPV vaccines are composed of VLPs derived by expression of major capsid protein L1. They confer protection generally restricted to infection by the αHPVs targeted by the up-to-9-valent vaccine, and their associated anogenital cancers and genital warts, but do not target βHPV that are associated with CSCC in EV and immunocompromised patients. We describe the development of a two-antigen vaccine protective in animal models against known oncogenic αHPVs as well as diverse βHPVs by incorporation into HPV16 and HPV18 L1 VLP of 20-amino-acid conserved protective epitopes derived from minor capsid protein L2.
Collapse
Affiliation(s)
- Pola Olczak
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Margaret Wong
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jade Alvarez
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Paul Lambert
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Neil D. Christensen
- The Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University, College of Medicine, Hershey, Pennsylvania, USA
- Department of Pathology, Pennsylvania State University, College of Medicine, Hershey, Pennsylvania, USA
| | - Jiafen Hu
- The Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University, College of Medicine, Hershey, Pennsylvania, USA
- Department of Pathology, Pennsylvania State University, College of Medicine, Hershey, Pennsylvania, USA
| | - Bettina Huber
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Reinhard Kirnbauer
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | | | | |
Collapse
|
15
|
McFall-Boegeman H, Huang X. Mechanisms of cellular and humoral immunity through the lens of VLP-based vaccines. Expert Rev Vaccines 2022; 21:453-469. [PMID: 35023430 PMCID: PMC8960355 DOI: 10.1080/14760584.2022.2029415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Vaccination can be effective defense against many infectious agents and the corresponding diseases. Discoveries elucidating the mechanisms of the immune system have given hopes to developing vaccines against diseases recalcitrant to current treatment/prevention strategies. One such finding is the ability of immunogenic biological nanoparticles to powerfully boost the immunogenicity of poorer antigens conjugated to them with virus-like particle (VLP)-based vaccines as a key example. VLPs take advantage of the well-defined molecular structures associated with sub-unit vaccines and the immunostimulatory nature of conjugate vaccines. AREAS COVERED In this review, we will discuss how advances in understanding the immune system can inform VLP-based vaccine design and how VLP-based vaccines have uncovered underlying mechanisms in the immune system. EXPERT OPINION As our understanding of mechanisms underlying the immune system increases, that knowledge should inform our vaccine design. Testing of proof-of-concept vaccines in the lab should seek to elucidate the underlying mechanisms of immune responses. The integration of these approaches will allow for VLP-based vaccines to live up to their promise as a powerful plug-and-play platform for next generation vaccine development.
Collapse
Affiliation(s)
- Hunter McFall-Boegeman
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, USA.,Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, USA
| | - Xuefei Huang
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, USA.,Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, USA.,Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan 48824, USA
| |
Collapse
|
16
|
Webster E, Seiger KW, Core SB, Collar AL, Knapp-Broas H, Graham J, Shrestha M, Afzaal S, Geisler WM, Wheeler CM, Chackerian B, Frietze KM, Lijek RS. Immunogenicity and Protective Capacity of a Virus-like Particle Vaccine against Chlamydia trachomatis Type 3 Secretion System Tip Protein, CT584. Vaccines (Basel) 2022; 10:vaccines10010111. [PMID: 35062772 PMCID: PMC8779370 DOI: 10.3390/vaccines10010111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/05/2022] [Accepted: 01/07/2022] [Indexed: 10/28/2022] Open
Abstract
An effective vaccine against Chlamydia trachomatis is urgently needed as infection rates continue to rise and C. trachomatis causes reproductive morbidity. An obligate intracellular pathogen, C. trachomatis employs a type 3 secretion system (T3SS) for host cell entry. The tip of the injectosome is composed of the protein CT584, which represents a potential target for neutralization with vaccine-induced antibody. Here, we investigate the immunogenicity and efficacy of a vaccine made of CT584 epitopes coupled to a bacteriophage virus-like particle (VLP), a novel platform for Chlamydia vaccines modeled on the success of HPV vaccines. Female mice were immunized intramuscularly, challenged transcervically with C. trachomatis, and assessed for systemic and local antibody responses and bacterial burden in the upper genital tract. Immunization resulted in a 3-log increase in epitope-specific IgG in serum and uterine homogenates and in the detection of epitope-specific IgG in uterine lavage at low levels. By contrast, sera from women infected with C. trachomatis and virgin controls had similarly low titers to CT584 epitopes, suggesting these epitopes are not systemically immunogenic during natural infection but can be rendered immunogenic by the VLP platform. C. trachomatis burden in the upper genital tract of mice varied after active immunization, yet passive protection was achieved when immune sera were pre-incubated with C. trachomatis prior to inoculation into the genital tract. These data demonstrate the potential for antibody against the T3SS to contribute to protection against C. trachomatis and the value of VLPs as a novel platform for C. trachomatis vaccines.
Collapse
Affiliation(s)
- Everett Webster
- Department of Biological Sciences, Mount Holyoke College, 50 College St., South Hadley, MA 01075, USA; (E.W.); (K.W.S.); (H.K.-B.); (J.G.); (M.S.); (S.A.)
| | - Kyra W. Seiger
- Department of Biological Sciences, Mount Holyoke College, 50 College St., South Hadley, MA 01075, USA; (E.W.); (K.W.S.); (H.K.-B.); (J.G.); (M.S.); (S.A.)
| | - Susan B. Core
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico, MSC 08-4660, 1 University of New Mexico, Albuquerque, NM 87131, USA; (S.B.C.); (A.L.C.); (B.C.); (K.M.F.)
| | - Amanda L. Collar
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico, MSC 08-4660, 1 University of New Mexico, Albuquerque, NM 87131, USA; (S.B.C.); (A.L.C.); (B.C.); (K.M.F.)
| | - Hannah Knapp-Broas
- Department of Biological Sciences, Mount Holyoke College, 50 College St., South Hadley, MA 01075, USA; (E.W.); (K.W.S.); (H.K.-B.); (J.G.); (M.S.); (S.A.)
| | - June Graham
- Department of Biological Sciences, Mount Holyoke College, 50 College St., South Hadley, MA 01075, USA; (E.W.); (K.W.S.); (H.K.-B.); (J.G.); (M.S.); (S.A.)
| | - Muskan Shrestha
- Department of Biological Sciences, Mount Holyoke College, 50 College St., South Hadley, MA 01075, USA; (E.W.); (K.W.S.); (H.K.-B.); (J.G.); (M.S.); (S.A.)
| | - Sarah Afzaal
- Department of Biological Sciences, Mount Holyoke College, 50 College St., South Hadley, MA 01075, USA; (E.W.); (K.W.S.); (H.K.-B.); (J.G.); (M.S.); (S.A.)
| | - William M. Geisler
- Department of Medicine, University of Alabama at Birmingham, 703 19th St. S, ZRB 242, Birmingham, AL 35294, USA;
| | - Cosette M. Wheeler
- Center for HPV Prevention, University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Sciences Center, MSC 08-4640, 1 University of New Mexico, Albuquerque, NM 87131, USA;
| | - Bryce Chackerian
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico, MSC 08-4660, 1 University of New Mexico, Albuquerque, NM 87131, USA; (S.B.C.); (A.L.C.); (B.C.); (K.M.F.)
| | - Kathryn M. Frietze
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico, MSC 08-4660, 1 University of New Mexico, Albuquerque, NM 87131, USA; (S.B.C.); (A.L.C.); (B.C.); (K.M.F.)
- Clinical and Translational Science Center, University of New Mexico Health Sciences, MSC 08-4635, 1 University of New Mexico, Albuquerque, NM 87131, USA
| | - Rebeccah S. Lijek
- Department of Biological Sciences, Mount Holyoke College, 50 College St., South Hadley, MA 01075, USA; (E.W.); (K.W.S.); (H.K.-B.); (J.G.); (M.S.); (S.A.)
- Correspondence: ; Tel.: +1-(413)-538-2487
| |
Collapse
|
17
|
Salinas-Montalvo AM, Supramaniam A, McMillan NA, Idris A. RNA-based gene targeting therapies for human papillomavirus driven cancers. Cancer Lett 2021; 523:111-120. [PMID: 34627949 DOI: 10.1016/j.canlet.2021.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/03/2021] [Accepted: 10/05/2021] [Indexed: 12/22/2022]
Abstract
While platinum-based chemotherapy, radiation therapy and or surgery are effective in reducing human papillomavirus (HPV) driven cancer tumours, they have some significant drawbacks, including low specificity for tumour, toxicity, and severe adverse effects. Though current therapies for HPV-driven cancers are effective, severe late toxicity associated with current treatments contributes to the deterioration of patient quality of life. This warrants the need for novel therapies for HPV derived cancers. In this short review, we examined RNA-based therapies targeting the major HPV oncogenes, including short-interfering RNAs (siRNAs) and clustered regularly interspaced short palindromic repeats (CRISPR) as putative treatment modalities. We also explore other potential RNA-based targeting approaches such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and mRNA vaccines as future treatment modalities for HPV cancers. Some of these technologies have already been approved for clinical use for a range of other human diseases but not for HPV cancers. Here we explore the emerging evidence supporting the effectiveness of some of these gene-based therapies for HPV malignancies. In short, the evidence sheds promising light on the feasibility of translating these technologies into a clinically relevant treatment modality for HPV derived cancers and potentially other virally driven human cancers.
Collapse
Affiliation(s)
- Ana María Salinas-Montalvo
- Menzies Health Institute Queensland and School of Medical Sciences, Griffith University, Gold Coast, QLD, Australia
| | - Aroon Supramaniam
- Menzies Health Institute Queensland and School of Medical Sciences, Griffith University, Gold Coast, QLD, Australia
| | - Nigel Aj McMillan
- Menzies Health Institute Queensland and School of Medical Sciences, Griffith University, Gold Coast, QLD, Australia
| | - Adi Idris
- Menzies Health Institute Queensland and School of Medical Sciences, Griffith University, Gold Coast, QLD, Australia.
| |
Collapse
|
18
|
Basu P, Malvi SG, Joshi S, Bhatla N, Muwonge R, Lucas E, Verma Y, Esmy PO, Poli URR, Shah A, Zomawia E, Pimple S, Jayant K, Hingmire S, Chiwate A, Divate U, Vashist S, Mishra G, Jadhav R, Siddiqi M, Sankaran S, Prabhu PR, Kannan TPRA, Varghese R, Shastri SS, Anantharaman D, Gheit T, Tommasino M, Sauvaget C, Pillai MR, Sankaranarayanan R. Vaccine efficacy against persistent human papillomavirus (HPV) 16/18 infection at 10 years after one, two, and three doses of quadrivalent HPV vaccine in girls in India: a multicentre, prospective, cohort study. Lancet Oncol 2021; 22:1518-1529. [PMID: 34634254 PMCID: PMC8560643 DOI: 10.1016/s1470-2045(21)00453-8] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/16/2021] [Accepted: 07/26/2021] [Indexed: 12/02/2022]
Abstract
BACKGROUND A randomised trial designed to compare three and two doses of quadrivalent human papillomavirus (HPV) vaccine in adolescent girls in India was converted to a cohort study after suspension of HPV vaccination in trials by the Indian Government. In this Article, the revised aim of the cohort study was to compare vaccine efficacy of single dose to that of three and two doses in protecting against persistent HPV 16 and 18 infection at 10 years post vaccination. METHODS In the randomised trial, unmarried girls aged 10-18 years were recruited from nine centres across India and randomly assigned to either two doses or three doses of the quadrivalent HPV vaccine (Gardasil [Merck Sharp & Dohme, Whitehouse Station, NJ, USA]; 0·5 mL administered intramuscularly). After suspension of recruitment and vaccination, the study became a longitudinal, prospective cohort study by default, and participants were allocated to four cohorts on the basis of the number vaccine doses received per protocol: the two-dose cohort (received vaccine on days 1 and 180 or later), three-dose cohort (days 1, 60, and 180 or later), two-dose default cohort (days 1 and 60 or later), and the single-dose default cohort. Participants were followed up yearly. Cervical specimens were collected from participants 18 months after marriage or 6 months after first childbirth, whichever was earlier, to assess incident and persistent HPV infections. Married participants were screened for cervical cancer as they reached 25 years of age. Unvaccinated women age-matched to the married vaccinated participants were recruited to serve as controls. Vaccine efficacy against persistent HPV 16 and 18 infections (the primary endpoint) was analysed for single-dose recipients and compared with that in two-dose and three-dose recipients after adjusting for imbalance in the distribution of potential confounders between the unvaccinated and vaccinated cohorts. This trial is registered with ISRCTN, ISRCTN98283094, and ClinicalTrials.gov, NCT00923702. FINDINGS Vaccinated participants were recruited between Sept 1, 2009, and April 8, 2010 (date of vaccination suspension), and followed up over a median duration of 9·0 years (IQR 8·2-9·6). 4348 participants had three doses, 4980 had two doses (0 and 6 months), and 4949 had a single dose. Vaccine efficacy against persistent HPV 16 and 18 infection among participants evaluable for the endpoint was 95·4% (95% CI 85·0-99·9) in the single-dose default cohort (2135 women assessed), 93·1% (77·3-99·8) in the two-dose cohort (1452 women assessed), and 93·3% (77·5-99·7) in three-dose recipients (1460 women assessed). INTERPRETATION A single dose of HPV vaccine provides similar protection against persistent infection from HPV 16 and 18, the genotypes responsible for nearly 70% of cervical cancers, to that provided by two or three doses. FUNDING Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Partha Basu
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, Lyon, France.
| | - Sylla G Malvi
- Tata Memorial Centre Rural Cancer Project, Nargis Dutt Memorial Cancer Hospital, Barshi District Solapur, Maharashtra, India
| | - Smita Joshi
- Jehangir Clinical Development Centre, Jehangir Hospital Premises, Pune, India
| | - Neerja Bhatla
- Department of Obstetrics and Gynaecology, All India Institute of Medical Sciences, New Delhi, India
| | - Richard Muwonge
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, Lyon, France
| | - Eric Lucas
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, Lyon, France
| | - Yogesh Verma
- Sikkim Manipal Institute of Medical Sciences, Sikkim Manipal University, Gangtok, Sikkim, India
| | - Pulikkottil O Esmy
- Christian Fellowship Community Health Centre, Ambillikai, Dindigul District, Tamil Nadu, India
| | | | - Anand Shah
- Department of Community Oncology, Gujarat Cancer and Research Institute, M P Shah Cancer Hospital, Civil Hospital Campus, Asarwa, Ahmedabad, India
| | | | - Sharmila Pimple
- Department of Preventive Oncology, Centre for Cancer Epidemiology, Tata Memorial Centre, Homi Bhabha National Institute, Parel, Mumbai, India
| | - Kasturi Jayant
- Tata Memorial Centre Rural Cancer Project, Nargis Dutt Memorial Cancer Hospital, Barshi District Solapur, Maharashtra, India
| | - Sanjay Hingmire
- Tata Memorial Centre Rural Cancer Project, Nargis Dutt Memorial Cancer Hospital, Barshi District Solapur, Maharashtra, India
| | - Aruna Chiwate
- Tata Memorial Centre Rural Cancer Project, Nargis Dutt Memorial Cancer Hospital, Barshi District Solapur, Maharashtra, India
| | - Uma Divate
- Jehangir Clinical Development Centre, Jehangir Hospital Premises, Pune, India
| | - Shachi Vashist
- Department of Obstetrics and Gynaecology, All India Institute of Medical Sciences, New Delhi, India
| | - Gauravi Mishra
- Department of Preventive Oncology, Centre for Cancer Epidemiology, Tata Memorial Centre, Homi Bhabha National Institute, Parel, Mumbai, India
| | - Radhika Jadhav
- Jehangir Clinical Development Centre, Jehangir Hospital Premises, Pune, India
| | | | - Subha Sankaran
- Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram, Kerala, India
| | - Priya Ramesh Prabhu
- Human Biology Division, Fred Hutchinson Cancer Research Centre, Seattle, WA, USA
| | | | - Rintu Varghese
- Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram, Kerala, India
| | - Surendra S Shastri
- Department of Health Disparities Research, Division of Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Devasena Anantharaman
- Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram, Kerala, India
| | - Tarik Gheit
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, Lyon, France
| | - Massimo Tommasino
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, Lyon, France
| | - Catherine Sauvaget
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, Lyon, France
| | - M Radhakrishna Pillai
- Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram, Kerala, India
| | | |
Collapse
|
19
|
Saied AA, Metwally AA, Mohamed HMA, Haridy MAM. The contribution of bovines to human health against viral infections. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:46999-47023. [PMID: 34272669 PMCID: PMC8284698 DOI: 10.1007/s11356-021-14941-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/12/2021] [Indexed: 04/12/2023]
Abstract
In the last 40 years, novel viruses have evolved at a much faster pace than other pathogens. Viral diseases pose a significant threat to public health around the world. Bovines have a longstanding history of significant contributions to human nutrition, agricultural, industrial purposes, medical research, drug and vaccine development, and livelihood. The life cycle, genomic structures, viral proteins, and pathophysiology of bovine viruses studied in vitro paved the way for understanding the human counterparts. Calf model has been used for testing vaccines against RSV, papillomavirus vaccines and anti-HCV agents were principally developed after using the BPV and BVDV model, respectively. Some bovine viruses-based vaccines (BPIV-3 and bovine rotaviruses) were successfully developed, clinically tried, and commercially produced. Cows, immunized with HIV envelope glycoprotein, produced effective broadly neutralizing antibodies in their serum and colostrum against HIV. Here, we have summarized a few examples of human viral infections for which the use of bovines has contributed to the acquisition of new knowledge to improve human health against viral infections covering the convergence between some human and bovine viruses and using bovines as disease models. Additionally, the production of vaccines and drugs, bovine-based products were covered, and the precautions in dealing with bovines and bovine-based materials.
Collapse
Affiliation(s)
- AbdulRahman A Saied
- Department of Food Establishments Licensing (Aswan Branch), National Food Safety Authority (NFSA), Aswan, 81511, Egypt.
- Touristic Activities and Interior Offices Sector (Aswan Office), Ministry of Tourism and Antiquities, Aswan, 81511, Egypt.
| | - Asmaa A Metwally
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Aswan University, Aswan, 81511, Egypt
| | - Hams M A Mohamed
- Department of Microbiology, Faculty of Veterinary Medicine, South Valley University, Qena, 83523, Egypt
| | - Mohie A M Haridy
- Department of Pathology and Clinical Pathology, Faculty of Veterinary Medicine, South Valley University, Qena, 83523, Egypt.
| |
Collapse
|
20
|
Hoes J, Pasmans H, Schurink-van 't Klooster TM, van der Klis FRM, Donken R, Berkhof J, de Melker HE. Review of long-term immunogenicity following HPV vaccination: Gaps in current knowledge. Hum Vaccin Immunother 2021; 18:1908059. [PMID: 34033518 PMCID: PMC8920133 DOI: 10.1080/21645515.2021.1908059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The licensed HPV vaccines are highly efficacious and induce high levels of neutralizing antibody levels, the assumed mediators of protection. However, a correlate of protection against HPV is lacking, and the evidence is still limited as to long-term persistence of antibodies, especially following reduced dosing schedules. The World Health Organization (WHO) urges immunization of young girls as part of the strategy to eliminate cervical cancer, thus long-lasting protection is required. The current review describes long-term follow-up regarding vaccine-induced seropositivity and antibody level development following the different vaccines and dosing schedules. Implications and opportunities of long-term vaccine-induced immune responses are discussed, such as the gaps in monitoring of long-term immunogenicity, the possibilities of reduced dosing schedules, and the importance of evidence for durable immunity.
Collapse
Affiliation(s)
- J Hoes
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands.,Department of Epidemiology and Data Science, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - H Pasmans
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - T M Schurink-van 't Klooster
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - F R M van der Klis
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - R Donken
- Department of Epidemiology and Data Science, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - J Berkhof
- Department of Epidemiology and Data Science, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - H E de Melker
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| |
Collapse
|
21
|
Kines RC, Thompson CD, Spring S, Li Z, de Los Pinos E, Monks S, Schiller JT. Virus-Like Particle-Drug Conjugates Induce Protective, Long-lasting Adaptive Antitumor Immunity in the Absence of Specifically Targeted Tumor Antigens. Cancer Immunol Res 2021; 9:693-706. [PMID: 33853825 DOI: 10.1158/2326-6066.cir-19-0974] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 12/24/2020] [Accepted: 04/07/2021] [Indexed: 12/30/2022]
Abstract
This study examined the ability of a papillomavirus-like particle drug conjugate, belzupacap sarotalocan (AU-011), to eradicate subcutaneous tumors after intravenous injection and to subsequently elicit long-term antitumor immunity in the TC-1 syngeneic murine tumor model. Upon in vitro activation with near-infrared light (NIR), AU-011-mediated cell killing was proimmunogenic in nature, resulting in the release of damage-associated molecular patterns such as DNA, ATP, and HMGB-1, activation of caspase-1, and surface relocalization of calreticulin and HSP70 on killed tumor cells. A single in vivo administration of AU-011 followed by NIR caused rapid cell death, leading to long-term tumor regression in ∼50% of all animals. Within hours of treatment, calreticulin surface expression, caspase-1 activation, and depletion of immunosuppressive leukocytes were observed in tumors. Combination of AU-011 with immune-checkpoint inhibitor antibodies, anti-CTLA-4 or anti-PD-1, improved therapeutic efficacy, resulting in 70% to 100% complete response rate that was durable 100 days after treatment, with 50% to 80% of those animals displaying protection from secondary tumor rechallenge. Depletion of CD4+ or CD8+ T cells, either at the time of AU-011 treatment or secondary tumor rechallenge of tumor-free mice, indicated that both cell populations are vital to AU-011's ability to eradicate primary tumors and induce long-lasting antitumor protection. Tumor-specific CD8+ T-cell responses could be observed in circulating peripheral blood mononuclear cells within 3 weeks of AU-011 treatment. These data, taken together, support the conclusion that AU-011 has a direct cytotoxic effect on tumor cells and induces long-term antitumor immunity, and this activity is enhanced when combined with checkpoint inhibitor antibodies.
Collapse
Affiliation(s)
| | - Cynthia D Thompson
- Laboratory of Cellular Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Zhenyu Li
- Aura Biosciences, Cambridge, Massachusetts
| | | | | | - John T Schiller
- Laboratory of Cellular Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
22
|
Abstract
In persistent high-risk HPV infection, viral gene expression can trigger some important early changes to immune capabilities which act to protect the lesion from immune attack and subsequently promote its growth and ability for sustained immune escape. This includes immune checkpoint-inhibitor ligand expression (e.g. PD-L1) by tumour or associated immune cells that can block any anti-tumour T-cell effectors. While there are encouraging signs of efficacy for cancer immunotherapies including with immune checkpoint inhibitors, therapeutic vaccines and adoptive cell therapies, overall response and survival rates remain relatively low. HPV oncogene vaccination has shown some useful efficacy in treatment of patients with high-grade lesions but was unable to control later stage cancers. To maximally exploit anti-tumour immune responses, the suppressive factors associated with HPV carcinogenesis must be countered. Importantly, a combination of chemotherapy, reducing immunosuppressive myeloid cells, with therapeutic HPV vaccination significantly improves impact on cancer treatment. Many clinical trials are investigating checkpoint inhibitor treatments in HPV associated cancers but response rates are limited; combination with vaccination is being tested. Further investigation of how chemo- and/or radio-therapy can influence the recovery of effective anti-tumour immunity is warranted. Understanding how to optimally deploy and sequence conventional and immunotherapies is the challenge.
Collapse
|
23
|
Affiliation(s)
- Dennis R Burton
- The Scripps Research Institute, La Jolla, CA, USA. .,Ragon Institute of MGH, Harvard and MIT, Cambridge, MA, USA.
| | - Eric J Topol
- The Scripps Research Institute, La Jolla, CA, USA. .,Scripps Translational Research Institute, La Jolla, CA, USA.
| |
Collapse
|
24
|
Cross-neutralizing antibody titres against non-vaccine types induced by a recombinant trivalent HPV vaccine (16/18/58) in rhesus macaques. PAPILLOMAVIRUS RESEARCH 2020; 10:100209. [PMID: 33197649 PMCID: PMC7704424 DOI: 10.1016/j.pvr.2020.100209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 12/05/2022]
Abstract
Human papillomavirus (HPV) causes not only most cervical cancers but also cancers of the vagina, vulva, penis, anus, rectum, and oropharynx. Every year, 200,000 women die of cervical cancer in the world, and China accounts for about 10%. HPV vaccines are effective in preventing HPV infections thus HPV-related cancers worldwide. Studies on the clinical trials of the 2v Cervarix™ and the 4v Gardasil® have suggested that immunization with either of these vaccines provided some level of protection against other HPV types that are closely related to the types contained in the vaccines. Here we conducted a preliminary evaluation on the ability to induce cross-neutralizing antibodies in rhesus monkeys by a 3v HPV vaccine that targets HPV16, 18, and 58 and it is specifically designed for Chinese women. We found that this vaccine is no less than Gardasil® in terms of the ability to induce NAbs against non-vaccine types of HPV in rhesus macaques. These results provided evidence from the immunogenicity point of view that the KLWS 3v HPV vaccine is a strong competitor to the imported 2v and 4v HPV vaccines currently available on the market.
Collapse
|
25
|
Progress in L2-Based Prophylactic Vaccine Development for Protection against Diverse Human Papillomavirus Genotypes and Associated Diseases. Vaccines (Basel) 2020; 8:vaccines8040568. [PMID: 33019516 PMCID: PMC7712070 DOI: 10.3390/vaccines8040568] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022] Open
Abstract
The human papillomaviruses (HPVs) are a family of small DNA tumor viruses including over 200 genotypes classified by phylogeny into several genera. Different genera of HPVs cause ano-genital and oropharyngeal cancers, skin cancers, as well as benign diseases including skin and genital warts. Licensed vaccines composed of L1 virus-like particles (VLPs) confer protection generally restricted to the ≤9 HPV types targeted. Here, we examine approaches aimed at broadening the protection against diverse HPV types by targeting conserved epitopes of the minor capsid protein, L2. Compared to L1 VLP, L2 is less immunogenic. However, with appropriate presentation to the immune system, L2 can elicit durable, broadly cross-neutralizing antibody responses and protection against skin and genital challenge with diverse HPV types. Such approaches to enhance the strength and breadth of the humoral response include the display of L2 peptides on VLPs or viral capsids, bacteria, thioredoxin and other platforms for multimerization. Neither L2 nor L1 vaccinations elicit a therapeutic response. However, fusion of L2 with early viral antigens has the potential to elicit both prophylactic and therapeutic immunity. This review of cross-protective HPV vaccines based on L2 is timely as several candidates have recently entered early-phase clinical trials.
Collapse
|
26
|
Garcea RL, Meinerz NM, Dong M, Funke H, Ghazvini S, Randolph TW. Single-administration, thermostable human papillomavirus vaccines prepared with atomic layer deposition technology. NPJ Vaccines 2020; 5:45. [PMID: 32528733 PMCID: PMC7265342 DOI: 10.1038/s41541-020-0195-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 05/12/2020] [Indexed: 01/05/2023] Open
Abstract
Cold-chain requirements affect worldwide distribution of many vaccines. In addition, vaccines requiring multiple doses impose logistical and financial burdens, as well as patient compliance barriers. To address such limitations, we have developed new technologies to prepare thermostable, single-shot, prime-boost microparticle vaccines. Antigen/adjuvant formulations containing glass-forming polymers and trehalose first are spray-dried to form glassy microparticles that confer thermostability. Atomic layer deposition (ALD) reactions conducted in fluidized beds are then used to coat the microparticles with defined numbers of molecular layers of alumina that modulate the timed release of the internalized antigen and act as adjuvants. We have used a model HPV16 L1 capsomere antigen to evaluate the properties of these technologies. Thermostabilized powders containing HPV16 L1 capsomeres were prepared by spray-drying, coated by ALD with up to 500 molecular layers of alumina, and injected into mice. Antigen distribution was assessed by live-animal IR dye tracking of injected labeled antigen. Antibody responses were measured weekly by ELISA, and neutralizing antibodies were measured by pseudovirus neutralization assays at selected time points. Thermostability was evaluated by measuring antibody responses after incubating ALD-coated antigen powders for one month at 50 °C. Single doses of the ALD-coated vaccine formulations elicited a prime-boost immune response, and produced neutralizing responses and antibody titers that were equivalent or superior to conventional prime-boost doses of liquid formulations. Antibody titers were unaffected by month-long incubation of the formulations at 50 °C. Single-dose, thermostable antigen preparations may overcome current limitations in HPV vaccine delivery as well as being widely applicable to other antigens.
Collapse
Affiliation(s)
- Robert L. Garcea
- The BioFrontiers Program, University of Colorado, Boulder, CO USA
- Department of Molecular, Cellular, Developmental Biology, University of Colorado, Boulder, CO USA
| | - Natalie M. Meinerz
- The BioFrontiers Program, University of Colorado, Boulder, CO USA
- Department of Molecular, Cellular, Developmental Biology, University of Colorado, Boulder, CO USA
| | - Miao Dong
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO USA
| | - Hans Funke
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO USA
| | - Saba Ghazvini
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO USA
| | - Theodore W. Randolph
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO USA
| |
Collapse
|
27
|
Hepatitis B Vaccination Induces Mucosal Antibody Responses in the Female Genital Tract, Indicating Potential Mechanisms of Protection Against Infection. Sex Transm Dis 2020; 46:e53-e56. [PMID: 30444798 DOI: 10.1097/olq.0000000000000949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Vaccines against hepatitis B virus confer effective protection. Enzyme-linked immunosorbent assay was developed to test for specific antibodies in female genital tract secretions. Anti-hepatitis B IgG and IgA were detected in the cervicovaginal secretions of women after hepatitis B vaccination, indicating a potential genital tract role for neutralizing antibodies against sexually transmitted hepatitis B virus.
Collapse
|
28
|
In RRP, serologic response to HPV is frequently absent and slow to develop. PLoS One 2020; 15:e0230106. [PMID: 32160246 PMCID: PMC7065799 DOI: 10.1371/journal.pone.0230106] [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: 02/18/2019] [Accepted: 02/23/2020] [Indexed: 01/08/2023] Open
Abstract
Background Recurrent respiratory papillomatosis (RRP) is characterized by repeated formation of papillomas in the respiratory tract and is caused by human papillomavirus (HPV) types 6 and 11. Women with genital HPV infection are slow to develop weak humoral immunity, but respond robustly to the HPV vaccine. We wondered if people with RRP had a similar immune response. Methods A convenience cross-sectional sample of patients with RRP were recruited into one of four groups: 1) adults and adolescents with active RRP, 2) children with active RRP, 3) RRP patients who had undergone HPV vaccination prior to enrollment and, 4) people with RRP who were in remission. Anti-HPV6 and HPV11 serology was determined by cLIA on a single blood draw. Results Of the 70 subjects enrolled, 36, 16, 8, and 10, were in groups 1, 2, 3, and 4, respectively. 47% of participants aged >11 years and 81% aged ≤11 years possessed no antibodies against HPV6 or HPV11 (ie. double seronegative). 61% of patients in remission were double seronegative. All participants who had received HPV vaccine previously were seropositive to at least one of these low risk HPV types (ie none of them were double seronegative). Among patients who had active RRP and never had HPV vaccination (n = 52) there was an association between duration of symptoms and seropositivity. Of those who were seropositive, the geometric mean duration of symptoms was 11 years compared to 4.7 years for those who were seronegative (p = 0.001). Conclusion People with RRP are capable of developing a humoral response to HPV6 and HPV11. That response appears to be robust when initiated by the HPV vaccine, but either nonexistent or slow to develop in response to infection. Most in remission do not have demonstrable antibody levels against HPV6 or HPV11.
Collapse
|
29
|
Smalley Rumfield C, Roller N, Pellom ST, Schlom J, Jochems C. Therapeutic Vaccines for HPV-Associated Malignancies. Immunotargets Ther 2020; 9:167-200. [PMID: 33117742 PMCID: PMC7549137 DOI: 10.2147/itt.s273327] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/03/2020] [Indexed: 12/13/2022] Open
Abstract
Human papillomavirus (HPV)-related malignancies are responsible for almost all cases of cervical cancer in women, and over 50% of all cases of head and neck carcinoma. Worldwide, HPV-positive malignancies account for 4.5% of the global cancer burden, or over 600,000 cases per year. HPV infection is a pressing public health issue, as more than 80% of all individuals have been exposed to HPV by age 50, representing an important target for vaccine development to reduce the incidence of cancer and the economic cost of HPV-related health issues. The approval of Gardasil® as a prophylactic vaccine for high-risk HPV 16 and 18 and low-risk HPV6 and 11 for people aged 11-26 in 2006, and of Cervarix® in 2009, revolutionized the field and has since reduced HPV infection in young populations. Unfortunately, prophylactic vaccination does not induce immunity in those with established HPV infections or HPV-induced neoplasms, and there are currently no therapeutic HPV vaccines approved by the US Food and Drug Administration. This comprehensive review will detail the progress made in the development of therapeutic vaccines against high-risk HPV types, and potential combinations with other immunotherapeutic agents for more efficient and rational designs of combination treatments for HPV-associated malignancies.
Collapse
Affiliation(s)
- Claire Smalley Rumfield
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nicholas Roller
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Samuel Troy Pellom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Correspondence: Jeffrey Schlom Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, 10 Center Drive, Room 8B09, Bethesda, MD20892, USATel +1 240-858-3463Fax +1 240-541-4558 Email
| | - Caroline Jochems
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
30
|
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
|
31
|
Plotkin SA. Updates on immunologic correlates of vaccine-induced protection. Vaccine 2019; 38:2250-2257. [PMID: 31767462 DOI: 10.1016/j.vaccine.2019.10.046] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/15/2019] [Accepted: 10/17/2019] [Indexed: 02/06/2023]
Abstract
Correlates of protection (CoPs) are increasingly important in the development and licensure of vaccines. Although the study of CoPs was initially directed at identifying a single immune function that could explain vaccine efficacy, it has become increasingly clear that there are often multiple functions responsible for efficacy. This review is meant to supplement prior articles on the subject, illustrating both simple and complex CoPs.
Collapse
Affiliation(s)
- Stanley A Plotkin
- Emeritus Professor of Pediatrics, University of Pennsylvania, Vaxconsult, 4650 Wismer Rd., Doylestown, PA 18902, United States.
| |
Collapse
|
32
|
Abstract
Antibody neutralization of a virus in vitro is often associated with protection against viral exposure in vivo, but the mechanisms operational in vivo are often unclear. By investigating a large number of antibodies, Earnest et al. (https://doi.org/10.1084/jem.20190736) show the importance of antibody effector function in neutralizing antibody protection against an emerging alphavirus in a mouse model.
Collapse
Affiliation(s)
- Dennis R Burton
- The Scripps Research Institute, La Jolla, CA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA
| |
Collapse
|
33
|
Human Papillomavirus 16 Capsids Mediate Nuclear Entry during Infection. J Virol 2019; 93:JVI.00454-19. [PMID: 31092566 DOI: 10.1128/jvi.00454-19] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 05/04/2019] [Indexed: 12/27/2022] Open
Abstract
Infectious human papillomavirus 16 (HPV16) L1/L2 pseudovirions were found to remain largely intact during vesicular transport to the nucleus. By electron microscopy, capsids with a diameter of 50 nm were clearly visible within small vesicles attached to mitotic chromosomes and to a lesser extent within interphase nuclei, implying nuclear disassembly. By confocal analysis, it was determined that nuclear entry of assembled L1 is dependent upon the presence of the minor capsid protein, L2, but independent of encapsidated DNA. We also demonstrate that L1 nuclear localization and mitotic chromosome association can occur in vivo in the murine cervicovaginal challenge model of HPV16 infection. These findings challenge the prevailing concepts of PV uncoating and disassembly. More generally, they document that a largely intact viral capsid can enter the nucleus within a transport vesicle, establishing a novel mechanism by which a virus accesses the nuclear cellular machinery.IMPORTANCE Papillomaviruses (PVs) comprise a large family of nonenveloped DNA viruses that include HPV16, among other oncogenic types, the causative agents of cervical cancer. Delivery of the viral DNA into the host cell nucleus is necessary for establishment of infection. This was thought to occur via a subviral complex following uncoating of the larger viral capsid. In this study, we demonstrate that little disassembly of the PV capsid occurs prior to nuclear delivery. These surprising data reveal a previously unrecognized viral strategy to access the nuclear replication machinery. Understanding viral entry mechanisms not only increases our appreciation of basic cell biological pathways but also may lead to more effective antiviral interventions.
Collapse
|
34
|
Zhu FC, Hu SY, Hong Y, Hu YM, Zhang X, Zhang YJ, Pan QJ, Zhang WH, Zhao FH, Zhang CF, Yang X, Yu JX, Zhu J, Zhu Y, Chen F, Zhang Q, Wang H, Wang C, Bi J, Xue S, Shen L, Zhang YS, He Y, Tang H, Karkada N, Suryakiran P, Bi D, Struyf F. Efficacy, immunogenicity and safety of the AS04-HPV-16/18 vaccine in Chinese women aged 18-25 years: End-of-study results from a phase II/III, randomised, controlled trial. Cancer Med 2019; 8:6195-6211. [PMID: 31305011 PMCID: PMC6797633 DOI: 10.1002/cam4.2399] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 06/18/2019] [Accepted: 06/20/2019] [Indexed: 12/20/2022] Open
Abstract
Background Cervical cancer is a major public health concern in China. We report the end‐of‐study results of a phase II/III trial to assess the efficacy, immunogenicity, and safety of the AS04‐human papillomavirus (HPV)‐16/18 vaccine in Chinese women aged 18‐25 years followed for up to 72 months after first vaccination. Results of approximately 57 months following first vaccination have been previously reported. Methods Healthy 18‐25‐year‐old women (N = 6051) were randomized (1:1) to receive three doses of AS04‐HPV‐16/18 vaccine or Al(OH)3 (control) at Months 0‐1‐6. Vaccine efficacy against HPV‐16/18 infection and cervical intraepithelial neoplasia (CIN), cross‐protective vaccine efficacy against infections and lesions associated with nonvaccine oncogenic HPV types, immunogenicity, and safety were assessed. Efficacy was assessed in the according‐to‐protocol efficacy (ATP‐E) cohort (vaccine N = 2888; control N = 2892), total vaccinated cohort for efficacy (TVC‐E; vaccine N = 2987; control N = 2985) and TVC‐naïve (vaccine N = 1660; control N = 1587). Results In initially HPV‐16/18 seronegative/DNA‐negative women, vaccine efficacy against HPV‐16/18‐associated CIN grade 2 or worse was 87.3% (95% CI: 5.5, 99.7) in the ATP‐E, 88.7% (95% CI: 18.5, 99.7) in the TVC‐E, and 100% (95% CI: 17.9, 100) in the TVC‐naïve. Cross‐protective efficacy against incident infection with HPV‐31, HPV‐33 and HPV‐45 was 59.6% (95% CI: 39.4, 73.5), 42.7% (95% CI: 15.6, 61.6), and 54.8% (95% CI: 19.3, 75.6), respectively (ATP‐E). At Month 72, >95% of initially seronegative women who received HPV vaccine in the ATP cohort for immunogenicity (N = 664) remained seropositive for anti‐HPV‐16/18 antibodies; anti‐HPV‐16 and anti‐HPV‐18 geometric mean titers were 678.1 EU/mL (95% CI: 552.9, 831.5) and 343.7 EU/mL (95% CI: 291.9, 404.8), respectively. Serious adverse events were infrequent (1.9% vaccine group [N = 3026]; 2.7% control group [N = 3025]). Three and zero women died in the control group and the vaccine group respectively. New onset autoimmune disease was reported in two women in the vaccine group and two in the control group. Conclusions This is the first large‐scale randomized clinical trial of HPV vaccination in China. High and sustained vaccine efficacy against HPV‐16/18‐associated infection and cervical lesions was demonstrated up to Month 72. The vaccine had an acceptable safety profile. Combined with screening, prophylactic HPV vaccination could potentially reduce the high burden of HPV infection and cervical cancer in China. Trial registration NCT00779766.
Collapse
Affiliation(s)
- Feng-Cai Zhu
- Jiangsu Province Center for Disease Prevention and Control, Nanjing, China
| | - Shang-Ying Hu
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Ying Hong
- Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yue-Mei Hu
- Jiangsu Province Center for Disease Prevention and Control, Nanjing, China
| | - Xun Zhang
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Yi-Ju Zhang
- Jiangsu Province Center for Disease Prevention and Control, Nanjing, China
| | - Qin-Jing Pan
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Wen-Hua Zhang
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Fang-Hui Zhao
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Cheng-Fu Zhang
- Lianshui Center for Disease Prevention and Control, Lianshui, China
| | - Xiaoping Yang
- Jintan Center for Disease Prevention and Control, Jintan, China
| | - Jia-Xi Yu
- Xuzhou Center for Disease Prevention and Control, Xuzhou, China
| | - Jiahong Zhu
- Lianshui Center for Disease Prevention and Control, Lianshui, China
| | - Yejiang Zhu
- Binhai Center for Disease Prevention and Control, Yancheng, China
| | - Feng Chen
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Qian Zhang
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Hong Wang
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Changrong Wang
- Jintan Center for Disease Prevention and Control, Jintan, China
| | - Jun Bi
- Xuzhou Center for Disease Prevention and Control, Xuzhou, China
| | - Shiyin Xue
- Lianshui Center for Disease Prevention and Control, Lianshui, China
| | - Lingling Shen
- Xuzhou Center for Disease Prevention and Control, Xuzhou, China
| | - Yan-Shu Zhang
- Binhai Center for Disease Prevention and Control, Yancheng, China
| | | | | | | | | | | | | |
Collapse
|
35
|
Thomson NA, Howe L, Weidgraaf K, Thomas DG, Young V, Ward VK, Munday JS. Felis catus papillomavirus type 2 virus-like particle vaccine is safe and immunogenic but does not reduce FcaPV-2 viral loads in adult cats. Vet Immunol Immunopathol 2019; 213:109888. [PMID: 31307673 DOI: 10.1016/j.vetimm.2019.109888] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/04/2019] [Accepted: 06/26/2019] [Indexed: 02/03/2023]
Abstract
Felis catus papillomavirus type 2 (FcaPV-2) commonly infects the skin of domestic cats and has been associated with the development of skin cancer. In the present study, a FcaPV-2 virus-like particle (VLP) vaccine was produced and assessed for vaccine safety, immunogenicity, and impact on FcaPV-2 viral load. This is the first report of the use of a papillomavirus VLP vaccine in domestic cats. The FcaPV-2 VLP vaccine was given to ten adult cats that were naturally infected with FcaPV-2, and a further ten naturally infected cats were sham vaccinated as a control group. The rationale for vaccinating cats already infected with the virus was to induce neutralizing antibody titers that could prevent reinfection of new areas of skin and reduce the overall viral load, as has been demonstrated in other species. Reducing the overall FcaPV-2 viral load could reduce the risk for subsequent PV-associated cancer. The vaccine in this study was well-tolerated, as none of the cats developed any signs of local reaction or systemic illness. In the treatment group, the geometric mean anti-papillomavirus endpoint antibody titers increased significantly following vaccination from 606 (95% CI 192-1913) to 4223 (2023-8814), a 7.0-fold increase, although the individual antibody response varied depending on the level of pre-existing antibodies. Despite the immunogenicity of the vaccine, there was no significant change in FcaPV-2 viral load in the treatment group compared to the control group, over the 24 week follow-up period. A possible reason is that FcaPV-2 was already widespread in the basal skin layer of these adult cats and so preventing further cells from becoming infected had no impact on the overall viral load. Therefore, these results do not support the use of a FcaPV-2 VLP vaccine to reduce the risk for PV-associated cancer in cats in which FcaPV-2 infection is already well established. However, these results justify future studies in which the vaccine is administered to younger cats prior to FcaPV-2 infection becoming fully established.
Collapse
Affiliation(s)
- Neroli A Thomson
- School of Veterinary Science, Massey University, Palmerston North 4472, New Zealand.
| | - Laryssa Howe
- School of Veterinary Science, Massey University, Palmerston North 4472, New Zealand
| | - Karin Weidgraaf
- School of Agriculture and Environment, Massey University, Palmerston North 4472, New Zealand
| | - David G Thomas
- School of Agriculture and Environment, Massey University, Palmerston North 4472, New Zealand
| | - Vivienne Young
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
| | - Vernon K Ward
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
| | - John S Munday
- School of Veterinary Science, Massey University, Palmerston North 4472, New Zealand
| |
Collapse
|
36
|
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: 1.8] [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
|
37
|
King E, Ottensmeier C, Pollock KGJ. Novel Approaches for Vaccination Against HPV-Induced Cancers. Curr Top Microbiol Immunol 2019; 405:33-53. [PMID: 25735921 DOI: 10.1007/82_2015_430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
To date, more than 5 % of all cancers are as a result of human papillomavirus (HPV) infection, and this incidence is increasing. Early recognition of disease is associated with good survival, but late presentation results in devastating consequences. Prevention is better than cure, and there are now successful prophylactic vaccination programmes in place. We discuss these and the prospect of therapeutic vaccinations in the near future to address a growing need for improved therapeutic options.
Collapse
Affiliation(s)
- Emma King
- University of Southampton, Southampton, UK.
| | | | | |
Collapse
|
38
|
Zaric M, Becker PD, Hervouet C, Kalcheva P, Doszpoly A, Blattman N, A O' Neill L, Yus BI, Cocita C, Kwon SY, Baker AH, Lord GM, Klavinskis LS. Skin immunisation activates an innate lymphoid cell-monocyte axis regulating CD8 + effector recruitment to mucosal tissues. Nat Commun 2019; 10:2214. [PMID: 31101810 PMCID: PMC6525176 DOI: 10.1038/s41467-019-09969-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 04/08/2019] [Indexed: 02/07/2023] Open
Abstract
CD8+ T cells provide a critical defence from pathogens at mucosal epithelia including the female reproductive tract (FRT). Mucosal immunisation is considered essential to initiate this response, however this is difficult to reconcile with evidence that antigen delivered to skin can recruit protective CD8+ T cells to mucosal tissues. Here we dissect the underlying mechanism. We show that adenovirus serotype 5 (Ad5) bio-distributes at very low level to non-lymphoid tissues after skin immunisation. This drives the expansion and activation of CD3- NK1.1+ group 1 innate lymphoid cells (ILC1) within the FRT, essential for recruitment of CD8+ T-cell effectors. Interferon gamma produced by activated ILC1 is critical to licence CD11b+Ly6C+ monocyte production of CXCL9, a chemokine required to recruit skin primed CXCR3+ CD8+T-cells to the FRT. Our findings reveal a novel role for ILC1 to recruit effector CD8+ T-cells to prevent virus spread and establish immune surveillance at barrier tissues.
Collapse
Affiliation(s)
- Marija Zaric
- School of Immunobiology and Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Pablo D Becker
- School of Immunobiology and Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Catherine Hervouet
- School of Immunobiology and Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Petya Kalcheva
- School of Immunobiology and Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Andor Doszpoly
- Centre for Cardiovascular Sciences, Queens Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Negin Blattman
- Biodesign Institute, Centre for Infectious Disease and Vaccinology, Arizona State University, Tempe, AZ, 85287, USA
| | - Lauren A O' Neill
- School of Immunobiology and Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Barbara Ibarzo Yus
- School of Immunobiology and Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Clement Cocita
- School of Immunobiology and Microbial Sciences, King's College London, London, SE1 9RT, UK
| | | | - Andrew H Baker
- Centre for Cardiovascular Sciences, Queens Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Graham M Lord
- School of Immunobiology and Microbial Sciences, King's College London, London, SE1 9RT, UK.,Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| | - Linda S Klavinskis
- School of Immunobiology and Microbial Sciences, King's College London, London, SE1 9RT, UK.
| |
Collapse
|
39
|
Frietze KM, Lijek R, Chackerian B. Applying lessons from human papillomavirus vaccines to the development of vaccines against Chlamydia trachomatis. Expert Rev Vaccines 2018; 17:959-966. [PMID: 30300019 PMCID: PMC6246778 DOI: 10.1080/14760584.2018.1534587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
INTRODUCTION Chlamydia trachomatis (Ct), the most common bacterial sexually transmitted infection (STI), leads to pelvic inflammatory disease, infertility, and ectopic pregnancy in women. In this Perspective, we discuss the successful human papillomavirus (HPV) vaccine as a case study to inform Ct vaccine efforts. Areas covered: The immunological basis of HPV vaccine-elicited protection is high-titer, long-lasting antibody responses in the genital tract which provides sterilizing immunity. These antibodies are elicited through parenteral administration of a subunit vaccine based on virus-like particles (VLPs) of HPV. We present three lessons learned from the successful HPV vaccine efforts: (1) antibodies alone can be sufficient to provide protection from STIs in the genital tract, (2) the successful generation of high antibody levels is due to the multivalent structure of HPV VLPs, (3) major challenges exist in designing vaccines that elicit appropriate effector T cells in the genital tract. We then discuss the possibility of antibody-based immunity for Ct. Expert commentary: In this Perspective, we present a case for developing antibody-eliciting vaccines, similar to the HPV vaccine, for Ct. Basic research into the mechanisms of Ct entry into host cells will reveal new vaccine targets, which may be antigens against which antibodies are not normally elicited during natural infection.
Collapse
Affiliation(s)
- Kathryn M Frietze
- a Department of Molecular Genetics and Microbiology , School of Medicine, University of New Mexico , Albuquerque , NM , USA
| | - Rebeccah Lijek
- b Department of Biological Sciences , Mount Holyoke College , South Hadley , MA , USA
| | - Bryce Chackerian
- a Department of Molecular Genetics and Microbiology , School of Medicine, University of New Mexico , Albuquerque , NM , USA
| |
Collapse
|
40
|
Wang A, Li N, Zhou J, Chen Y, Jiang M, Qi Y, Liu H, Liu Y, Liu D, Zhao J, Wang Y, Zhang G. Mapping the B cell epitopes within the major capsid protein L1 of human papillomavirus type 16. Int J Biol Macromol 2018; 118:1354-1361. [DOI: 10.1016/j.ijbiomac.2018.06.094] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 04/13/2018] [Accepted: 06/20/2018] [Indexed: 10/28/2022]
|
41
|
Bywaters SM, Brendle SA, Biryukov J, Wang JW, Walston J, Milici J, Roden RB, Meyers C, Christensen ND. Production and characterization of a novel HPV anti-L2 monoclonal antibody panel. Virology 2018; 524:106-113. [PMID: 30170240 DOI: 10.1016/j.virol.2018.08.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 08/17/2018] [Accepted: 08/20/2018] [Indexed: 12/21/2022]
Abstract
The major capsid protein of HPV, L1, assembles into pentamers that form a T = 7 icosahedral particle, but the location of the co-assembled minor capsid protein, L2, remains controversial. Several researchers have developed useful monoclonal antibodies targeting L2, but most react with linear epitopes toward the N-terminus. As a means to better define the virus capsid and better assess the localization and exposure of L2 epitopes in the context of assembled HPV, we have developed a panel of 30 monoclonal antibodies (mAbs) which target the N-terminus of L2 amino acids 11-200, previously defined as a broadly protective immunogen. Select mAbs were processed with enzymes and anti-L2 Fabs were generated. These new mAb/Fab probes will be beneficial in future studies to unravel the placement of L2 and to help better define the role of L2 in the HPV lifecycle and the nature of the broadly protective epitopes.
Collapse
Affiliation(s)
- S M Bywaters
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - S A Brendle
- Jake Gittlen Laboratories for Cancer Research, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - J Biryukov
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - J W Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA.
| | - J Walston
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - J Milici
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - R B Roden
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA.
| | - C Meyers
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - N D Christensen
- Jake Gittlen Laboratories for Cancer Research, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
42
|
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: 8.1] [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
|
43
|
Roles of Fc Domain and Exudation in L2 Antibody-Mediated Protection against Human Papillomavirus. J Virol 2018; 92:JVI.00572-18. [PMID: 29743371 DOI: 10.1128/jvi.00572-18] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 05/07/2018] [Indexed: 12/18/2022] Open
Abstract
To address how L2-specific antibodies prevent human papillomavirus (HPV) infection of the genital tract, we generated neutralizing monoclonal antibodies (MAbs) WW1, a rat IgG2a that binds L2 residues 17 to 36 (like mouse MAb RG1), and JWW3, a mouse IgG2b derivative of Mab24 specific for L2 residues 58 to 64. By Western blotting, WW1 recognized L2 of 29/34 HPV genotypes tested, compared to only 13/34 for RG1 and 25/34 for JWW3. WW1 IgG and F(ab')2 bound HPV16 pseudovirions similarly; however, whole IgG provided better protection against HPV vaginal challenge. Passive transfer of WW1 IgG was similarly protective in wild-type and neonatal Fc receptor (FcRn)-deficient mice, suggesting that protection by WW1 IgG is not mediated by FcRn-dependent transcytosis. Rather, local epithelial disruption, required for genital infection and induced by either brushing or nonoxynol-9 treatment, released serum IgG in the genital tract, suggesting Fc-independent exudation. Depletion of neutrophils and macrophages reduced protection of mice upon passive transfer of whole WW1 or JWW3 IgGs. Similarly, IgG-mediated protection by L2 MAbs WW1, JWW3, and RG1 was reduced in Fc receptor knockout compared to wild-type mice. However, levels of in vitro neutralization by WW1 IgG were similar in TRIM21 knockout and wild-type cells, indicating that Fc does not contribute to antibody-dependent intracellular neutralization (ADIN). In conclusion, the Fc domain of L2-specific IgGs is not active for ADIN, but it opsonizes bound extracellular pseudovirions for phagocytes in protecting mice from intravaginal HPV challenge. Systemically administered neutralizing IgG can access the site of infection in an abrasion via exudation without the need for FcRn-mediated transcytosis.IMPORTANCE At least 15 alpha HPV types are causative agents for 5% of all cancers worldwide, and beta types have been implicated in nonmelanoma skin cancer, whereas others produce benign papillomas, such as genital warts, associated with considerable morbidity and health systems costs. Vaccines targeting the minor capsid protein L2 have the potential to provide broad-spectrum immunity against medically relevant HPVs of divergent genera via the induction of broadly cross-neutralizing serum IgG. Here we examine the mechanisms by which L2-specific serum IgG reaches the viral inoculum in the genital tract to effect protection. Abrasion of the vaginal epithelium allows the virus to access and infect basal keratinocytes, and our findings suggest that this also permits the local exudation of neutralizing IgG and vaccine-induced sterilizing immunity. We also demonstrate the importance of Fc-mediated phagocytosis of L2 antibody-virion complexes for humoral immunity, a protective mechanism that is not detected by current in vitro neutralization assays.
Collapse
|
44
|
Bhatla N, Nene BM, Joshi S, Esmy PO, Poli URR, Joshi G, Verma Y, Zomawia E, Pimple S, Prabhu PR, Basu P, Muwonge R, Hingmire S, Sauvaget C, Lucas E, Pawlita M, Gheit T, Jayant K, Malvi SG, Siddiqi M, Michel A, Butt J, Sankaran S, Kannan TPRA, Varghese R, Divate U, Willhauck-Fleckenstein M, Waterboer T, Müller M, Sehr P, Kriplani A, Mishra G, Jadhav R, Thorat R, Tommasino M, Pillai MR, Sankaranarayanan R. Are two doses of human papillomavirus vaccine sufficient for girls aged 15-18 years? Results from a cohort study in India. PAPILLOMAVIRUS RESEARCH (AMSTERDAM, NETHERLANDS) 2018; 5:163-171. [PMID: 29578097 PMCID: PMC6047463 DOI: 10.1016/j.pvr.2018.03.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 03/20/2018] [Accepted: 03/21/2018] [Indexed: 01/08/2023]
Abstract
Extending two-dose recommendations of HPV vaccine to girls between 15 and 18 years will reduce program cost and improve compliance. Immunogenicity and vaccine targeted HPV infection outcomes were compared between 1795 girls aged 15-18 years receiving two (1-180 days) and 1515 girls of same age receiving three (1-60-180 days) doses. Immunogenicity outcomes in 15-18 year old two-dose recipients were also compared with the 10-14 year old three-dose (N = 2833) and two-dose (N = 3184) recipients. The 15-18 year old two-dose recipients had non-inferior L1-binding antibody titres at seven months against vaccine-targeted HPV types compared to three-dose recipients at 15-18 years and three-dose recipients at 10-14 years of age. Neutralizing antibody titres at 18 months in 15-18 year old two-dose recipients were non-inferior to same age three-dose recipients for all except HPV 18. The titres were inferior to those in the 10-14 year old three-dose recipients for all targeted types. Frequency of incident infections from vaccine-targeted HPV types in the 15-18 year old two-dose recipients was similar to the three dose recipients. None of the girls receiving two or three doses had persistent infection from vaccine-targeted types. These findings support that two doses of HPV vaccine can be extended to girls aged 15-18 years.
Collapse
Affiliation(s)
- Neerja Bhatla
- Department of Obstetrics & Gynaecology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Bhagwan M Nene
- Tata Memorial Centre Rural Cancer Project, Nargis Dutt Memorial Cancer Hospital, Barshi District, Solapur, Maharashtra 413401, India
| | - Smita Joshi
- Jehangir Clinical Development Centre, Jehangir Hospital Premises, Pune 411001, India
| | - Pulikottil O Esmy
- Christian Fellowship Community Health Centre, Ambillikai (near Oddanchathram), Dindigul District, Tamil Nadu 624612, India
| | - Usha Rani Reddy Poli
- MNJ Institute of Oncology & Regional Cancer Center, Red Hills, Lakadikapul, Hyderabad, Andhra Pradesh 500004, India
| | - Geeta Joshi
- Gujarat Cancer & Research Institute (GCRI), M.P. Shah Cancer Hospital, Civil Hospital Campus, Asarwa, Ahmedabad 380016, India
| | - Yogesh Verma
- Sikkim Manipal Institute of Medical Sciences, Sikkim Manipal University, Sikkim 737101, Sikkim, India
| | | | - Sharmila Pimple
- Department of Preventive Oncology, Tata Memorial Center, Tata Memorial Hospital & Cancer Research Inst, Parel, Mumbai 400012, India
| | - Priya R Prabhu
- Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram, 695014 Kerala, India
| | - Partha Basu
- Screening Group, Section of Early Detection and Prevention, International Agency for Research on Cancer, 50 cours Albert Thomas, 69372 Lyon Cedex 08, France
| | - Richard Muwonge
- Screening Group, Section of Early Detection and Prevention, International Agency for Research on Cancer, 50 cours Albert Thomas, 69372 Lyon Cedex 08, France
| | - Sanjay Hingmire
- Tata Memorial Centre Rural Cancer Project, Nargis Dutt Memorial Cancer Hospital, Barshi District, Solapur, Maharashtra 413401, India
| | - Catherine Sauvaget
- Screening Group, Section of Early Detection and Prevention, International Agency for Research on Cancer, 50 cours Albert Thomas, 69372 Lyon Cedex 08, France
| | - Eric Lucas
- Screening Group, Section of Early Detection and Prevention, International Agency for Research on Cancer, 50 cours Albert Thomas, 69372 Lyon Cedex 08, France
| | - Michael Pawlita
- Infection, inflammation and Cancer Program, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, D-69120 Heidelberg, Germany
| | - Tarik Gheit
- Section of Infections and Cancer Biology, International Agency for Research on Cancer, Lyon, France
| | - Kasturi Jayant
- Tata Memorial Centre Rural Cancer Project, Nargis Dutt Memorial Cancer Hospital, Barshi District, Solapur, Maharashtra 413401, India
| | - Sylla G Malvi
- Tata Memorial Centre Rural Cancer Project, Nargis Dutt Memorial Cancer Hospital, Barshi District, Solapur, Maharashtra 413401, India
| | - Maqsood Siddiqi
- Cancer Foundation of India, Kolkata, West Bengal 700039, India
| | - Angelika Michel
- Infection, inflammation and Cancer Program, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, D-69120 Heidelberg, Germany
| | - Julia Butt
- Infection, inflammation and Cancer Program, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, D-69120 Heidelberg, Germany
| | - Subha Sankaran
- Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram, 695014 Kerala, India
| | | | - Rintu Varghese
- Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram, 695014 Kerala, India
| | - Uma Divate
- Jehangir Clinical Development Centre, Jehangir Hospital Premises, Pune 411001, India
| | - Martina Willhauck-Fleckenstein
- Infection, inflammation and Cancer Program, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, D-69120 Heidelberg, Germany
| | - Tim Waterboer
- Infection, inflammation and Cancer Program, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, D-69120 Heidelberg, Germany
| | - Martin Müller
- Infection, inflammation and Cancer Program, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, D-69120 Heidelberg, Germany
| | - Peter Sehr
- EMBL-DKFZ Chemical Biology Core Facility, European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - Alka Kriplani
- Department of Obstetrics & Gynaecology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Gauravi Mishra
- Department of Preventive Oncology, Tata Memorial Center, Tata Memorial Hospital & Cancer Research Inst, Parel, Mumbai 400012, India
| | - Radhika Jadhav
- Jehangir Clinical Development Centre, Jehangir Hospital Premises, Pune 411001, India
| | - Ranjit Thorat
- Tata Memorial Centre Rural Cancer Project, Nargis Dutt Memorial Cancer Hospital, Barshi District, Solapur, Maharashtra 413401, India
| | - Massimo Tommasino
- Section of Infections and Cancer Biology, International Agency for Research on Cancer, Lyon, France
| | - M Radhakrishna Pillai
- Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram, 695014 Kerala, India
| | - Rengaswamy Sankaranarayanan
- Screening Group, Section of Early Detection and Prevention, International Agency for Research on Cancer, 50 cours Albert Thomas, 69372 Lyon Cedex 08, France.
| |
Collapse
|
45
|
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: 0.9] [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
|
46
|
Abstract
The discovery of genotype 16 as the prototype oncogenic human papillomavirus (HPV) initiated a quarter century of laboratory and epidemiological studies that demonstrated their necessary, but not sufficient, aetiological role in cervical and several other anogenital and oropharyngeal cancers. Early virus-induced immune deviation can lead to persistent subclinical infection that brings the risk of progression to cancer. Effective secondary prevention of cervical cancer through cytological and/or HPV screening depends on regular and widespread use in the general population, but coverage is inadequate in low-resource settings. The discovery that the major capsid antigen L1 could self-assemble into empty virus-like particles (VLPs) that are both highly immunogenic and protective led to the licensure of several prophylactic VLP-based HPV vaccines for the prevention of cervical cancer. The implementation of vaccination programmes in adolescent females is underway in many countries, but their impact critically depends on the population coverage and is improved by herd immunity. This Review considers how our expanding knowledge of the virology and immunology of HPV infection can be exploited to improve vaccine technologies and delivery of such preventive strategies to maximize reductions in HPV-associated disease, including incorporation of an HPV vaccine covering oncogenic types within a standard multitarget paediatric vaccine.
Collapse
Affiliation(s)
| | - Peter L. Stern
- Division of Molecular and Clinical Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
47
|
Garolla A, De Toni L, Bottacin A, Valente U, De Rocco Ponce M, Di Nisio A, Foresta C. Human Papillomavirus Prophylactic Vaccination improves reproductive outcome in infertile patients with HPV semen infection: a retrospective study. Sci Rep 2018; 8:912. [PMID: 29343824 PMCID: PMC5772512 DOI: 10.1038/s41598-018-19369-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 12/29/2017] [Indexed: 12/17/2022] Open
Abstract
In this study we aimed to evaluate the effect on reproductive outcome of HPV vaccination in male subjects of infertile couples with HPV semen infection. In this single-center study, we retrospectively enrolled 151 infertile couples with detection of HPV in semen, attending our Hospital Unit of Andrology between January 2013 and June 2015, counseled to receive adjuvant HPV vaccination. Seventy-nine accepted vaccination (vaccine group) whilst 72 did not (control group). Our protocol of follow-up, aimed to evaluate HPV viral clearance, consisted in semen analysis, INNO-LiPA and FISH for HPV in semen cells after 6 and 12 months from basal evaluation. Spontaneous pregnancies, miscarriages and live births were recorded. Progressive sperm motility and anti-sperm antibodies were improved in the vaccine group at both time points (p < 0,05 vs control arm). Forty-one pregnancies, 11 in the control group and 30 in the vaccine group, were recorded (respectively 15% and 38,9%, p < 0,05) and resulted into 4 deliveries and 7 miscarriages (control group) and 29 deliveries and one miscarriage (vaccine group, p < 0,05 vs control group). HPV detection on sperms was predictive of negative pregnancy outcome. Adjuvant vaccination associated with enhanced HPV healing in semen cells and increased rate of natural pregnancies and live births.
Collapse
Affiliation(s)
- Andrea Garolla
- Department of Medicine, Unit of Andrology and Reproductive Medicine, Section of Endocrinology & Centre for Male Gamete Cryopreservation, University of Padova, Padova, Italy
| | - Luca De Toni
- Department of Medicine, Unit of Andrology and Reproductive Medicine, Section of Endocrinology & Centre for Male Gamete Cryopreservation, University of Padova, Padova, Italy
| | - Alberto Bottacin
- Department of Medicine, Unit of Andrology and Reproductive Medicine, Section of Endocrinology & Centre for Male Gamete Cryopreservation, University of Padova, Padova, Italy
| | - Umberto Valente
- Department of Medicine, Unit of Andrology and Reproductive Medicine, Section of Endocrinology & Centre for Male Gamete Cryopreservation, University of Padova, Padova, Italy
| | - Maurizio De Rocco Ponce
- Department of Medicine, Unit of Andrology and Reproductive Medicine, Section of Endocrinology & Centre for Male Gamete Cryopreservation, University of Padova, Padova, Italy
| | - Andrea Di Nisio
- Department of Medicine, Unit of Andrology and Reproductive Medicine, Section of Endocrinology & Centre for Male Gamete Cryopreservation, University of Padova, Padova, Italy
| | - Carlo Foresta
- Department of Medicine, Unit of Andrology and Reproductive Medicine, Section of Endocrinology & Centre for Male Gamete Cryopreservation, University of Padova, Padova, Italy.
| |
Collapse
|
48
|
Abstract
HPV L1 virus-like particle (VLP) vaccines administered in a prime/boost series of three injections over six months have demonstrated remarkable prophylactic efficacy in clinical trials and effectiveness in national immunization programs with high rates of coverage. There is mounting evidence that the vaccines have similar efficacy and effectiveness even when administered in a single dose. The unexpected potency of one dose of these VLP vaccines may largely be attributed to structural features of the particles, which lead to the efficient generation of long-lived antigen-specific antibody-producing cells and unique features of the virus life cycle that make the HPV virions highly susceptible to antibody-mediated inhibition of infection.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/immunology
- 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/therapeutic use
- Humans
- Immunization Schedule
- Mass Vaccination
- Mice
- Papillomaviridae/immunology
- Papillomavirus Infections/prevention & control
- Papillomavirus Infections/therapy
- Papillomavirus Vaccines/administration & dosage
- Papillomavirus Vaccines/therapeutic use
- Pre-Exposure Prophylaxis/methods
- Program Evaluation
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/therapeutic use
- Virion/immunology
Collapse
Affiliation(s)
- John Schiller
- Laboratory of Cellular Oncology, Center for Cancer Research, NCI, Bethesda, MD, USA.
| | - Doug Lowy
- Laboratory of Cellular Oncology, Center for Cancer Research, NCI, Bethesda, MD, USA
| |
Collapse
|
49
|
|
50
|
Kalnin K, Chivukula S, Tibbitts T, Yan Y, Stegalkina S, Shen L, Cieszynski J, Costa V, Sabharwal R, Anderson SF, Christensen N, Jagu S, Roden RBS, Kleanthous H. Incorporation of RG1 epitope concatemers into a self-adjuvanting Flagellin-L2 vaccine broaden durable protection against cutaneous challenge with diverse human papillomavirus genotypes. Vaccine 2017; 35:4942-4951. [PMID: 28778613 PMCID: PMC6454882 DOI: 10.1016/j.vaccine.2017.07.086] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/14/2017] [Accepted: 07/23/2017] [Indexed: 12/23/2022]
Abstract
AIM To achieve durable and broad protection against human papillomaviruses by vaccination with multimers of minor capsid antigen L2 using self-adjuvanting fusions with the toll-like receptor-5 (TLR5) ligand bacterial flagellin (Fla) instead of co-formulation with alum. METHODS Fla fusions with L2 protective epitopes comprising residues 11-200, 11-88 and/or 17-38 of a single or multiple HPV types were produced in E. coli and their capacity to activate TLR5 signaling was assessed. Immunogenicity was evaluated serially following administration of 3 intramuscular doses of Fla-L2 multimer without exogenous adjuvant, followed by challenge 1, 3, 6 or 12months later, and efficacy compared to vaccination with human doses of L1 VLP vaccines (Gardasil and Cervarix) or L2 multimer formulated in alum. Serum antibody responses were assessed by peptide ELISA, in vitro neutralization assays and passive transfer to naïve rabbits in which End-Point Protection Titers (EPPT) were determined using serial dilutions of pooled immune sera collected 1, 3, 6 or 12months after completing active immunization. Efficacy was assessed by determining wart volume following concurrent challenge at different sites with HPV6/16/18/31/45/58 'quasivirions' containing cottontail rabbit papillomavirus (CRPV) genomes. RESULTS Vaccination in the absence of exogenous adjuvant with Fla-HPV16 L2 11-200 fusion protein elicited durable protection against HPV16, but limited cross-protection against other HPV types. Peptide mapping data suggested the importance of the 17-38 aa region in conferring immunity. Indeed, addition of L2 residues 17-38 of HPV6/18/31/39/52 to a Fla-HPV16 L2 11-200 or 11-88 elicited broader protection via active or passive immunization, similar to that seen with vaccination with an alum-adjuvanted L2 multimer comprising the aa 11-88 peptides of five or eight genital HPV types. CONCLUSIONS Vaccination with flagellin fused L2 multimers provided lasting (>1year) immunity without the need for an exogenous adjuvant. Inclusion of the L2 amino acid 17-38 region in such multi-HPV type fusions expanded the spectrum of protection.
Collapse
Affiliation(s)
- Kirill Kalnin
- Research, Sanofi Pasteur, 38 Sidney Street, Cambridge, MA, USA.
| | | | | | - Yanhua Yan
- Research, Sanofi Pasteur, 38 Sidney Street, Cambridge, MA, USA
| | | | - Lihua Shen
- Research, Sanofi Pasteur, 38 Sidney Street, Cambridge, MA, USA
| | | | - Victor Costa
- Research, Sanofi Pasteur, 38 Sidney Street, Cambridge, MA, USA
| | | | | | - Neil Christensen
- Department of Pathology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Subhashini Jagu
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Richard B S Roden
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | | |
Collapse
|