1
|
Hussain Z, Zhang Y, Qiu L, Gou S, Liu K. Exploring Clec9a in dendritic cell-based tumor immunotherapy for molecular insights and therapeutic potentials. NPJ Vaccines 2025; 10:27. [PMID: 39920156 PMCID: PMC11806010 DOI: 10.1038/s41541-025-01084-2] [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: 07/18/2024] [Accepted: 01/30/2025] [Indexed: 02/09/2025] Open
Abstract
The pivotal role of type 1 conventional dendritic cells (cDC1s) in the field of dendritic cell (DC)-based tumor immunotherapies has been gaining increasing recognition due to their superior antigen cross-presentation abilities and essential role in modulating immune responses. This review specifically highlights the C-type lectin receptor family 9 member A (Clec9a or DNGR-1), which is exclusively expressed on cDC1s and plays a pivotal role in augmenting antigen cross-presentation and cytotoxic T lymphocyte (CTL) responses while simultaneously mitigating off-target effects. These effects include the enhancement of the cDC1s cross-presentation, reducing autoimmune responses and systemic inflammation, as well as preventing the non-specific activation of other immune cells. Consequently, these actions may contribute to reduced toxicity and enhanced treatment efficacy in immunotherapy. The exceptional ability of Clec9a to cross-present dead cell-associated antigens and enhance both humoral and CTL responses makes it an optimal receptor for DC-based strategies aimed at strengthening antitumor immunity. This review provides a comprehensive overview of the molecular characterization, expression, and signaling mechanisms of Clec9a. Furthermore, it discusses the role of Clec9a in the induction and functional activation of Clec9a+ cDC1s, with a particular focus on addressing the challenges related to off-target effects and immune tolerance in the development of tumor vaccines. Additionally, this review explores the potential of Clec9a-targeted approaches to enhance the immunogenicity of tumor vaccines and addresses the utilization of Clec9a as a delivery target for specific agonists (such as STING agonists and αGC) to enhance their therapeutic effects. This novel approach leverages Clec9a's capacity to improve the precision and efficacy of these immunomodulatory molecules in tumor treatment. In summary, this review presents compelling evidence positioning Clec9a as a promising target for DC-based tumor immunotherapy, capable of enhancing the efficacy of vaccines and immune responses while minimizing adverse effects.
Collapse
Affiliation(s)
- Zubair Hussain
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Metabolic dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, China
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China
| | - Yueteng Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Lu Qiu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shanshan Gou
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- State Key Laboratory of Metabolic dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, China.
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China.
- China‒US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China.
| |
Collapse
|
2
|
Akumbom AM, Bergman AJ, Strickler H, Budhathoki C, Nkimbeng M, Grant R, Reynolds NR, Talaat KR. Understanding human papillomavirus vaccine response and efficacy in people living with HIV: A systematic mixed studies review and meta-analysis. PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0003931. [PMID: 39705286 PMCID: PMC11661617 DOI: 10.1371/journal.pgph.0003931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 10/21/2024] [Indexed: 12/22/2024]
Abstract
Coinfection with human papillomavirus (HPV) and HIV compounds the risks of developing cervical, anal, and HPV-associated oral neoplasia. Safe prophylactic vaccines are available to prevent HPV infections in people with HIV(PWH). Yet, vaccine efficacy and duration of protection remain questionable. Historically, the efficacy of vaccines has been suboptimal in PWH compared to people without HIV (PWoH).A systematic review of HPV vaccine trials in PWH was conducted using PRISMA guidelines. Outcomes of interest were vaccine efficacy, immunogenicity, and predictors of HPV vaccine efficacy. A secondary outcome was to assess age and sex differences. Efficacy was reviewed as cervical/anal/oral lesions or neoplasia, and incident or persistent HPV infection following vaccination. A random effects meta-analysis was performed comparing geometric mean titer (GMT) in PWH to PWoH. Twenty-eight studies out of 988 were eligible for inclusion in our study, and qualitatively synthesized. Eight of these studies were meta-analyzed. GMT results of HPV16 and HPV18 genotypes were significantly lower in PWH; Hedges's g -0.434 (95% CI: -0.823, -0.046) and Hedges's g -0.57 (95% CI: -0.72, -0.43), respectively. The mean difference in GMT for HPV18 between PWH and PWoH was -536.23 (95% CI: -830.66, -241.81); approximately 22 times higher than HPV18 seropositivity cut-offs, assuming milli-Merck Units per milliliter. Risk factors for incident or persistent infections in PWH included: failure to seroconvert after vaccination, baseline CD4+ T-cell count <500 cells/mm3, early age of sexual debut, HIV viral load ≥ 400 copies/mL. There was a trend towards decreased HPV vaccine efficacy in studies that included enrollees with a history of AIDS or AIDS-defining illness.Applying existing evidence of HPV vaccine efficacy on meaningful clinical outcomes in PWH is questionable. This could be influenced by the diversity of eligibility criteria across clinical trials of HPV vaccine efficacy. Precision medicine may offer novel alternatives for evaluating HPV vaccine efficacy in PWH.
Collapse
Affiliation(s)
- Alvine M. Akumbom
- Center for Infectious Disease and Nursing Innovation, Johns Hopkins School of Nursing, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Alanna J. Bergman
- University of Virginia School of Nursing, Charlottesville, Virginia, United States of America
| | - Howard Strickler
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Chakra Budhathoki
- Center for Infectious Disease and Nursing Innovation, Johns Hopkins School of Nursing, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Manka Nkimbeng
- Division of Health Policy and Management, University of Minnesota School of Public Health, Minneapolis, Minnesota, United States of America
| | - Raeven Grant
- David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, Los Angeles, California, United States of America
| | - Nancy R. Reynolds
- Center for Infectious Disease and Nursing Innovation, Johns Hopkins School of Nursing, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Kawsar R. Talaat
- Department of International Health, Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| |
Collapse
|
3
|
Șandru F, Radu AM, Petca A, Dumitrașcu MC, Petca RC, Roman AM. Unveiling the Therapeutic Horizon: HPV Vaccines and Their Impact on Cutaneous Diseases-A Comprehensive Review. Vaccines (Basel) 2024; 12:228. [PMID: 38543862 PMCID: PMC10974301 DOI: 10.3390/vaccines12030228] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/18/2024] [Accepted: 02/21/2024] [Indexed: 03/30/2025] Open
Abstract
Human papillomavirus (HPV) encompasses a diverse array of viruses, comprising approximately 200 serotypes that affect humans. While the majority of HPV strains are associated with benign skin or mucous membrane growths, a subset is implicated in severe health conditions, such as cervical, anal, vulvar, and vaginal cancers. Despite the established effectiveness of HPV vaccines in preventing cervical and anal carcinomas in particular, their therapeutic potential in addressing cutaneous diseases linked to diverse HPV strains remains an intriguing area of investigation. This narrative review critically examines the existing literature to assess the viability of HPV immunization as a therapeutic intervention for prevalent cutaneous conditions. These include genital and extragenital cutaneous warts, epidermodysplasia verruciformis, and keratinocyte carcinomas. The findings suggest a promising dual role for HPV vaccines in preventing and treating dermatologic conditions while emphasizing future research directions, including the immunization perspective against β-HPVs. Moreover, the presence of conflicting study outcomes underscores the imperative for larger-scale, randomized trials with well-matched control groups to validate the efficacy of HPV immunization in the dermatologic context. This review contributes valuable insights into the evolving landscape of HPV-vaccine applications in the field of dermatology.
Collapse
Affiliation(s)
- Florica Șandru
- Department of Dermatovenerology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania (A.-M.R.); (A.-M.R.)
- Dermatology Department, “Elias” University Emergency Hospital, 011461 Bucharest, Romania
| | - Andreea-Maria Radu
- Department of Dermatovenerology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania (A.-M.R.); (A.-M.R.)
- Dermatology Department, “Elias” University Emergency Hospital, 011461 Bucharest, Romania
| | - Aida Petca
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Obstetrics and Gynecology, Elias Emergency University Hospital, 011461 Bucharest, Romania
| | - Mihai Cristian Dumitrașcu
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Obstetrics and Gynecology, University Emergency Hospital of Bucharest, 050098 Bucharest, Romania
| | - Răzvan-Cosmin Petca
- Department of Urology, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Urology, ‘Prof. Dr. Th. Burghele’ Clinical Hospital, 050659 Bucharest, Romania
| | - Alexandra-Maria Roman
- Department of Dermatovenerology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania (A.-M.R.); (A.-M.R.)
- Dermatology Department, “Elias” University Emergency Hospital, 011461 Bucharest, Romania
| |
Collapse
|
4
|
Akbari E, Milani A, Seyedinkhorasani M, Bolhassani A. HPV co-infections with other pathogens in cancer development: A comprehensive review. J Med Virol 2023; 95:e29236. [PMID: 37997472 DOI: 10.1002/jmv.29236] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/13/2023] [Accepted: 11/02/2023] [Indexed: 11/25/2023]
Abstract
High-risk human papillomaviruses (HR-HPVs) cause various malignancies in the anogenital and oropharyngeal regions. About 70% of cervical and oropharyngeal cancers are caused by HPV types 16 and 18. Notably, some viruses including herpes simplex virus, Epstein-Barr virus, and human immunodeficiency virus along with various bacteria often interact with HPV, potentially impacting its replication, persistence, and cancer progression. Thus, HPV infection can be significantly influenced by co-infecting agents that influence infection dynamics and disease progression. Bacterial co-infections (e.g., Chlamydia trachomatis) along with bacterial vaginosis-related species also interact with HPV in genital tract leading to viral persistence and disease outcomes. Co-infections involving HPV and diverse infectious agents have significant implications for disease transmission and clinical progression. This review explores multiple facets of HPV infection encompassing the co-infection dynamics with other pathogens, interaction with the human microbiome, and its role in disease development.
Collapse
Affiliation(s)
- Elahe Akbari
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Alireza Milani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | | | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
5
|
Waheed DEN, Burdier FR, Eklund C, Baussano I, Mariz FC, Téblick L, Mugo N, Watson-Jones D, Stanley M, Baay M, Vorsters A. An update on one-dose HPV vaccine studies, immunobridging and humoral immune responses - A meeting report. Prev Med Rep 2023; 35:102368. [PMID: 37680853 PMCID: PMC10480621 DOI: 10.1016/j.pmedr.2023.102368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/09/2023] [Accepted: 08/13/2023] [Indexed: 09/09/2023] Open
Abstract
The 12th HPV Prevention and Control meeting was held on June 2-3, 2022, in Antwerp, Belgium. This technical meeting focused on several topics. This report summarises the discussions and lessons learned on two topics: an update on one-dose HPV vaccination studies and humoral immune responses upon HPV vaccination. Long-term follow-up studies from Costa Rica (eleven years) and India (ten years) report stable levels of antibodies after a single HPV vaccination. High vaccine effectiveness against incident persistent HPV 16/18 infection was seen in India (95.4%, 85.0-99.9) ten years postvaccination and in Kenya (97.5%, 81.7-99.7) eighteen months postvaccination, an important observation in a setting with a higher HPV prevalence. The potential impact of HPV vaccination using a one-dose schedule in India was modelled and showed that implementation of one-dose schedule can contribute towards achieving WHO Cervical Cancer elimination goals. These data support the WHO SAGE recommendations for adopting a one-dose schedule for females aged 9-20 years. Immunobridging studies were discussed during the meeting. General agreement was reached that when thoughtfully applied, they can support and accelerate the expanded use of HPV vaccine with new vaccine schedules, age cohorts, or vaccine formulations. Internationally standardised measurements of HPV immune responses important for the progress of HPV vaccinology field. Humoral immune responses upon HPV vaccination plateau at 24 months regardless of number of doses, therefore, data should be analysed after at least 24 months of follow-up to bridge studies accurately.
Collapse
Affiliation(s)
- Dur-e-Nayab Waheed
- Centre for Evaluation of Vaccination, Vaccine and Infectious Disease Institute, University of Antwerp, Belgium
| | - F. Ricardo Burdier
- Centre for Evaluation of Vaccination, Vaccine and Infectious Disease Institute, University of Antwerp, Belgium
| | - Carina Eklund
- Karolinska Institutet, Department of Laboratory Medicine, Huddinge, Sweden
| | - Iacopo Baussano
- International Agency for Research on Cancer, Early Detection, Prevention and Infections Branch Lyon, France
| | - Filipe Colaço Mariz
- German Cancer Research Center, DKFZ, Tumorvirus-Specific Vaccination Strategies, Heidelberg, Germany
| | - Laura Téblick
- Centre for Evaluation of Vaccination, Vaccine and Infectious Disease Institute, University of Antwerp, Belgium
| | - Nelly Mugo
- Kenya Medical Research Institute, Nairobi, Kenya
| | - Deborah Watson-Jones
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Margaret Stanley
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Marc Baay
- P95 Epidemiology & Pharmacovigilance, Leuven, Belgium
| | - Alex Vorsters
- Centre for Evaluation of Vaccination, Vaccine and Infectious Disease Institute, University of Antwerp, Belgium
| |
Collapse
|
6
|
Losada C, Samaha H, Scherer EM, Kazzi B, Khalil L, Ofotokun I, Rouphael N. Efficacy and Durability of Immune Response after Receipt of HPV Vaccines in People Living with HIV. Vaccines (Basel) 2023; 11:1067. [PMID: 37376456 DOI: 10.3390/vaccines11061067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/24/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
People living with HIV (PLH) experience higher rates of HPV infection as well as an increased risk of HPV-related disease, including malignancies. Although they are considered a high-priority group for HPV vaccination, there are limited data regarding the long-term immunogenicity and efficacy of HPV vaccines in this population. Seroconversion rates and geometric mean titers elicited by vaccination are lower in PLH compared to immunocompetent participants, especially in individuals with CD4 counts below 200 cells/mm3 and a detectable viral load. The significance of these differences is still unclear, as a correlate of protection has not been identified. Few studies have focused on demonstrating vaccine efficacy in PLH, with variable results depending on the age at vaccination and baseline seropositivity. Although waning humoral immunity for HPV seems to be more rapid in this population, there is evidence that suggests that seropositivity lasts at least 2-4 years following vaccination. Further research is needed to determine the differences between vaccine formulations and the impact of administrating additional doses on durability of immune protection.
Collapse
Affiliation(s)
- Cecilia Losada
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, Decatur, GA 30030, USA
| | - Hady Samaha
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, Decatur, GA 30030, USA
| | - Erin M Scherer
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, Decatur, GA 30030, USA
| | - Bahaa Kazzi
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, Decatur, GA 30030, USA
| | - Lana Khalil
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, Decatur, GA 30030, USA
| | - Ighovwerha Ofotokun
- Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Nadine Rouphael
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, Decatur, GA 30030, USA
| |
Collapse
|
7
|
Kheirvari M, Liu H, Tumban E. Virus-like Particle Vaccines and Platforms for Vaccine Development. Viruses 2023; 15:1109. [PMID: 37243195 PMCID: PMC10223759 DOI: 10.3390/v15051109] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Virus-like particles (VLPs) have gained a lot of interest within the past two decades. The use of VLP-based vaccines to protect against three infectious agents-hepatitis B virus, human papillomavirus, and hepatitis E virus-has been approved; they are very efficacious and offer long-lasting immune responses. Besides these, VLPs from other viral infectious agents (that infect humans, animals, plants, and bacteria) are under development. These VLPs, especially those from human and animal viruses, serve as stand-alone vaccines to protect against viruses from which the VLPs were derived. Additionally, VLPs, including those derived from plant and bacterial viruses, serve as platforms upon which to display foreign peptide antigens from other infectious agents or metabolic diseases such as cancer, i.e., they can be used to develop chimeric VLPs. The goal of chimeric VLPs is to enhance the immunogenicity of foreign peptides displayed on VLPs and not necessarily the platforms. This review provides a summary of VLP vaccines for human and veterinary use that have been approved and those that are under development. Furthermore, this review summarizes chimeric VLP vaccines that have been developed and tested in pre-clinical studies. Finally, the review concludes with a snapshot of the advantages of VLP-based vaccines such as hybrid/mosaic VLPs over conventional vaccine approaches such as live-attenuated and inactivated vaccines.
Collapse
Affiliation(s)
| | | | - Ebenezer Tumban
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX 79106, USA
| |
Collapse
|
8
|
Staadegaard L, Rönn MM, Soni N, Bellerose ME, Bloem P, Brisson M, Maheu-Giroux M, Barnabas RV, Drolet M, Mayaud P, Dalal S, Boily MC. Immunogenicity, safety, and efficacy of the HPV vaccines among people living with HIV: A systematic review and meta-analysis. EClinicalMedicine 2022; 52:101585. [PMID: 35936024 PMCID: PMC9350866 DOI: 10.1016/j.eclinm.2022.101585] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/08/2022] [Accepted: 07/08/2022] [Indexed: 01/21/2023] Open
Abstract
Background Vaccines have been demonstrated to protect against high-risk human papillomavirus infection (HPV), including HPV-16/18, and cervical lesions among HIV negative women. However, their efficacy remains uncertain for people living with HIV (PLHIV).We systematically reviewed available evidence on HPV vaccine on immunological, virological, or other biological outcomes in PLHIV. Methods We searched five electronic databases (PubMed, Medline and Embase, clinicaltrials.gov and the WHO clinical trial database) for longitudinal prospective studies reporting immunogenicity, virological, cytological, histological, clinical or safety endpoints following prophylactic HPV vaccination among PLHIV. We included studies published by February 11th, 2021. We summarized results, assessed study quality, and conducted meta-analysis and subgroup analyses, where possible. Findings We identified 43 publications stemming from 18 independent studies (Ns =18), evaluating the quadrivalent (Ns =15), bivalent (Ns =4) and nonavalent (Ns =1) vaccines. A high proportion seroconverted for the HPV vaccine types. Pooled proportion seropositive by 28 weeks following 3 doses with the bivalent, quadrivalent, and nonavalent vaccines were 0.99 (95% confidence interval: 0.95-1.00, Ns =1), 0.99 (0.98-1.00, Ns =9), and 1.00 (0.99-1.00, Ns =1) for HPV-16 and 0.99 (0.96-1.00, Ns =1), 0.94 (0.91-0.96, Ns =9), and 1.00 (0.99-1.00, Ns =1) for HPV-18, respectively. Seropositivity remained high among people who received 3 doses despite some declines in antibody titers and lower seropositivity over time, especially for HPV-18, for the quadrivalent than the bivalent vaccine, and for HIV positive than negative individuals. Seropositivity for HPV-18 at 29-99 weeks among PLHIV was 0.72 (0.66-0.79, Ns =8) and 0.96 (0.92-0.99, Ns =2) after 3 doses of the quadrivalent and bivalent vaccine, respectively and 0.94 (0.90-0.98, Ns =3) among HIV-negative historical controls. Evidence suggests that the seropositivity after vaccination declines over time but it can lasts at least 2-4 years. The vaccines were deemed safe among PLHIV with few serious adverse events. Evidence of HPV vaccine efficacy against acquisition of HPV infection and/or associated disease from the eight trials available was inconclusive due to the low quality. Interpretation PLHIV have a robust and safe immune response to HPV vaccination. Antibody titers and seropositivity rates decline over time but remain high. The lack of a formal correlate of protection and efficacy results preclude definitive conclusions on the clinical benefits. Nevertheless, given the burden of HPV disease in PLHIV, although the protection may be shorter or less robust against HPV-18, the robust immune response suggests that PLHIV may benefit from receiving HPV vaccination after acquiring HIV. Better quality studies are needed to demonstrate the clinical efficacy among PLHIV. Funding World Health Organization. MRC Centre for Global Infectious Disease Analysis, Canadian Institutes of Health Research, UK Medical Research Council (MRC).
Collapse
Affiliation(s)
- Lisa Staadegaard
- Department of Infectious Diseases Epidemiology, Imperial College, London, United Kingdom
| | - Minttu M. Rönn
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, United States
| | - Nirali Soni
- Department of Infectious Diseases Epidemiology, Imperial College, London, United Kingdom
- Medical Research Council Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, United Kingdom
| | - Meghan E. Bellerose
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, United States
| | - Paul Bloem
- World Health Organization, Geneva, Switzerland
| | - Marc Brisson
- Laval University, Québec, Canada
- Centre de recherche du CHU de Québec-Université Laval, Canada
| | - Mathieu Maheu-Giroux
- Department of Epidemiology and Biostatistics, School of Population and Global Health, McGill University, Montreal, Canada
| | - Ruanne V. Barnabas
- Department of Global Health, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Melanie Drolet
- Centre de recherche du CHU de Québec-Université Laval, Canada
| | - Philippe Mayaud
- Faculty of Infectious & Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Shona Dalal
- World Health Organization, Geneva, Switzerland
| | - Marie-Claude Boily
- Department of Infectious Diseases Epidemiology, Imperial College, London, United Kingdom
- Medical Research Council Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, United Kingdom
| |
Collapse
|
9
|
Pérez-González A, Cachay E, Ocampo A, Poveda E. Update on the Epidemiological Features and Clinical Implications of Human Papillomavirus Infection (HPV) and Human Immunodeficiency Virus (HIV) Coinfection. Microorganisms 2022; 10:1047. [PMID: 35630489 PMCID: PMC9147826 DOI: 10.3390/microorganisms10051047] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/12/2022] [Accepted: 05/17/2022] [Indexed: 12/01/2022] Open
Abstract
Human papillomavirus (HPV) infection is the most common sexually transmitted infection (STI) worldwide. Although most HPV infections will spontaneously resolve, a considerable proportion of them will persist, increasing the risk of anogenital dysplasia, especially within certain populations, such as patients infected with human immunodeficiency virus (HIV). Furthermore, high-risk oncogenic HPV types (HR-HPV) are the main cause of cervix and other anogenital cancers, such as cancer of the vagina, vulva, penis, or anus. HIV and HPV coinfection is common among people living with HIV (PLWH) but disproportionally affects men who have sex with men (MSM) for whom the rate of persistent HPV infection and reinfection is noteworthy. The molecular interactions between HIV and HPV, as well as the interplay between both viruses and the immune system, are increasingly being understood. The immune dysfunction induced by HIV infection impairs the rate of HPV clearance and increases its oncogenic risk. Despite the availability of effective antiretroviral therapy (ART), the incidence of several HPV-related cancers is higher in PLWH, and the burden of persistent HPV-related disease has become a significant concern in an aging HIV population. Several public health strategies have been developed to reduce the transmission of HIV and HPV and mitigate the consequences of this type of coinfection. Universal HPV vaccination is the most effective preventive tool to reduce the incidence of HPV disease. In addition, screening programs for HPV-related cervical and vulvovaginal diseases in women are well-recognized strategies to prevent cervical cancer. Similarly, anal dysplasia screening programs are being implemented worldwide for the prevention of anal cancer among PLWH. Herein, the main epidemiological features and clinical implications of HIV and HPV coinfection are reviewed, focusing mainly on the relationship between HIV immune status and HPV-related diseases and the current strategies used to reduce the burden of HPV-related disease.
Collapse
Affiliation(s)
- Alexandre Pérez-González
- Group of Virology and Pathogenesis, Galicia Sur Health Research Institute (IIS Galicia Sur), 36312 Vigo, Spain;
- Infectious Disease Unit, Internal Medicine Department, Hospital Álvaro Cunqueiro, 36312 Vigo, Spain;
| | - Edward Cachay
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California at San Diego, San Diego, CA 92093, USA;
| | - Antonio Ocampo
- Infectious Disease Unit, Internal Medicine Department, Hospital Álvaro Cunqueiro, 36312 Vigo, Spain;
| | - Eva Poveda
- Group of Virology and Pathogenesis, Galicia Sur Health Research Institute (IIS Galicia Sur), 36312 Vigo, Spain;
| |
Collapse
|
10
|
Kang M, Umbleja T, Ellsworth G, Aberg J, Wilkin T. Effects of Sex, Existing Antibodies, and HIV-1-Related and Other Baseline Factors on Antibody Responses to Quadrivalent HPV Vaccine in Persons With HIV. J Acquir Immune Defic Syndr 2022; 89:414-422. [PMID: 34907980 PMCID: PMC8881300 DOI: 10.1097/qai.0000000000002891] [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: 06/23/2021] [Accepted: 12/06/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND We compared antibody (Ab) responses to a quadrivalent (types 6, 11, 16, and 18) human papillomavirus (HPV) vaccine between men and women with HIV-1. METHODS A retrospective analysis of participant-level data from published clinical trials of HPV vaccine administered at study entry and at weeks 8 and 24 was conducted separately for baseline Ab undetectable and baseline Ab detectable using Ab titers and titer changes from baseline, respectively, at week 28 and year 1.5. Generalized estimating equations accounted for multiple HPV types and were adjusted for multiple baseline factors, including existing HPV antibodies before vaccination from natural exposure. RESULTS We evaluated 575 participants with CD4+ count >200 cells/mm3, 323 men and 252 women: median ages 46 and 38 years, respectively. Week 28 and year 1.5 Ab titers were similar between men and women regardless of the baseline Ab detection in multivariate models. HIV-1 RNA ≥400 copies/mm3 was associated with a lower week 28 Ab response; in baseline Ab detectable, the baseline HPV Ab titer level, HPV DNA detection, and lower CD4+/CD8+ ratio were also associated with a lower response. CD4+/CD8+ ratio was a stronger predictor in the year 1.5 Ab analysis than in the week 28 analysis. Ab responses among baseline Ab detectable were only somewhat higher than those among baseline Ab undetectable (eg, type 16 week 28 median 3.46 vs 3.20 log10 mMU/mL) despite the existing baseline titer (median 1.74). CONCLUSIONS We did not find any sex differences of serologic response to HPV vaccine. Ab titer gain was lower in those with preexisting antibodies due to previous natural infection.
Collapse
Affiliation(s)
- Minhee Kang
- Center for Biostatistics in AIDS Research in the Department
of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Triin Umbleja
- Center for Biostatistics in AIDS Research in the Department
of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Grant Ellsworth
- Division of Infectious Diseases, Weill Cornell Medicine,
New York, NY
| | - Judith Aberg
- Division of Infectious Diseases, Icahn School of Medicine
at Mount Sinai, New York, NY
| | - Timothy Wilkin
- Division of Infectious Diseases, Weill Cornell Medicine,
New York, NY
| |
Collapse
|
11
|
Castle PE, Einstein MH, Sahasrabuddhe VV. Cervical cancer prevention and control in women living with human immunodeficiency virus. CA Cancer J Clin 2021; 71:505-526. [PMID: 34499351 PMCID: PMC10054840 DOI: 10.3322/caac.21696] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/07/2021] [Accepted: 08/10/2021] [Indexed: 12/16/2022] Open
Abstract
Despite being highly preventable, cervical cancer is the fourth most common cancer and cause of cancer death in women globally. In low-income countries, cervical cancer is often the leading cause of cancer-related morbidity and mortality. Women living with human immunodeficiency virus (HIV)/acquired immunodeficiency syndrome are at a particularly high risk of cervical cancer because of an impaired immune response to human papillomavirus, the obligate cause of virtually all cervical cancers. Globally, approximately 1 in 20 cervical cancers is attributable to HIV; in sub-Saharan Africa, approximately 1 in 5 cervical cancers is due to HIV. Here, the authors provide a critical appraisal of the evidence to date on the impact of HIV disease on cervical cancer risk, describe key methodologic issues, and frame the key outstanding research questions, especially as they apply to ongoing global efforts for prevention and control of cervical cancer. Expanded efforts to integrate HIV care with cervical cancer prevention and control, and vice versa, could assist the global effort to eliminate cervical cancer as a public health problem.
Collapse
Affiliation(s)
- Philip E. Castle
- Division of Cancer PreventionNational Cancer InstituteNational Institutes of HealthRockvilleMaryland
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteNational Institutes of HealthRockvilleMaryland
| | - Mark H. Einstein
- Department of Obstetrics, Gynecology, and Reproductive HealthRutgers New Jersey Medical SchoolNewarkNew Jersey
| | - Vikrant V. Sahasrabuddhe
- Division of Cancer PreventionNational Cancer InstituteNational Institutes of HealthRockvilleMaryland
| |
Collapse
|
12
|
Shin MB, Liu G, Mugo N, Garcia PJ, Rao DW, Broshkevitch CJ, Eckert LO, Pinder LF, Wasserheit JN, Barnabas RV. A Framework for Cervical Cancer Elimination in Low-and-Middle-Income Countries: A Scoping Review and Roadmap for Interventions and Research Priorities. Front Public Health 2021; 9:670032. [PMID: 34277540 PMCID: PMC8281011 DOI: 10.3389/fpubh.2021.670032] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 05/19/2021] [Indexed: 12/14/2022] Open
Abstract
The World Health Organization announced an ambitious call for cervical cancer elimination worldwide. With existing prevention and treatment modalities, cervical cancer elimination is now within reach for high-income countries. Despite limited financing and capacity constraints in low-and-middle-income countries (LMICs), prevention and control efforts can be supported through integrated services and new technologies. We conducted this scoping review to outline a roadmap toward cervical cancer elimination in LMICs and highlight evidence-based interventions and research priorities to accelerate cervical cancer elimination. We reviewed and synthesized literature from 2010 to 2020 on primary and secondary cervical cancer prevention strategies. In addition, we conducted expert interviews with gynecologic and infectious disease providers, researchers, and LMIC health officials. Using these data, we developed a logic model to summarize the current state of science and identified evidence gaps and priority research questions for each prevention strategy. The logic model for cervical cancer elimination maps the needs for improved collaboration between policy makers, production and supply, healthcare systems, providers, health workers, and communities. The model articulates responsibilities for stakeholders and visualizes processes to increase access to and coverage of prevention methods. We discuss the challenges of contextual factors and highlight innovation needs. Effective prevention methods include HPV vaccination, screening using visual inspection and HPV testing, and thermocoagulation. However, vaccine coverage remains low in LMICs. New strategies, including single-dose vaccination could enhance impact. Loss to follow-up and treatment delays could be addressed by improved same-day screen-and-treat technologies. We provide a practical framework to guide cervical cancer elimination in LMICs. The scoping review highlights existing and innovative strategies, unmet needs, and collaborations required to achieve elimination across implementation contexts.
Collapse
Affiliation(s)
- Michelle B. Shin
- School of Nursing, University of Washington, Seattle, WA, United States
| | - Gui Liu
- Department of Epidemiology, University of Washington, Seattle, WA, United States
| | - Nelly Mugo
- Department of Global Health, University of Washington, Seattle, WA, United States
- Center for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Patricia J. Garcia
- Department of Global Health, University of Washington, Seattle, WA, United States
- School of Public Health, Cayetano Heredia University, Lima, Peru
| | - Darcy W. Rao
- Department of Epidemiology, University of Washington, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Cara J. Broshkevitch
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Linda O. Eckert
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Leeya F. Pinder
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
- Department of Obstetrics and Gynecology, University of Zambia, Lusaka, Zambia
| | - Judith N. Wasserheit
- Department of Epidemiology, University of Washington, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Medicine, University of Washington, Seattle, WA, United States
| | - Ruanne V. Barnabas
- Department of Epidemiology, University of Washington, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Medicine, University of Washington, Seattle, WA, United States
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| |
Collapse
|
13
|
Co-Administration of Aluminium Hydroxide Nanoparticles and Protective Antigen Domain 4 Encapsulated Non-Ionic Surfactant Vesicles Show Enhanced Immune Response and Superior Protection against Anthrax. Vaccines (Basel) 2020; 8:vaccines8040571. [PMID: 33019545 PMCID: PMC7711981 DOI: 10.3390/vaccines8040571] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 12/11/2022] Open
Abstract
Aluminium salts have been the adjuvant of choice in more than 100 licensed vaccines. Here, we have studied the synergistic effect of aluminium hydroxide nanoparticles (AH np) and non-ionic surfactant-based vesicles (NISV) in modulating the immune response against protective antigen domain 4 (D4) of Bacillus anthracis. NISV was prepared from Span 60 and cholesterol, while AH np was prepared from aluminium chloride and sodium hydroxide. AH np was co-administered with NISV encapsulating D4 (NISV-D4) to formulate AHnp/NISV-D4. The antigen-specific immune response of AHnp/NISV-D4 was compared with that of commercial alhydrogel (alhy) co-administered with NISV-D4 (alhydrogel/NISV-D4), NISV-D4, AHnp/D4, and alhydrogel/D4. Co-administration of NISV-D4 with AH np greatly improved the D4-specific antibody titer as compared to the control groups. Based on IgG isotyping and ex vivo cytokine analysis, AHnp/NISV-D4 generated a balanced Th1/Th2 response. Furthermore, AH np/NISV-D4 showed superior protection against anthrax spore challenge in comparison to other groups. Thus, we demonstrate the possibility of developing a novel combinatorial nanoformulation capable of augmenting both humoral and cellular response, paving the way for adjuvant research.
Collapse
|
14
|
Folschweiller N, Teixeira J, Joshi S, Goldani LZ, Supparatpinyo K, Basu P, Chotpitayasunondh T, Chetchotisakd P, Ruxrungtham K, Roteli-Martins C, Grinsztejn B, Quintana SM, Kumarasamy N, Poongulali S, Kulkarni V, Lin L, Datta SK, Descamps D, Dodet M, Dubin G, Friel D, Hezareh M, Karkada N, Meric Camilleri D, Poncelet S, Salaun B, Tavares-da-Silva F, Thomas-Jooris F, Struyf F. Immunogenicity and safety of the AS04-HPV-16/18 and HPV-6/11/16/18 human papillomavirus vaccines in asymptomatic young women living with HIV aged 15-25 years: A phase IV randomized comparative study. EClinicalMedicine 2020; 23:100353. [PMID: 32639485 PMCID: PMC7329699 DOI: 10.1016/j.eclinm.2020.100353] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 03/27/2020] [Accepted: 04/11/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Women living with HIV (WLWH) are at higher risk of acquisition and progression of human papillomavirus (HPV) infection. Evidence on effect of HPV vaccination in this population is limited. METHODS This phase IV randomized controlled observer-blind study assessed immunogenicity and safety of two HPV vaccines (AS04-HPV-16/18 vs. 4vHPV) given in WLWH (stage 1) and HIV- females aged 15-25 years. Co-primary endpoints were to demonstrate, in WLWH subjects, non-inferiority (and if demonstrated, superiority) of AS04-HPV-16/18 vs. 4vHPV for HPV-16 and HPV-18 by pseudovirion-based neutralization assay (PBNA) at month 7 and safety. Non-inferiority criteria was lower limit (LL) of the 95% confidence interval (CI) of the GMT ratio AS04-HPV-16/18/4vHPV above 0.5, in the according to protocol population. NCT01031069. FINDINGS Among 873 subjects recruited between 26-Oct-2010 and 14-May-2015, 546 were randomized (1:1) and received at least one vaccine dose (total vaccinated cohort, TVC): 257 were WLWH (129 AS04-HPV-16/18; 128 4vHPV) and 289 were subjects without HIV (144 AS04-HPV-16/18; 145 4vHPV). Baseline CD4 cell count in WLWH was at least 350 cells/mm3.At month 7, AS04-HPV-16/18 showed immunological superiority to 4vHPV in WLWH. Neutralizing anti-HPV-16 and HPV-18 antibody GMTs were 2·74 (95% CI: 1·83; 4·11) and 7·44 (95% CI: 4·79; 11·54) fold higher in AS04-HPV-16/18 vs. 4vHPV (LL of the GMT ratio >1 in TVC, p<0·0001), respectively. Similar results were observed by ELISA up to month 24.Solicited local and general symptoms were in line with product labels. The number of reported serious adverse events (SAEs) was balanced throughout the study. INTERPRETATION Both vaccines showed an acceptable safety profile in all subjects. Despite the absence of an immunological correlate of protection for HPV, differences in immune responses elicited by the vaccines especially for HPV-18 may translate into longer lasting or more robust protection against cervical cancer with the AS04-HPV-16/18 vaccine in WLWH.
Collapse
Affiliation(s)
| | | | - Smita Joshi
- Jehangir Clinical Development Centre and Prayas, Pune, India
| | - Luciano Z Goldani
- Hospital de Clinicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre Brazil
| | | | - Partha Basu
- Chittaranjan National Cancer Institute, Kolkata, India
| | | | | | | | | | - Beatriz Grinsztejn
- Instituto de pesquisa Clínica Evandro Chagas (IPEC), Rio de Janeiro, Brazil
| | | | | | | | - Vinay Kulkarni
- Jehangir Clinical Development Centre and Prayas, Pune, India
| | - Lan Lin
- GSK, Avenue Fleming 20, 1300 Wavre, Belgium
| | | | | | | | - Gary Dubin
- GSK, Avenue Fleming 20, 1300 Wavre, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Kim M, Kim S, Shin JY. Post-approval Safety Monitoring of Quadrivalent and Bivalent Human Papillomavirus Vaccines Based on Real-world Data from the Korea Adverse Events Reporting System (KAERS). Clin Drug Investig 2020; 40:727-735. [PMID: 32474825 DOI: 10.1007/s40261-020-00930-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND OBJECTIVES The vaccine adjuvant, AS04, present in bivalent human papilloma virus (bHPV) vaccines, induces greater local immune responses than the aluminum adjuvant in quadrivalent HPV (qHPV) vaccines. These distinctions might also result in disparities in immunogenicity and responsiveness, possibly contributing to differences in adverse events (AEs) between these vaccines. Here we comparatively analyzed AEs between qHPV and bHPV vaccines based on 10-year real-world AE data. METHODS We evaluated the Korea Adverse Events Reporting System (KAERS), a nationwide drug database including vaccines from January 2007 to December 2016, analyzing AEs reported for qHPV and bHPV vaccines. Vaccine-AE pairs were generated, and the characteristics of all reported AEs were analyzed. Signals were derived using the disproportionality method of signal detection algorithms (reporting odds ratios and information component). RESULTS Of the total 2566 HPV vaccine-associated AE reports, 2686 and 1994 were vaccine-AE pairs for qHPV and bHPV, respectively. Application site disorders were the most frequent AEs for both vaccines but were more frequently reported with the bHPV vaccine. The characteristics of non-application site AEs between the two vaccines were generally similar, but systemic AEs such as fever and fatigue were more common with the bHPV vaccine. Tremor, rash, eye pain, myopathy and circulatory failure were identified as signals in both qHPV and bHPV vaccines. CONCLUSIONS Both application site disorders and systemic AEs were somewhat more frequent with the bHPV vaccine than with the qHPV vaccine. This might be caused by an immune response induced by adjuvants contained within the vaccines.
Collapse
Affiliation(s)
- MoonJung Kim
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, South Korea
| | - SangHee Kim
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, South Korea
| | - Ju-Young Shin
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, South Korea.
| |
Collapse
|
16
|
The effectiveness of vaccination to prevent the papillomavirus infection: a systematic review and meta-analysis. Epidemiol Infect 2020; 147:e156. [PMID: 31063090 PMCID: PMC6518793 DOI: 10.1017/s0950268818003679] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Our purpose was to determine the effectiveness and harms of vaccination in patients with any sexual history to prevent the prevalence of papillomavirus infection. A search strategy was conducted in the MEDLINE, CENTRAL, EMBASE and LILACS databases. Searches were also conducted in other databases and unpublished literature. The risk of bias was evaluated with the Cochrane Collaboration's tool. Analysis of fixed effects was conducted. The primary outcome was the infection by any and each human papillomavirus (HPV) genotype, serious adverse effects and short-term adverse effects. The measure of the effect was the risk difference (RD) with a 95% confidence interval (CI). The planned interventions were bivalent vaccine/tetravalent/nonavalent vs. placebo/no intervention/other vaccines. We included 29 studies described in 35 publications. Bivalent HPV vaccine offers protection against HPV16 (RD −0.05, 95% CI −0.098 to −0.0032), HPV18 (RD −0.03, 95% CI −0.062 to −0.0004) and HPV16/18 genotypes (RD of −0.1, 95% CI −0.16 to −0.04). On the other side, tetravalent HPV vaccine offered protection against HPV6 (RD of −0.0500, 95% CI −0.0963 to −0.0230), HPV11 (RD −0.0198, 95% CI −0.0310 to −0.0085). Also, against HPV16 (RD of −0.0608, 95% CI −0.1126 to −0.0091) and HPV18 (RD of −0.0200, 95% CI −0.0408 to −0.0123). There was a reduction in the prevalence of HPV16, 18 and 16/18 genotypes when applying the bivalent vaccine, with no increase in adverse effects. Regarding the tetravalent vaccine, we found a reduction in the prevalence of HPV6, 11, 16 and 18 genotypes, with no increase in adverse effects.
Collapse
|
17
|
Yadav R, Zhai L, Tumban E. Virus-like Particle-Based L2 Vaccines against HPVs: Where Are We Today? Viruses 2019; 12:v12010018. [PMID: 31877975 PMCID: PMC7019592 DOI: 10.3390/v12010018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/15/2019] [Accepted: 12/18/2019] [Indexed: 12/17/2022] Open
Abstract
Human papillomaviruses (HPVs) are the most common sexually transmitted infections worldwide. Ninety percent of infected individuals clear the infection within two years; however, in the remaining 10% of infected individuals, the infection(s) persists and ultimately leads to cancers (anogenital cancers and head and neck cancers) and genital warts. Fortunately, three prophylactic vaccines have been approved to protect against HPV infections. The most recent HPV vaccine, Gardasil-9 (a nonavalent vaccine), protects against seven HPV types associated with ~90% of cervical cancer and against two HPV types associated with ~90% genital warts with little cross-protection against non-vaccine HPV types. The current vaccines are based on virus-like particles (VLPs) derived from the major capsid protein, L1. The L1 protein is not conserved among HPV types. The minor capsid protein, L2, on the other hand, is highly conserved among HPV types and has been an alternative target antigen, for over two decades, to develop a broadly protective HPV vaccine. The L2 protein, unlike the L1, cannot form VLPs and as such, it is less immunogenic. This review summarizes current studies aimed at developing HPV L2 vaccines by multivalently displaying L2 peptides on VLPs derived from bacteriophages and eukaryotic viruses. Recent data show that a monovalent HPV L1 VLP as well as bivalent MS2 VLPs displaying HPV L2 peptides (representing amino acids 17–36 and/or consensus amino acids 69–86) elicit robust broadly protective antibodies against diverse HPV types (6/11/16/18/26/31/33/34/35/39/43/44/45/51/52/53/56/58/59/66/68/73) associated with cancers and genital warts. Thus, VLP-based L2 vaccines look promising and may be favorable, in the near future, over current L1-based HPV vaccines and should be explored further.
Collapse
Affiliation(s)
- Rashi Yadav
- Department of Biological Sciences, Michigan Technological University, Houghton, MI 49931, USA; (R.Y.); (L.Z.)
| | - Lukai Zhai
- Department of Biological Sciences, Michigan Technological University, Houghton, MI 49931, USA; (R.Y.); (L.Z.)
- Current address: Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Ebenezer Tumban
- Department of Biological Sciences, Michigan Technological University, Houghton, MI 49931, USA; (R.Y.); (L.Z.)
- Correspondence: ; Tel.: +1-906-487-2256; Fax: +1-906-487-3167
| |
Collapse
|
18
|
Bergman H, Buckley BS, Villanueva G, Petkovic J, Garritty C, Lutje V, Riveros‐Balta AX, Low N, Henschke N, Cochrane Gynaecological, Neuro‐oncology and Orphan Cancer Group. Comparison of different human papillomavirus (HPV) vaccine types and dose schedules for prevention of HPV-related disease in females and males. Cochrane Database Syst Rev 2019; 2019:CD013479. [PMID: 31755549 PMCID: PMC6873216 DOI: 10.1002/14651858.cd013479] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Uptake of human papillomavirus (HPV) vaccine remains low in many countries, although the bivalent and quadrivalent HPV vaccines given as a three-dose schedule are effective in the prevention of precancerous lesions of the cervix in women. Simpler immunisation schedules, such as those with fewer doses, might reduce barriers to vaccination, as may programmes that include males. OBJECTIVES To evaluate the efficacy, immunogenicity, and harms of different dose schedules and different types of HPV vaccines in females and males. SEARCH METHODS We conducted electronic searches on 27 September 2018 in Ovid MEDLINE, the Cochrane Central Register of Controlled Trials (CENTRAL) (in the Cochrane Library), and Ovid Embase. We also searched the WHO International Clinical Trials Registry Platform, and ClinicalTrials.gov (both 27 September 2018), vaccine manufacturer websites, and checked reference lists from an index of HPV studies and other relevant systematic reviews. SELECTION CRITERIA We included randomised controlled trials (RCTs) with no language restriction. We considered studies if they enrolled HIV-negative males or females aged 9 to 26 years, or HIV-positive males or females of any age. DATA COLLECTION AND ANALYSIS We used methods recommended by Cochrane. We use the term 'control' to refer to comparator products containing an adjuvant or active vaccine and 'placebo' to refer to products that contain no adjuvant or active vaccine. Most primary outcomes in this review were clinical outcomes. However, for comparisons comparing dose schedules, the included RCTs were designed to measure antibody responses (i.e. immunogenicity) as the primary outcome, rather than clinical outcomes, since it is unethical to collect cervical samples from girls under 16 years of age. We analysed immunogenicity outcomes (i.e. geometric mean titres) with ratios of means, clinical outcomes (e.g. cancer and intraepithelial neoplasia) with risk ratios or rate ratios and, for serious adverse events and deaths, we calculated odds ratios. We rated the certainty of evidence with GRADE. MAIN RESULTS We included 20 RCTs with 31,940 participants. The length of follow-up in the included studies ranged from seven months to five years. Two doses versus three doses of HPV vaccine in 9- to 15-year-old females Antibody responses after two-dose and three-dose HPV vaccine schedules were similar after up to five years of follow-up (4 RCTs, moderate- to high-certainty evidence). No RCTs collected clinical outcome data. Evidence about serious adverse events in studies comparing dose schedules was of very low-certainty owing to imprecision and indirectness (three doses 35/1159; two doses 36/1158; 4 RCTs). One death was reported in the three-dose group (1/898) and none in the two-dose group (0/899) (low-certainty evidence). Interval between doses of HPV vaccine in 9- to 14-year-old females and males Antibody responses were stronger with a longer interval (6 or 12 months) between the first two doses of HPV vaccine than a shorter interval (2 or 6 months) at up to three years of follow-up (4 RCTs, moderate- to high-certainty evidence). No RCTs collected data about clinical outcomes. Evidence about serious adverse events in studies comparing intervals was of very low-certainty, owing to imprecision and indirectness. No deaths were reported in any of the studies (0/1898, 3 RCTs, low-certainty evidence). HPV vaccination of 10- to 26-year-old males In one RCT there was moderate-certainty evidence that quadrivalent HPV vaccine, compared with control, reduced the incidence of external genital lesions (control 36 per 3081 person-years; quadrivalent 6 per 3173 person-years; rate ratio 0.16, 95% CI 0.07 to 0.38; 6254 person-years) and anogenital warts (control 28 per 2814 person-years; quadrivalent 3 per 2831 person-years; rate ratio 0.11, 95% CI 0.03 to 0.38; 5645 person-years). The quadrivalent vaccine resulted in more injection-site adverse events, such as pain or redness, than control (537 versus 601 per 1000; risk ratio (RR) 1.12, 95% CI 1.06 to 1.18, 3895 participants, high-certainty evidence). There was very low-certainty evidence from two RCTs about serious adverse events with quadrivalent vaccine (control 12/2588; quadrivalent 8/2574), and about deaths (control 11/2591; quadrivalent 3/2582), owing to imprecision and indirectness. Nonavalent versus quadrivalent vaccine in 9- to 26-year-old females and males Three RCTs were included; one in females aged 9- to 15-years (n = 600), one in females aged 16- to 26-years (n = 14,215), and one in males aged 16- to 26-years (n = 500). The RCT in 16- to 26-year-old females reported clinical outcomes. There was little to no difference in the incidence of the combined outcome of high-grade cervical epithelial neoplasia, adenocarcinoma in situ, or cervical cancer between the HPV vaccines (quadrivalent 325/6882, nonavalent 326/6871; OR 1.00, 95% CI 0.85 to 1.16; 13,753 participants; high-certainty evidence). The other two RCTs did not collect data about clinical outcomes. There were slightly more local adverse events with the nonavalent vaccine (905 per 1000) than the quadrivalent vaccine (846 per 1000) (RR 1.07, 95% CI 1.05 to 1.08; 3 RCTs, 15,863 participants; high-certainty evidence). Comparative evidence about serious adverse events in the three RCTs (nonavalent 243/8234, quadrivalent 192/7629; OR 0.60, 95% CI 0.14 to 2.61) was of low certainty, owing to imprecision and indirectness. HPV vaccination for people living with HIV Seven RCTs reported on HPV vaccines in people with HIV, with two small trials that collected data about clinical outcomes. Antibody responses were higher following vaccination with either bivalent or quadrivalent HPV vaccine than with control, and these responses could be demonstrated to have been maintained for up to 24 months in children living with HIV (low-certainty evidence). The evidence about clinical outcomes and harms for HPV vaccines in people with HIV is very uncertain (low- to very low-certainty evidence), owing to imprecision and indirectness. AUTHORS' CONCLUSIONS The immunogenicity of two-dose and three-dose HPV vaccine schedules, measured using antibody responses in young females, is comparable. The quadrivalent vaccine probably reduces external genital lesions and anogenital warts in males compared with control. The nonavalent and quadrivalent vaccines offer similar protection against a combined outcome of cervical, vaginal, and vulval precancer lesions or cancer. In people living with HIV, both the bivalent and quadrivalent HPV vaccines result in high antibody responses. For all comparisons of alternative HPV vaccine schedules, the certainty of the body of evidence about serious adverse events reported during the study periods was low or very low, either because the number of events was low, or the evidence was indirect, or both. Post-marketing surveillance is needed to continue monitoring harms that might be associated with HPV vaccines in the population, and this evidence will be incorporated in future updates of this review. Long-term observational studies are needed to determine the effectiveness of reduced-dose schedules against HPV-related cancer endpoints, and whether adopting these schedules improves vaccine coverage rates.
Collapse
Affiliation(s)
- Hanna Bergman
- CochraneCochrane ResponseSt Albans House57‐59 HaymarketLondonUKSW1Y 4QX
| | - Brian S Buckley
- CochraneCochrane ResponseSt Albans House57‐59 HaymarketLondonUKSW1Y 4QX
- University of PhillipinesDepartment of SurgeryManilaPhilippines
| | - Gemma Villanueva
- CochraneCochrane ResponseSt Albans House57‐59 HaymarketLondonUKSW1Y 4QX
| | - Jennifer Petkovic
- CochraneCochrane ResponseSt Albans House57‐59 HaymarketLondonUKSW1Y 4QX
- University of OttawaBruyère Research Institute43 Bruyère StAnnex E, room 312OttawaONCanadaK1N 5C8
| | - Chantelle Garritty
- CochraneCochrane ResponseSt Albans House57‐59 HaymarketLondonUKSW1Y 4QX
- Ottawa Hospital Research InstituteOttawa Methods Centre, Clinical Epidemiology ProgramOttawaOntarioCanadaK1H 8L1
| | - Vittoria Lutje
- Liverpool School of Tropical MedicineDepartment of Clinical SciencesPembroke PlaceLiverpoolUKL3 5QA
| | | | - Nicola Low
- University of BernInstitute of Social and Preventive Medicine (ISPM)Finkenhubelweg 11BernSwitzerlandCH‐3012
| | - Nicholas Henschke
- CochraneCochrane ResponseSt Albans House57‐59 HaymarketLondonUKSW1Y 4QX
| | | |
Collapse
|
19
|
Zhan Y, Liu X, Feng Y, Wu S, Jiang Y. Safety and efficacy of human papillomavirus vaccination for people living with HIV: A systematic review and meta-analysis. Int J STD AIDS 2019; 30:1105-1115. [PMID: 31551002 DOI: 10.1177/0956462419852224] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The current evidence regarding the safety and immunogenicity of human papillomavirus (HPV) vaccinations for people living with HIV (PLWH) is unclear. We searched PubMed, Embase, Cochrane Library and Web of Science databases from inception to 23 November 2018. The pooled proportion, relative risk (RR) and the standardized mean difference (SMD) with 95% confidence intervals (CIs) were calculated. Twenty-four studies consisting of 7507 participants were identified. The pooled proportion of adverse events in HIV-infected vaccinees was 60% and the antibody seroconversion rates in HPV-6, -11, -16, -18 subtypes were all above 90%. When compared with the placebo groups, the risk of adverse events was not different except for the injection site reactions (RR: 2.63, 95% CI: 1.72–4.01, p < 0.001), and the level of CD4 was relatively lower (SMD: −0.17, 95% CI: −0.29 to −0.04, p = 0.01) in the HIV-positive vaccinees groups. When compared with HIV-negative vaccinees, the risk of adverse events was not different, but the pooled RR and SMD indicated that antibody seroconversion and geometric mean titer for HPV-18 in HIV-positive groups was lower (RR: 0.91, 95% CI: 0.87–0.95, p < 0.001; SMD: −0.43, 95% CI: −0.62 to −0.24, p < 0.001). The study proves that HPV vaccine is safe and efficacious for PLWH and has important implications for international guidelines and strategies for HPV vaccination.
Collapse
Affiliation(s)
- Yongle Zhan
- School of Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Xuan Liu
- School of Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Yahui Feng
- School of Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Sansan Wu
- School of Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Yu Jiang
- School of Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
20
|
Mavundza EJ, Wiyeh AB, Mahasha PW, Halle-Ekane G, Wiysonge CS. A systematic review of immunogenicity, clinical efficacy and safety of human papillomavirus vaccines in people living with the human immunodeficiency virus. Hum Vaccin Immunother 2019; 16:426-435. [PMID: 31448991 PMCID: PMC7062428 DOI: 10.1080/21645515.2019.1656481] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/26/2019] [Accepted: 08/09/2019] [Indexed: 02/07/2023] Open
Abstract
The human papillomavirus (HPV) is the most prevalent sexually transmitted infection worldwide. People living with the human immunodeficiency virus (HIV) are at high risk of HPV infection. This systematic review evaluates the immunogenicity, clinical efficacy, and safety of prophylactic HPV vaccines in people living with HIV. We registered the protocol for this review in the International Prospective Register of Systematic Reviews (CRD42018109898) and prepared the review following the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA). Five randomized trials with 1042 participants are included in this review. One trial with 120 participants compared the bivalent HPV vaccine to placebo, three trials with 830 participants compared the quadrivalent vaccine to placebo, and another trial with 92 participants compared the quadrivalent to the bivalent vaccine. There was low to moderate certainty evidence suggesting that seroconversion was higher among participants in the vaccine arms compared to the placebo arms for both vaccines. In one study with very low certainty evidence, participants who received the bivalent vaccine had higher anti-HPV-18 geometric mean titers (GMTs) compared to those who received the quadrivalent vaccine, despite little difference in anti-HPV-16 GMTs between the two vaccines. There were no differences in the incident and persistent HPV infections in both groups. None of the studies reported data on the incidence of precancerous lesions, or cancer. There were no reports of serious adverse events following vaccination in any of the trials. None of the included studies assessed the effects of HPV vaccines in adolescents living with HIV. Very limited evidence suggests lower immunogenicity of HPV vaccines in HIV positive compared to HIV-negative people. Finally, the long-term effect of the HPV vaccine in the incidence of cervical precancerous lesions and cervical cancer needs to be monitored. There is an urgent need for more high-quality randomized controlled trials that can address these gaps.
Collapse
Affiliation(s)
- Edison J. Mavundza
- Cochrane South Africa, South African Medical Research Council, Cape Town, South Africa
| | - Alison B. Wiyeh
- Cochrane South Africa, South African Medical Research Council, Cape Town, South Africa
| | - Phetole W. Mahasha
- Cochrane South Africa, South African Medical Research Council, Cape Town, South Africa
| | | | - Charles S. Wiysonge
- Cochrane South Africa, South African Medical Research Council, Cape Town, South Africa
- Division of Epidemiology and Biostatistics, Department of Global Health, Stellenbosch University, Cape Town, South Africa
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
21
|
Todorova A, Schwierzeck V, Turek D, Zink A, Schwerdtfeger C, Kaliebe K, Spinner CD, Traidl-Hoffmann C. Evaluation of anal carcinoma screening in male and female HIV patients at an interdisciplinary HIV therapy centre. J Eur Acad Dermatol Venereol 2019; 33:1595-1601. [PMID: 30883929 DOI: 10.1111/jdv.15575] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/01/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Incidence of anal carcinoma is increased in people living with HIV (PLWH). Due to the improved life expectancy in PLWH, identifying appropriate prevention strategies for non-AIDS-defining cancer types such as anal carcinoma has become a priority in managing PLWH today. OBJECTIVE We aimed to evaluate anal cytology assessment as screening tool for anal dysplasia and/or carcinoma in PLWH, regardless of gender or sexual orientation. Additionally, we investigated the correlation between cancer risk factors and abnormal screening results in our patient cohort. METHODS People living with HIV from the Interdisciplinary HIV Centre of the University Hospital rechts der Isar in Munich, Germany (IZAR), were screened for anal carcinoma by single cytobrush examination and anal Papanicolaou (PAP) smear assessment from 2013 to 2015. Patients with abnormal PAP smear result were offered a follow-up examination after 12 months. Differences between two groups were tested for statistical significance using Student's t-test and Mann-Whitney U-test, as appropriate. RESULTS In total, 101 PLWH were included. 26.7% of subjects (n = 27) were PAP IIID, and 9.9% (n = 10) were PAP IVa. Seven female subjects had an abnormal finding at screening. Smoking was significantly associated with abnormal findings at screening (P = 0.005). In addition, our study found an association between sexually transmitted infections (STI) and anal dysplasia. Condylomata acuminata were increased in subjects with PAP IIID/PAP IVa (P = 0.045). Reactive syphilis serology was found to be significantly associated with abnormal screening results (P = 0.016), respectively. CONCLUSION Our results demonstrate that smoking and two common STIs, condylomata acuminata and syphilis, are risk factors associated with advanced anal intraepithelial neoplasia (AIN) stages in our PLWH cohort. While further analysis is needed to determine diagnostic guidelines concerning AIN in PLWH, these results suggest that interdisciplinary lifestyle prevention strategies are required to reduce the risk factors for AIN in PLWH in an outpatient setting.
Collapse
Affiliation(s)
- A Todorova
- Interdisciplinary HIV Center (IZAR), University Hospital rechts der Isar, Technical University of Munich, Munich, Germany.,Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and Helmholtz Zentrum Munich - German Research Center for Environmental Health (GmbH), Munich, Germany
| | - V Schwierzeck
- Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and Helmholtz Zentrum Munich - German Research Center for Environmental Health (GmbH), Munich, Germany
| | - D Turek
- Interdisciplinary HIV Center (IZAR), University Hospital rechts der Isar, Technical University of Munich, Munich, Germany.,Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and Helmholtz Zentrum Munich - German Research Center for Environmental Health (GmbH), Munich, Germany
| | - A Zink
- Interdisciplinary HIV Center (IZAR), University Hospital rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Dermatology and Allergy, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - C Schwerdtfeger
- Interdisciplinary HIV Center (IZAR), University Hospital rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Medicine II, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - K Kaliebe
- Interdisciplinary HIV Center (IZAR), University Hospital rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Dermatology and Allergy, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - C D Spinner
- Interdisciplinary HIV Center (IZAR), University Hospital rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Medicine II, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - C Traidl-Hoffmann
- Interdisciplinary HIV Center (IZAR), University Hospital rechts der Isar, Technical University of Munich, Munich, Germany.,Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and Helmholtz Zentrum Munich - German Research Center for Environmental Health (GmbH), Munich, Germany
| |
Collapse
|
22
|
Martínez-Gómez X, Curran A, Campins M, Alemany L, Rodrigo-Pendás JÁ, Borruel N, Castellsagué X, Díaz-de-Heredia C, Moraga-Llop FA, Del Pino M, Torné A. Multidisciplinary, evidence-based consensus guidelines for human papillomavirus (HPV) vaccination in high-risk populations, Spain, 2016. Euro Surveill 2019; 24:1700857. [PMID: 30782268 PMCID: PMC6381660 DOI: 10.2807/1560-7917.es.2019.24.7.1700857] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 10/15/2018] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Although human papillomavirus (HPV) routine vaccination programmes have been implemented around the world and recommendations have been expanded to include other high-risk individuals, current recommendations often differ between countries in Europe, as well as worldwide. AIM To find and summarise the best available evidence of HPV vaccination in high-risk patients aiding clinicians and public health workers in the day-to-day vaccine decisions relating to HPV in Spain. METHODS We conducted a systematic review of the immunogenicity, safety and efficacy/effectiveness of HPV vaccination in high-risk populations between January 2006 and June 2016. HPV vaccination recommendations were established with levels of evidence according to the Grading of Recommendations Assessment, Development and Evaluation (GRADE) system. RESULTS A strong recommendation about HPV vaccination was made in the following groups: HIV infected patients aged 9-26 years; men who have sex with men aged 9-26 years; women with precancerous cervical lesions; patients with congenital bone marrow failure syndrome; women who have received a solid organ transplant or hematopoietic stem cell transplantation aged 9-26 years; and patients diagnosed with recurrent respiratory papillomatosis. CONCLUSIONS Data concerning non-routine HPV vaccination in populations with a high risk of HPV infection and associated lesions were scarce. We have developed a document to evaluate and establish evidence-based guidelines on HPV vaccination in high-risk populations in Spain, based on best available scientific evidence.
Collapse
Affiliation(s)
- Xavier Martínez-Gómez
- Servei de Medicina Preventiva i Epidemiologia, Hospital Universitari Vall d'Hebron - Universitat Autònoma de Barcelona, Barcelona, España
| | - Adrian Curran
- Servei de Malalties Infeccioses, Hospital Universitari Vall d'Hebron - Universitat Autònoma de Barcelona, Barcelona, España
| | - Magda Campins
- Servei de Medicina Preventiva i Epidemiologia, Hospital Universitari Vall d'Hebron - Universitat Autònoma de Barcelona, Barcelona, España
| | - Laia Alemany
- Programa de Recerca en Epidemiologia del Càncer, Institut Català d'Oncologia - IDIBELL CIBER Epidemiología y Salud Pública, Barcelona, España
| | - José Ángel Rodrigo-Pendás
- Servei de Medicina Preventiva i Epidemiologia, Hospital Universitari Vall d'Hebron - Universitat Autònoma de Barcelona, Barcelona, España
| | - Natalia Borruel
- Unitat d'Atenció Crohn-Colitis, Servei d'Aparell Digestiu; Hospital Universitari Vall d'Hebron - Universitat Autònoma de Barcelona, Barcelona, España
| | - Xavier Castellsagué
- Programa de Recerca en Epidemiologia del Càncer, Institut Català d'Oncologia - IDIBELL CIBER Epidemiología y Salud Pública, Barcelona, España
| | - Cristina Díaz-de-Heredia
- Servei d'Oncologia i Hematologia Pediàtrica, Hospital Universitari Vall d'Hebron - Universitat Autònoma de Barcelona, Barcelona, España
| | | | - Marta Del Pino
- Unidad de Ginecología Oncológica, Instituto Clínico de Ginecología y Obstetricia y Neonatología (ICGON), Hospital Clínic de Barcelona, Barcelona, España
- Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Facultad de Medicina, Universidad de Barcelona, Barcelona, España
| | - Aureli Torné
- Unidad de Ginecología Oncológica, Instituto Clínico de Ginecología y Obstetricia y Neonatología (ICGON), Hospital Clínic de Barcelona, Barcelona, España
- Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Facultad de Medicina, Universidad de Barcelona, Barcelona, España
| |
Collapse
|
23
|
Phillips A, Patel C, Pillsbury A, Brotherton J, Macartney K. Safety of Human Papillomavirus Vaccines: An Updated Review. Drug Saf 2018; 41:329-346. [PMID: 29280070 DOI: 10.1007/s40264-017-0625-z] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Human papillomavirus (HPV) vaccines are now included in immunisation programmes in 71 countries. Unfortunately, uptake has been impacted in some countries by reduced confidence in the safety of the HPV vaccine. In 2013, we published an extensive review demonstrating a reassuring safety profile for bivalent (2vHPV) and quadrivalent (4vHPV) vaccines. A nonavalent (9vHPV) vaccine is now available and HPV immunisation programmes have been extended to males in 11 countries. The aim of this updated narrative review was to examine the evidence on HPV vaccine safety, focusing on the 9vHPV vaccine, special populations and adverse events of special interest (AESI). The previous searches were replicated to identify studies to August 2016, including additional search terms for AESI. We identified 109 studies, including 15 population-based studies in over 2.5 million vaccinated individuals across six countries. All vaccines demonstrated an acceptable safety profile; injection-site reactions were slightly more common for 9vHPV vaccine than for 4vHPV vaccine. There was no consistent evidence of an increased risk of any AESI, including demyelinating syndromes or neurological conditions such as complex regional pain or postural orthostatic tachycardia syndromes. The risk-benefit profile for HPV vaccines remains highly favourable.
Collapse
Affiliation(s)
| | - Cyra Patel
- National Centre for Immunisation Research and Surveillance, Kids Research Institute, The Sydney Children's Hospitals Network, Cnr Hawkesbury Road and Hainsworth Street, Westmead, NSW, 2145, Australia
| | - Alexis Pillsbury
- National Centre for Immunisation Research and Surveillance, Kids Research Institute, The Sydney Children's Hospitals Network, Cnr Hawkesbury Road and Hainsworth Street, Westmead, NSW, 2145, Australia
| | - Julia Brotherton
- National HPV Vaccination Program Register, Victorian Cytology Service, Level 6, 176 Wellington Parade, East Melbourne, Carlton, VIC, 3002, Australia
- The University of Melbourne, Melbourne, VIC, Australia
| | - Kristine Macartney
- The University of Sydney, Sydney, NSW, Australia.
- National Centre for Immunisation Research and Surveillance, Kids Research Institute, The Sydney Children's Hospitals Network, Cnr Hawkesbury Road and Hainsworth Street, Westmead, NSW, 2145, Australia.
| |
Collapse
|
24
|
Tian T, Wang D, Papamichael C, Yan Z, Guoyao S, Zhanlin Z, Mahan Y, Xiaoqing T, Zheng G, Jianghong D. HPV vaccination acceptability among men who have sex with men in Urumqi, China. Hum Vaccin Immunother 2018; 15:1005-1012. [PMID: 30230946 PMCID: PMC6628869 DOI: 10.1080/21645515.2018.1520591] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background: Human papillomavirus (HPV) infection is common among men who have sex with men (MSM), especially among Human Immunodeficiency Virus (HIV)-infected MSM. The prevalence of HPV among MSM, accounts for the higher incidence of HPV-related cancer observed in this population. It is well known that targeted HPV vaccination is an effective way to prevent HPV infection; an intervention which could be beneficial for a high-risk group such as MSM. The current study aimed to assess the attitudes towards and acceptability of the HPV vaccine among MSM in in Urumqi, China. Methods: A total of 253 HIV-uninfected and 205 HIV-infected MSM (in Urumqi, China participated in the current cross-sectional study. Information on HPV-related knowledge, willingness to be vaccinated as well as demographic data were collected through a self-administered survey. A logistic regression model was applied to determine the predictors of HPV vaccine acceptability among the population. Results: The survey results indicated that only 218 (47.6%) of MSM questioned were aware of the term HPV, nevertheless, once awareness was established the prevalence of acceptability of free HPV vaccine was recorded at 96.7% of the total MSM sample. However, HIV-uninfected and HIV-infected MSM demonstrated significantly different attitudes in regard to their acceptability of free HPV vaccination (94.9% vs. 99.0%, p < 0.001) as well as their willingness to pay for the HPV vaccination (64.8% vs. 80.5%, p < 0.001), with the HIV-infected group being significantly more receptive towards HPV immunization. A multivariate logistic regression analysis indicated that local residency, employment status, hepatitis B vaccination status, previous awareness of HPV and HIV status were independent predictors of the participants’ willingness to pay for HPV vaccination. Conclusions: Participants in the current study demonstrated poor knowledge of HPV but the majority of MSM were willing to accept HPV vaccine after consultation; with HIV-infected MSM displaying higher acceptability towards a potential HPV vaccination than HIV-uninfected MSM. MSM who were previously aware of HPV were more likely to be willing to pay for HPV vaccine.
Collapse
Affiliation(s)
- Tian Tian
- a Department of Epidemiology and Health Statistics, School of Public Health , Xinjiang Medical University , Urumqi , Xinjiang , China
| | - Duolao Wang
- b Department of Clinical Sciences , Liverpool School of Tropical Medicine , Liverpool , UK
| | - Christiana Papamichael
- b Department of Clinical Sciences , Liverpool School of Tropical Medicine , Liverpool , UK
| | - Zhang Yan
- c Youth Road Hospital , Friendship Hospital , Urumqi , China
| | - Sang Guoyao
- d Clinical laboratory of Xinjiang Medical University First Affiliated Hospital , Urumqi , Xinjiang , China
| | - Zhang Zhanlin
- a Department of Epidemiology and Health Statistics, School of Public Health , Xinjiang Medical University , Urumqi , Xinjiang , China
| | - Yeledan Mahan
- a Department of Epidemiology and Health Statistics, School of Public Health , Xinjiang Medical University , Urumqi , Xinjiang , China
| | - Tuo Xiaoqing
- a Department of Epidemiology and Health Statistics, School of Public Health , Xinjiang Medical University , Urumqi , Xinjiang , China
| | - Gong Zheng
- a Department of Epidemiology and Health Statistics, School of Public Health , Xinjiang Medical University , Urumqi , Xinjiang , China
| | - Dai Jianghong
- a Department of Epidemiology and Health Statistics, School of Public Health , Xinjiang Medical University , Urumqi , Xinjiang , China
| |
Collapse
|
25
|
Geretti AM, Brook G, Cameron C, Chadwick D, French N, Heyderman R, Ho A, Hunter M, Ladhani S, Lawton M, MacMahon E, McSorley J, Pozniak A, Rodger A. British HIV Association Guidelines on the Use of Vaccines in HIV-Positive Adults 2015. HIV Med 2018; 17 Suppl 3:s2-s81. [PMID: 27568789 DOI: 10.1111/hiv.12424] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Anna Maria Geretti
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | | | | | | | | | | | | | | | | | - Mark Lawton
- Royal Liverpool University Hospital, Liverpool, UK
| | - Eithne MacMahon
- Guy's & St Thomas' NHS Foundation Trust, London, UK.,King's College London, London, UK
| | | | - Anton Pozniak
- Chelsea and Westminster Hospital, NHS Foundation Trust, London, UK
| | | |
Collapse
|
26
|
Arbyn M, Xu L, Simoens C, Martin‐Hirsch PPL, Cochrane Gynaecological, Neuro‐oncology and Orphan Cancer Group. Prophylactic vaccination against human papillomaviruses to prevent cervical cancer and its precursors. Cochrane Database Syst Rev 2018; 5:CD009069. [PMID: 29740819 PMCID: PMC6494566 DOI: 10.1002/14651858.cd009069.pub3] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Persistent infection with high-risk human papillomaviruses (hrHPV) types is causally linked with the development of cervical precancer and cancer. HPV types 16 and 18 cause approximately 70% of cervical cancers worldwide. OBJECTIVES To evaluate the harms and protection of prophylactic human papillomaviruses (HPV) vaccines against cervical precancer and HPV16/18 infection in adolescent girls and women. SEARCH METHODS We searched MEDLINE, Cochrane Central Register of Controlled Trials (CENTRAL) and Embase (June 2017) for reports on effects from trials. We searched trial registries and company results' registers to identify unpublished data for mortality and serious adverse events. SELECTION CRITERIA Randomised controlled trials comparing efficacy and safety in females offered HPV vaccines with placebo (vaccine adjuvants or another control vaccine). DATA COLLECTION AND ANALYSIS We used Cochrane methodology and GRADE to rate the certainty of evidence for protection against cervical precancer (cervical intraepithelial neoplasia grade 2 and above [CIN2+], CIN grade 3 and above [CIN3+], and adenocarcinoma-in-situ [AIS]), and for harms. We distinguished between the effects of vaccines by participants' baseline HPV DNA status. The outcomes were precancer associated with vaccine HPV types and precancer irrespective of HPV type. Results are presented as risks in control and vaccination groups and risk ratios (RR) with 95% confidence intervals in brackets. MAIN RESULTS We included 26 trials (73,428 participants). Ten trials, with follow-up of 1.3 to 8 years, addressed protection against CIN/AIS. Vaccine safety was evaluated over a period of 6 months to 7 years in 23 studies. Studies were not large enough or of sufficient duration to evaluate cervical cancer outcomes. All but one of the trials was funded by the vaccine manufacturers. We judged most included trials to be at low risk of bias. Studies involved monovalent (N = 1), bivalent (N = 18), and quadrivalent vaccines (N = 7). Most women were under 26 years of age. Three trials recruited women aged 25 and over. We summarize the effects of vaccines in participants who had at least one immunisation.Efficacy endpoints by initial HPV DNA statushrHPV negativeHPV vaccines reduce CIN2+, CIN3+, AIS associated with HPV16/18 compared with placebo in adolescent girls and women aged 15 to 26. There is high-certainty evidence that vaccines lower CIN2+ from 164 to 2/10,000 (RR 0.01 (0 to 0.05)) and CIN3+ from 70 to 0/10,000 (RR 0.01 (0.00 to 0.10). There is moderate-certainty evidence that vaccines reduce the risk of AIS from 9 to 0/10,000 (RR 0.10 (0.01 to 0.82).HPV vaccines reduce the risk of any CIN2+ from 287 to 106/10,000 (RR 0.37 (0.25 to 0.55), high certainty) and probably reduce any AIS lesions from 10 to 0/10,000 (RR 0.1 (0.01 to 0.76), moderate certainty). The size of reduction in CIN3+ with vaccines differed between bivalent and quadrivalent vaccines (bivalent: RR 0.08 (0.03 to 0.23), high certainty; quadrivalent: RR 0.54 (0.36 to 0.82), moderate certainty). Data in older women were not available for this comparison.HPV16/18 negativeIn those aged 15 to 26 years, vaccines reduce CIN2+ associated with HPV16/18 from 113 to 6 /10,000 (RR 0.05 (0.03 to 0.10). In women 24 years or older the absolute and relative reduction in the risk of these lesions is smaller (from 45 to 14/10,000, (RR 0.30 (0.11 to 0.81), moderate certainty). HPV vaccines reduce the risk of CIN3+ and AIS associated with HPV16/18 in younger women (RR 0.05 (0.02 to 0.14), high certainty and RR 0.09 (0.01 to 0.72), moderate certainty, respectively). No trials in older women have measured these outcomes.Vaccines reduce any CIN2+ from 231 to 95/10,000, (RR 0.41 (0.32 to 0.52)) in younger women. No data are reported for more severe lesions.Regardless of HPV DNA statusIn younger women HPV vaccines reduce the risk of CIN2+ associated with HPV16/18 from 341 to 157/10,000 (RR 0.46 (0.37 to 0.57), high certainty). Similar reductions in risk were observed for CIN3+ associated with HPV16/18 (high certainty). The number of women with AIS associated with HPV16/18 is reduced from 14 to 5/10,000 with HPV vaccines (high certainty).HPV vaccines reduce any CIN2+ from 559 to 391/10,000 (RR 0.70 (0.58 to 0.85, high certainty) and any AIS from 17 to 5/10,000 (RR 0.32 (0.15 to 0.67), high certainty). The reduction in any CIN3+ differed by vaccine type (bivalent vaccine: RR 0.55 (0.43 to 0.71) and quadrivalent vaccine: RR 0.81 (0.69 to 0.96)).In women vaccinated at 24 to 45 years of age, there is moderate-certainty evidence that the risks of CIN2+ associated with HPV16/18 and any CIN2+ are similar between vaccinated and unvaccinated women (RR 0.74 (0.52 to 1.05) and RR 1.04 (0.83 to 1.30) respectively). No data are reported in this age group for CIN3+ or AIS.Adverse effectsThe risk of serious adverse events is similar between control and HPV vaccines in women of all ages (669 versus 656/10,000, RR 0.98 (0.92 to 1.05), high certainty). Mortality was 11/10,000 in control groups compared with 14/10,000 (9 to 22) with HPV vaccine (RR 1.29 [0.85 to 1.98]; low certainty). The number of deaths was low overall but there is a higher number of deaths in older women. No pattern in the cause or timing of death has been established.Pregnancy outcomesAmong those who became pregnant during the studies, we did not find an increased risk of miscarriage (1618 versus 1424/10,000, RR 0.88 (0.68 to 1.14), high certainty) or termination (931 versus 838/10,000 RR 0.90 (0.80 to 1.02), high certainty). The effects on congenital abnormalities and stillbirths are uncertain (RR 1.22 (0.88 to 1.69), moderate certainty and (RR 1.12 (0.68 to 1.83), moderate certainty, respectively). AUTHORS' CONCLUSIONS There is high-certainty evidence that HPV vaccines protect against cervical precancer in adolescent girls and young women aged 15 to 26. The effect is higher for lesions associated with HPV16/18 than for lesions irrespective of HPV type. The effect is greater in those who are negative for hrHPV or HPV16/18 DNA at enrolment than those unselected for HPV DNA status. There is moderate-certainty evidence that HPV vaccines reduce CIN2+ in older women who are HPV16/18 negative, but not when they are unselected by HPV DNA status.We did not find an increased risk of serious adverse effects. Although the number of deaths is low overall, there were more deaths among women older than 25 years who received the vaccine. The deaths reported in the studies have been judged not to be related to the vaccine. Increased risk of adverse pregnancy outcomes after HPV vaccination cannot be excluded, although the risk of miscarriage and termination are similar between trial arms. Long-term of follow-up is needed to monitor the impact on cervical cancer, occurrence of rare harms and pregnancy outcomes.
Collapse
Affiliation(s)
- Marc Arbyn
- SciensanoUnit of Cancer Epidemiology, Belgian Cancer CentreJuliette Wytsmanstreet 14BrusselsBelgiumB‐1050
| | - Lan Xu
- SciensanoUnit of Cancer Epidemiology, Belgian Cancer CentreJuliette Wytsmanstreet 14BrusselsBelgiumB‐1050
| | - Cindy Simoens
- University of AntwerpLaboratory of Cell Biology and HistologyGroenenborgerlaan 171AntwerpBelgiumB‐2020
| | - Pierre PL Martin‐Hirsch
- Royal Preston Hospital, Lancashire Teaching Hospital NHS TrustGynaecological Oncology UnitSharoe Green LaneFullwoodPrestonLancashireUKPR2 9HT
| | | |
Collapse
|
27
|
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
|
28
|
Boda D, Docea AO, Calina D, Ilie MA, Caruntu C, Zurac S, Neagu M, Constantin C, Branisteanu DE, Voiculescu V, Mamoulakis C, Tzanakakis G, Spandidos DA, Drakoulis N, Tsatsakis AM. Human papilloma virus: Apprehending the link with carcinogenesis and unveiling new research avenues (Review). Int J Oncol 2018; 52:637-655. [PMID: 29393378 PMCID: PMC5807043 DOI: 10.3892/ijo.2018.4256] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 12/28/2017] [Indexed: 12/17/2022] Open
Abstract
Human papilloma viruses (HPV) are a small group of non‑enveloped viruses belonging to the Papillomaviridae family with strong similarities to polyoma viruses. The viral particles consist of a genome in the form of a circular double‑stranded DNA, encompassing eight open reading frames, as well as a non‑enveloped icosahedral capsid. HPV infection is considered the most common sexually transmitted disease in both sexes and is strongly implicated in the pathogenesis of different types of cancer. 'High‑risk' mucosal HPV types, predominantly types 16, 18, 31, 33 and 35, are associated with most cervical, penile, vulvar, vaginal, anal, oropharyngeal cancers and pre‑cancers. Screening for HPV is necessary for the prognosis and for determining treatment strategies for cancer. Novel HPV markers, including proteomic and genomic markers, as well as anti‑papillomavirus vaccines are currently available. The aim of this comprehensive review was to thoroughly present the updated information on virus development, cancer occurrence, treatment and prevention strategies, in an attempt to shed further light into the field, including novel research avenues.
Collapse
Affiliation(s)
- Daniel Boda
- Dermatology Research Laboratory, ‘Carol Davila’ University of Medicine and Pharmacy, 030167 Bucharest
- Department of Dermatology, ‘Prof. N. Paulescu’ National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest
| | | | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova
| | - Mihaela Adriana Ilie
- Dermatology Research Laboratory, ‘Carol Davila’ University of Medicine and Pharmacy, 030167 Bucharest
- Department of Biochemistry
| | - Constantin Caruntu
- Dermatology Research Laboratory, ‘Carol Davila’ University of Medicine and Pharmacy, 030167 Bucharest
- Department of Dermatology, ‘Prof. N. Paulescu’ National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest
- Department of Physiology
| | - Sabina Zurac
- Department of Pathology, ‘Carol Davila’ University of Medicine and Pharmacy, 030167 Bucharest
- Colentina University Hospital, Sector 2 19-21, Bucharest
| | - Monica Neagu
- ‘Victor Babes’ National Institute of Pathology, 050096 Bucharest
| | | | | | - Vlad Voiculescu
- Department of Dermatology and Allergology, Elias Emergency University Hospital, 011461 Bucharest, Romania
| | - Charalampos Mamoulakis
- Department of Urology, University General Hospital of Heraklion, University of Crete Medical School
| | | | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Crete
| | - Nikolaos Drakoulis
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15771 Athens
| | - Aristides M. Tsatsakis
- Laboratory of Toxicology, Medical School, University of Crete, 71003 Heraklion, Crete, Greece
| |
Collapse
|
29
|
Beck Z, Torres OB, Matyas GR, Lanar DE, Alving CR. Immune response to antigen adsorbed to aluminum hydroxide particles: Effects of co-adsorption of ALF or ALFQ adjuvant to the aluminum-antigen complex. J Control Release 2018; 275:12-19. [PMID: 29432824 DOI: 10.1016/j.jconrel.2018.02.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 02/04/2018] [Indexed: 12/12/2022]
Abstract
Aluminum salts have been used as vaccine adjuvants for >50 years, and they are currently present in at least 146 licensed vaccines worldwide. In this study we examined whether adsorption of Army Liposome Formulation (ALF) to an aluminum salt that already has an antigen adsorbed to it might result in improved immune potency of the aluminum-adsorbed antigen. ALF is composed of a family of anionic liposome-based adjuvants, in which the liposomes contain synthetic phospholipids having dimyristoyl fatty acyl groups, cholesterol and monophosphoryl lipid A (MPLA). For certain candidate vaccines, ALF has been added to aluminum hydroxide (AH) gel as a second adjuvant to form ALFA. Here we show that different methods of preparation of ALF changed the physical structures of both ALF and ALFA. Liposomes containing the saponin QS21 (ALFQ) have also been mixed with AH to form ALFQA as an effective combination. In this study, we first adsorbed one of two different antigens to AH, either tetanus toxoid conjugated to 34 copies of a hapten (MorHap), which has been used in a candidate heroin vaccine, or gp140 protein derived from the envelope protein of HIV-1. We then co-adsorbed ALF or ALFQ to the AH to form ALFA or ALFQA. In each case, the immune potency of the antigen adsorbed to AH was greatly increased by co-adsorbing either ALF or ALFQ to the AH. Based on IgG subtype and cytokine analysis by ELISPOT, ALFA induced predominately a Th2-type response and ALFQ and ALFQA each induced more balanced Th1/Th2 responses.
Collapse
Affiliation(s)
- Zoltan Beck
- U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA; Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
| | - Oscar B Torres
- U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA; Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
| | - Gary R Matyas
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
| | - David E Lanar
- Malaria Vaccine Branch, US Military Malaria Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Ave, Silver Spring, MD 20910, USA
| | - Carl R Alving
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA.
| |
Collapse
|
30
|
Leung TF, Liu APY, Lim FS, Thollot F, Oh HML, Lee BW, Rombo L, Tan NC, Rouzier R, De Simoni S, Suryakiran P, Hezareh M, Thomas F, Folschweiller N, Struyf F. Comparative immunogenicity and safety of human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine and 4vHPV vaccine administered according to two- or three-dose schedules in girls aged 9-14 years: Results to month 36 from a randomized trial. Vaccine 2018; 36:98-106. [PMID: 29174109 DOI: 10.1016/j.vaccine.2017.11.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/26/2017] [Accepted: 11/13/2017] [Indexed: 12/20/2022]
Abstract
This observer-blind study (clinicaltrials.gov NCT01462357) compared the immunogenicity and safety of two doses (2D) of the HPV-16/18 AS04-adjuvanted vaccine (2D of AS04-HPV-16/18) vs. two or three doses of the 4vHPV vaccine [2D or 3D of 4vHPV] in 1075 healthy girls aged 9-14 years. Girls were randomized (1:1:1) to receive 2D of AS04-HPV-16/18 at months (M) 0, 6 (N = 359), 2D of 4vHPV at M0, 6 (N = 358) or 3D of 4vHPV at M0, 2, 6 (N = 358). 351, 339 and 346 girls, respectively, returned for the concluding visit at M36. Superiority was demonstrated at M7 and M12; comparison of the immune response to both vaccine antigens was made between 2D of AS04-HPV-16/18 and 2D or 3D of 4vHPV at subsequent time points in the according-to-protocol immunogenicity cohort (ATP-I; N = 958 at M36) and the total vaccinated cohort (TVC: N = 1036 at M36). HPV-16/18-specific T-cell- and B-cell-mediated immune responses and safety were also investigated. At M36, anti-HPV-16/18 ELISA responses in the 2D AS04-HPV-16/18 group remained superior to those of the 2D and 3D 4vHPV groups. In the M36 TVC, geometric mean titers were 2.78-fold (HPV-16) and 6.84-fold (HPV-18) higher for 2D of AS04-HPV-16/18 vs. 2D of 4vHPV and 2.3-fold (HPV-16) and 4.14-fold (HPV-18) higher vs. 3D of 4vHPV. Results were confirmed by vaccine pseudovirion-based neutralisation assay. Numbers of circulating CD4+ T cells and B cells appeared similar across groups. Safety was in line with the known safety profiles of both vaccines. In conclusion, superior HPV-16/18 antibody responses were elicited by 2D of the AS04-HPV-16/18 compared with 2D or 3D of the 4vHPV vaccine in girls aged 9-14 years. CLINICAL TRIAL REGISTRATION NCT0146235.
Collapse
Affiliation(s)
- Ting Fan Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong.
| | - Anthony Pak-Yin Liu
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Fong Seng Lim
- National Healthcare Group Polyclinics and National University of Singapore, Singapore
| | - Franck Thollot
- Association Française de Pédiatrie Ambulatoire (AFPA), Essey Les Nancy, France
| | - Helen May Lin Oh
- Division of Infectious Disease, Changi General Hospital, Singapore
| | - Bee Wah Lee
- Mount Elizabeth Medical Centre and National University of Singapore, Singapore
| | - Lars Rombo
- Centre for Clinical Research, Sormland County Council, Uppsala University and Karolinska Institute, Stockholm, Sweden
| | - Ngiap Chuan Tan
- SingHealth Polyclinics and DUKE-NUS Graduate Medical School, Singapore, Singapore
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Zurek Munk-Madsen M, Toft L, Kube T, Richter R, Ostergaard L, Søgaard OS, Tolstrup M, Kaufmann AM. Cellular immunogenicity of human papillomavirus vaccines Cervarix and Gardasil in adults with HIV infection. Hum Vaccin Immunother 2017; 14:909-916. [PMID: 29172992 PMCID: PMC5893199 DOI: 10.1080/21645515.2017.1407896] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Human papillomavirus (HPV) infection is a frequent cause of malignant and non-malignant disease, in particular among persons with HIV. HPV serotype-specific anti L1 antibodies protect against HPV infection but little is known about prophylactic HPV vaccine-induced cell-mediated immunity against HPV in high-risk individuals. We recently showed that both HPV vaccines (Gardasil® and Cervarix®) induce solid, serological immune responses in HIV-infected persons. This study aimed to characterize HPV-specific CD4 T cells in HIV-infected HPV-vaccine recipients, T cell responses being critical for B cell activation and antibody-isotype switching. Thirty HIV-infected patients on long-term antiretroviral treatment (ART) received 3 doses of either Cervarix (n = 15) or Gardasil (n = 15) vaccine at month 0, 1.5 and 6. Cryopreserved peripheral blood mononuclear cells (PBMC) from baseline, 7 and 12 months were subjected to 24-hour stimulation with specific pools of HPV L1-peptides (HPV6, 11, 16, 18, 31 and 45) and HPV E6/E7-peptide pools (HPV6/11 and HPV16/18). Fluorescence-activated cell sorting with intracellular staining (IC-FACS) against CD4, CD154, IL-2, and IFNγ was performed. Frequencies (%) of HPV-antigen specific CD4+ T cells (CD154+/IL-2+ or CD154+/ IFNγ+) were determined. Both HPV-vaccines significantly and comparably enhanced cell-mediated vaccine L1 antigen-specific immunity in HIV-positive adults receiving ART therapy at month 7 and 12 after first vaccine dose. This suggests that the vaccines induce CD4 T cellular memory despite HIV-induced immune compromisation.
Collapse
Affiliation(s)
- Maria Zurek Munk-Madsen
- a Clinic for Gynecology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health , Berlin , Germany.,b Department of Infectious Diseases , Aarhus University Hospital , Aarhus , Denmark
| | - Lars Toft
- b Department of Infectious Diseases , Aarhus University Hospital , Aarhus , Denmark
| | - Tina Kube
- a Clinic for Gynecology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health , Berlin , Germany
| | - Rolf Richter
- a Clinic for Gynecology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health , Berlin , Germany
| | - Lars Ostergaard
- b Department of Infectious Diseases , Aarhus University Hospital , Aarhus , Denmark
| | - Ole S Søgaard
- b Department of Infectious Diseases , Aarhus University Hospital , Aarhus , Denmark
| | - Martin Tolstrup
- b Department of Infectious Diseases , Aarhus University Hospital , Aarhus , Denmark
| | - Andreas M Kaufmann
- a Clinic for Gynecology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health , Berlin , Germany
| |
Collapse
|
32
|
Bacik LC, Chung C. Human papillomavirus-associated cutaneous disease burden in human immunodeficiency virus (HIV)-positive patients: the role of human papillomavirus vaccination and a review of the literature. Int J Dermatol 2017; 57:627-634. [PMID: 29152727 DOI: 10.1111/ijd.13819] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 09/21/2017] [Accepted: 09/26/2017] [Indexed: 11/30/2022]
Abstract
Human papillomavirus (HPV) infection is related to the development of cutaneous squamous cell carcinoma, oropharyngeal carcinoma, and anogenital malignancies. Patients infected with human immunodeficiency virus (HIV) have impaired cell-mediated immunity, placing them at risk for more prolonged infection with a greater likelihood of disease expression. This presents important implications for screening and treatment of HPV in the HIV patient population. The use of prophylactic vaccines directed against HPV has been a promising clinical development, though the immunogenicity of these vaccines in the immunocompromised host and in patients with previously established HPV infections has not been well established. In this review, we describe the pathogenesis and epidemiology of HPV-related cutaneous malignancies in patients with HIV. We outline the current guidelines and recent advances in the field of HPV vaccination. It is our hope that increasing awareness of the HPV-related HIV comorbidities will lead to developments in preventative medicine capable of reducing the burden of these diseases. We recognize the importance of prevention as a primary defense against disease and hope that this article organizes and disseminates recent findings in the field of HPV-associated comorbidities in the HIV population.
Collapse
Affiliation(s)
- Lindsay C Bacik
- Department of Dermatology, Penn State Hershey Medical Center, Hershey, PA, USA
| | - Catherine Chung
- Department of Dermatology, Ohio State University, Columbus, OH, USA
| |
Collapse
|
33
|
Pierini S, Perales-Linares R, Uribe-Herranz M, Pol JG, Zitvogel L, Kroemer G, Facciabene A, Galluzzi L. Trial watch: DNA-based vaccines for oncological indications. Oncoimmunology 2017; 6:e1398878. [PMID: 29209575 DOI: 10.1080/2162402x.2017.1398878] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 10/24/2017] [Indexed: 12/16/2022] Open
Abstract
DNA-based vaccination is a promising approach to cancer immunotherapy. DNA-based vaccines specific for tumor-associated antigens (TAAs) are indeed relatively simple to produce, cost-efficient and well tolerated. However, the clinical efficacy of DNA-based vaccines for cancer therapy is considerably limited by central and peripheral tolerance. During the past decade, considerable efforts have been devoted to the development and characterization of novel DNA-based vaccines that would circumvent this obstacle. In this setting, particular attention has been dedicated to the route of administration, expression of modified TAAs, co-expression of immunostimulatory molecules, and co-delivery of immune checkpoint blockers. Here, we review preclinical and clinical progress on DNA-based vaccines for cancer therapy.
Collapse
Affiliation(s)
- Stefano Pierini
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Ovarian Cancer Research Center (OCRC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Renzo Perales-Linares
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Ovarian Cancer Research Center (OCRC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mireia Uribe-Herranz
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Ovarian Cancer Research Center (OCRC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan G Pol
- Université Paris Descartes/Paris V, France.,Université Pierre et Marie Curie/Paris VI, Paris.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, Paris, France
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France.,Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, France.,Université Pierre et Marie Curie/Paris VI, Paris.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden.,Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP; Paris, France
| | - Andrea Facciabene
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Ovarian Cancer Research Center (OCRC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, France.,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
34
|
Poljak M, Šterbenc A, Lunar MM. Prevention of human papillomavirus (HPV)-related tumors in people living with human immunodeficiency virus (HIV). Expert Rev Anti Infect Ther 2017; 15:987-999. [PMID: 29027811 DOI: 10.1080/14787210.2017.1392854] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION In comparison to their HIV-negative counterparts, people living with HIV (PLWH) have a higher prevalence of human papillomavirus (HPV) infection in various anatomical sites coupled with increased HPV persistence, higher risk of HPV-related tumors, and faster disease progression. Areas covered: Gender-neutral prevention strategies for HPV-related cancers in PLWH discussed: ABC approach, HPV vaccination, antiretroviral treatment (ART), anal cancer screening, and smoking cessation. Gender specific strategies: cervical cancer screening reduces the incidence and mortality of cervical cancer and circumcision might reduce the risk of HPV infections in men. Expert commentary: HPV-related cancer incidence has not declined (e.g. cervical cancer) and has even increased (e.g. anal cancer) in the ART era, demanding an effective HPV prevention strategy. HPV vaccination should be introduced into national prevention programs worldwide immediately because current prophylactic vaccines are safe, tolerable, and immunogenic in PLWH. HPV vaccine efficacy trials in PLWH are essential to determine the most appropriate immunization schedule. The population most at risk of anal cancer is HIV-positive men who have sex with men, who are not protected by herd immunity if only the female population is vaccinated. Unvaccinated PLWH need enhanced surveillance for early detection of HPV-related cancers and their precursors.
Collapse
Affiliation(s)
- Mario Poljak
- a Institute of Microbiology and Immunology, Faculty of Medicine , University of Ljubljana , Ljubljana , Slovenia
| | - Anja Šterbenc
- a Institute of Microbiology and Immunology, Faculty of Medicine , University of Ljubljana , Ljubljana , Slovenia
| | - Maja M Lunar
- a Institute of Microbiology and Immunology, Faculty of Medicine , University of Ljubljana , Ljubljana , Slovenia
| |
Collapse
|
35
|
Poddighe D. 2-Dose Schedule of AS04-Adjuvanted Human Papillomavirus Types 16/18 Vaccine. J Infect Dis 2017; 216:782-783. [PMID: 28934441 DOI: 10.1093/infdis/jix364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/14/2017] [Indexed: 09/21/2023] Open
Affiliation(s)
- Dimitri Poddighe
- Department of Pediatrics, ASST Melegnano e Martesana, Vizzolo Predabissi, Milano
- Department of Pediatrics, Università degli Studi di Pavia, Pavia, Italy
| |
Collapse
|
36
|
Dreyer G. Clinical implications of the interaction between HPV and HIV infections. Best Pract Res Clin Obstet Gynaecol 2017; 47:95-106. [PMID: 28958633 DOI: 10.1016/j.bpobgyn.2017.08.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 08/27/2017] [Indexed: 01/01/2023]
Abstract
HIV-related immunodeficiency has complex effects on female genital HPV, which include increased risks of infection, multiple types, persistence, reactivation and the risk to develop pre-invasive and invasive disease. Reconstitution of immunity with anti-viral drugs improves cellular immunity, but the risk of HPV-related malignancy remains higher than background incidences and presents at younger ages. Early initiation of antiretroviral therapy (ART) allows improved retention of immune memory through existing antibodies and T-cell clones and improves long-term outcomes. Suggestions of a higher risk to contract HIV if there is existing genital HPV infection are supported and explained by pathophysiological cervical changes, including inflammation. HIV-HPV interactions should influence public health decisions towards prioritising large-scale prepubertal HPV-vaccine roll-out, secondary cervical cancer prevention and early detection programmes for HIV-infected women and early initiation of ART. This chapter will also focus on special considerations for the management of women with co-infection with these two viruses and genital HPV-related diseases.
Collapse
Affiliation(s)
- Greta Dreyer
- Department Obstetrics and Gynaecology, University of Pretoria, Pretoria, South Africa.
| |
Collapse
|
37
|
Crum-Cianflone NF, Sullivan E. Vaccinations for the HIV-Infected Adult: A Review of the Current Recommendations, Part I. Infect Dis Ther 2017; 6:303-331. [PMID: 28779442 PMCID: PMC5595780 DOI: 10.1007/s40121-017-0166-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Indexed: 12/19/2022] Open
Abstract
Vaccination is a critical component for ensuring the health of those living with the human immunodeficiency virus (HIV) by protection against vaccine-preventable diseases. Since HIV-infected persons may have reduced immune responses and shorter durations of protection post-vaccination, HIV-specific guidelines have been published by global and national advisory organizations to address these potential concerns. This article provides a comprehensive review of the current guidelines and evidence-based data for vaccinating HIV-infected adults, including guidance on modified vaccine dosing and testing strategies, as well as safety considerations, to enhance protection among this vulnerable population. In the current article, part I of the two-part series, inactivated vaccines with broad indications as well as vaccines for specific risk and age groups will be discussed.
Collapse
Affiliation(s)
- Nancy F Crum-Cianflone
- Internal Medicine Department, Scripps Mercy Hospital, San Diego, CA, USA.
- Infectious Disease Division, Scripps Mercy Hospital, San Diego, CA, USA.
- Infectious Disease Division, Naval Medical Center San Diego, San Diego, CA, USA.
| | - Eva Sullivan
- Pharmacy Department, Scripps Mercy Hospital, San Diego, CA, USA
| |
Collapse
|
38
|
Hidalgo-Tenorio C, Ramírez-Taboada J, Gil-Anguita C, Esquivias J, Omar-Mohamed-Balgahata M, SamPedro A, Lopez-Ruz M, Pasquau J. Safety and immunogenicity of the quadrivalent human papillomavirus (qHPV) vaccine in HIV-positive Spanish men who have sex with men (MSM). AIDS Res Ther 2017; 14:34. [PMID: 28720147 PMCID: PMC5516305 DOI: 10.1186/s12981-017-0160-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/11/2017] [Indexed: 01/05/2023] Open
Abstract
Background Safety and immunogenicity of the quadrivalent human papillomavirus (qHPV) vaccine were evaluated in HIV-positive Spanish MSM. The prevalence of High Squamous Intraepithelial Lesions (HSIL) and genotypes of high-risk human papillomavirus (HR-HPV) were also determined, as well as risk factors associated with the presence of HR-HPV in anal mucosa. Methods This is a randomised, double blind, placebo-controlled trial of the quadrivalent HPV (qHPV) vaccine. The study enrolled from May 2012 to May 2014. Vaccine and placebo were administered at 0, 2 and 6 months (V1, V2, V3 clinical visits). Vaccine antibody titres were evaluated at 7 months. Cytology (Thin Prep® Pap Test), HPV PCR genotyping (Linear Array HPV Genotyping Test), and high-resolution anoscopy (Zeiss 150 fc© colposcope) were performed at V1. Results Patients (n = 162; mean age 37.9 years) were screened for inclusion; 14.2% had HSIL, 73.1% HR-HPV and 4.5% simultaneous infection with HPV16 and 18. Study participants (n = 129) were randomized to qHPV vaccine or placebo. The most common adverse event was injection-site pain predominating in the placebo group [the first dose (83.6% vs. 56.1%; p = 0.0001]; the second dose (87.8% vs. 98.4%; p = 0.0001); the third dose (67.7% vs. 91.9%; p = 0.0001). The vaccine did not influence either the viral load of HIV or the levels of CD4. Of those vaccinated, 76% had antibodies to HPV vs. 30.2% of those receiving placebo (p = 0.0001). In the multivariate analysis, Older age was associated with lower HR-HPV infection (RR 0.97; 95% CI 0.96–0.99), and risk factor were viral load of HIV >200 copies/µL (RR 1.42 95% CI 1.17–1.73) and early commencement of sexual activity (RR 1.35; 95% CI 1.001–1.811). Conclusions This trial showed significantly higher anti-HR-HPV antibody titres in vaccinated individuals than in unvaccinated controls. There were no serious adverse events attributable to the vaccine. In our cohort, 1 of every 7 patients had HSIL and the prevalence of combined infection by genotypes 16 and 18 was low. This suggests that patients could benefit from receiving qHPV vaccine. Older age was the main protective factor against HR-HPV infection, and non-suppressed HIV viremia was a risk factor. Clinical trial registration: ISRCTN14732216 (http://www.isrctn.com/ISRCTN14732216).
Collapse
|
39
|
Bissett SL, Godi A, Jit M, Beddows S. Seropositivity to non-vaccine incorporated genotypes induced by the bivalent and quadrivalent HPV vaccines: A systematic review and meta-analysis. Vaccine 2017. [PMID: 28633892 DOI: 10.1016/j.vaccine.2017.06.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Human papillomavirus vaccines have demonstrated remarkable efficacy against persistent infection and disease associated with vaccine-incorporated genotypes and a degree of efficacy against some genetically related, non-vaccine-incorporated genotypes. The vaccines differ in the extent of cross-protection against these non-vaccine genotypes. Data supporting the role for neutralizing antibodies as a correlate or surrogate of cross-protection are lacking, as is a robust assessment of the seroconversion rates against these non-vaccine genotypes. METHODS We performed a systematic review and meta-analysis of available data on vaccine-induced neutralizing antibody seropositivity to non-vaccine incorporated HPV genotypes. RESULTS Of 304 articles screened, 9 were included in the analysis representing ca. 700 individuals. The pooled estimate for seropositivity against HPV31 for the bivalent vaccine (86%; 95%CI 78-91%) was higher than that for the quadrivalent vaccine (61%; 39-79%; p=0.011). The pooled estimate for seropositivity against HPV45 for the bivalent vaccine (50%; 37-64%) was also higher than that for the quadrivalent vaccine (16%; 6-36%; p=0.007). Seropositivity against HPV33, HPV52 and HPV58 were similar between the vaccines. Mean seropositivity rates across non-vaccine genotypes were positively associated with the corresponding vaccine efficacy data reported from vaccine trials. CONCLUSIONS These data improve our understanding of vaccine-induced functional antibody specificity against non-vaccine incorporated genotypes and may help to parameterize vaccine-impact models and improve patient management in a post-vaccine setting.
Collapse
Affiliation(s)
- Sara L Bissett
- Virus Reference Department, Public Health England, London, UK
| | - Anna Godi
- Virus Reference Department, Public Health England, London, UK
| | - Mark Jit
- Modelling and Economics Unit, Public Health England, London, UK; Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Simon Beddows
- Virus Reference Department, Public Health England, London, UK.
| |
Collapse
|
40
|
Howitt BE, Herfs M, Tomoka T, Kamiza S, Gheit T, Tommasino M, Delvenne P, Crum CP, Milner D. Comprehensive Human Papillomavirus Genotyping in Cervical Squamous Cell Carcinomas and Its Relevance to Cervical Cancer Prevention in Malawian Women. J Glob Oncol 2017; 3:227-234. [PMID: 28717764 PMCID: PMC5493214 DOI: 10.1200/jgo.2015.001909] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
PURPOSE Cervical squamous cell carcinoma (SCC) continues to be a significant cause of cancer morbidity and is the third leading cause of cancer-related death in women worldwide. In sub-Saharan Africa, cervical cancer is not only the most common female cancer but also the leading cause of cancer-related deaths in women. Malawi, in particular, has the highest burden of cervical cancer. With the increasing use of human papillomavirus (HPV) vaccination, documenting the prevalent HPV types in those high-risk populations is necessary to both manage expectations of HPV vaccination and guide future vaccine development. MATERIALS AND METHODS In this study, we performed HPV typing on 474 cervical SCC samples and analyzed the potential impact of HPV vaccination in this population. RESULTS Ninety-seven percent of tumors were positive for at least one HPV type, and 54% harbored more than one HPV type. HPV 16 was the most common type (82%), followed by HPV 18 (34%), HPV 35 (24%), and HPV 31 (12%). A vaccine against HPV 16 and 18 would ideally prevent 53% of cervical SCC, and the nonavalent HPV vaccine (covering HPV 16, 18, 31, 33, 45, 52, and 58) would prevent 71% of cervical SCC in Malawi (assuming 100% vaccine efficacy). The main reason for a lack of coverage was high prevalence of HPV 35, which was also present as a single infection in a small subset of patients. CONCLUSION Although any HPV vaccination in this population would likely prevent a significant proportion of cervical cancer, the nonavalent vaccine would provide better coverage. Furthermore, investigation of the role of HPV 35 in this population, including possible cross-protection with other HPV types, should be pursued.
Collapse
Affiliation(s)
- Brooke E. Howitt
- Brooke E. Howitt, Christopher P. Crum, and Danny Milner, Brigham and Women’s Hospital, Boston, MA; Michael Herfs and Philippe Delvenne, University of Liege, Liege, Belgium; Tamiwe Tomoka, Steve Kamiza, and Danny Milner, Malawi College of Medicine, Blantyre, Malawi; and Tarik Gheit and Massimo Tommasino, International Agency for Research on Cancer, Lyon, France
| | - Michael Herfs
- Brooke E. Howitt, Christopher P. Crum, and Danny Milner, Brigham and Women’s Hospital, Boston, MA; Michael Herfs and Philippe Delvenne, University of Liege, Liege, Belgium; Tamiwe Tomoka, Steve Kamiza, and Danny Milner, Malawi College of Medicine, Blantyre, Malawi; and Tarik Gheit and Massimo Tommasino, International Agency for Research on Cancer, Lyon, France
| | - Tamiwe Tomoka
- Brooke E. Howitt, Christopher P. Crum, and Danny Milner, Brigham and Women’s Hospital, Boston, MA; Michael Herfs and Philippe Delvenne, University of Liege, Liege, Belgium; Tamiwe Tomoka, Steve Kamiza, and Danny Milner, Malawi College of Medicine, Blantyre, Malawi; and Tarik Gheit and Massimo Tommasino, International Agency for Research on Cancer, Lyon, France
| | - Steve Kamiza
- Brooke E. Howitt, Christopher P. Crum, and Danny Milner, Brigham and Women’s Hospital, Boston, MA; Michael Herfs and Philippe Delvenne, University of Liege, Liege, Belgium; Tamiwe Tomoka, Steve Kamiza, and Danny Milner, Malawi College of Medicine, Blantyre, Malawi; and Tarik Gheit and Massimo Tommasino, International Agency for Research on Cancer, Lyon, France
| | - Tarik Gheit
- Brooke E. Howitt, Christopher P. Crum, and Danny Milner, Brigham and Women’s Hospital, Boston, MA; Michael Herfs and Philippe Delvenne, University of Liege, Liege, Belgium; Tamiwe Tomoka, Steve Kamiza, and Danny Milner, Malawi College of Medicine, Blantyre, Malawi; and Tarik Gheit and Massimo Tommasino, International Agency for Research on Cancer, Lyon, France
| | - Massimo Tommasino
- Brooke E. Howitt, Christopher P. Crum, and Danny Milner, Brigham and Women’s Hospital, Boston, MA; Michael Herfs and Philippe Delvenne, University of Liege, Liege, Belgium; Tamiwe Tomoka, Steve Kamiza, and Danny Milner, Malawi College of Medicine, Blantyre, Malawi; and Tarik Gheit and Massimo Tommasino, International Agency for Research on Cancer, Lyon, France
| | - Philippe Delvenne
- Brooke E. Howitt, Christopher P. Crum, and Danny Milner, Brigham and Women’s Hospital, Boston, MA; Michael Herfs and Philippe Delvenne, University of Liege, Liege, Belgium; Tamiwe Tomoka, Steve Kamiza, and Danny Milner, Malawi College of Medicine, Blantyre, Malawi; and Tarik Gheit and Massimo Tommasino, International Agency for Research on Cancer, Lyon, France
| | - Christopher P. Crum
- Brooke E. Howitt, Christopher P. Crum, and Danny Milner, Brigham and Women’s Hospital, Boston, MA; Michael Herfs and Philippe Delvenne, University of Liege, Liege, Belgium; Tamiwe Tomoka, Steve Kamiza, and Danny Milner, Malawi College of Medicine, Blantyre, Malawi; and Tarik Gheit and Massimo Tommasino, International Agency for Research on Cancer, Lyon, France
| | - Danny Milner
- Brooke E. Howitt, Christopher P. Crum, and Danny Milner, Brigham and Women’s Hospital, Boston, MA; Michael Herfs and Philippe Delvenne, University of Liege, Liege, Belgium; Tamiwe Tomoka, Steve Kamiza, and Danny Milner, Malawi College of Medicine, Blantyre, Malawi; and Tarik Gheit and Massimo Tommasino, International Agency for Research on Cancer, Lyon, France
| |
Collapse
|
41
|
Haskins-Coulter T, Southern J, Andrews N, Miller E. Reactogenicity of Cervarix and Gardasil human papillomavirus (HPV) vaccines in a randomized single blind trial in healthy UK adolescent females. Hum Vaccin Immunother 2017; 13:1-9. [PMID: 28319456 DOI: 10.1080/21645515.2016.1277846] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
One hundred and ninety eight females aged 12-15 y were enrolled in an observer-blinded randomized trial to assess the immunogenicity and reactogenicity of the tetravalent HPV vaccine Gardasil® (group 2), in comparison to the bivalent HPV vaccine, Cervarix® (group 1), which was routinely offered in the national vaccination schedule at the time. Participants were blinded to treatment group until all 3 vaccinations had been given, while laboratory staff were masked during testing. For the majority of local and general reactions, recipients of both vaccines reported comparable frequencies. Local and systemic events were rarely of high severity, except for tenderness at the injection site which reached a severe level after at least one of the doses in 24% of the Cervarix® group and 7% of the Gardasil® group (p = 0.001 comparing groups). For most reactions, no dose response was recorded, except for swelling with higher reporting at dose 3 (17.7%) than dose 1 (3.1%) for Cervarix®. SAE reporting was low (n = 3) and considered unrelated to either vaccine. This paper supports the body of evidence that Gardasil® has an acceptable safety profile when compared with Cervarix® and other vaccines given in the national program.
Collapse
Affiliation(s)
- Tao Haskins-Coulter
- a Immunisation, Hepatitis and Blood Safety Department , Public Health England , London , UK
| | - Jo Southern
- a Immunisation, Hepatitis and Blood Safety Department , Public Health England , London , UK
| | - Nick Andrews
- a Immunisation, Hepatitis and Blood Safety Department , Public Health England , London , UK
| | - Elizabeth Miller
- a Immunisation, Hepatitis and Blood Safety Department , Public Health England , London , UK
| |
Collapse
|
42
|
Levin MJ, Huang S, Moscicki AB, Song LY, Read JS, Meyer WA, Saah AJ, Richardson K, Weinberg A. Four-year persistence of type-specific immunity after quadrivalent human papillomavirus vaccination in HIV-infected children: Effect of a fourth dose of vaccine. Vaccine 2017; 35:1712-1720. [PMID: 28238631 DOI: 10.1016/j.vaccine.2017.02.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/02/2017] [Accepted: 02/09/2017] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Although HIV-infected children are recommended to receive quadrivalent human papillomavirus vaccine (QHPV) there is limited information on their response to QHPV. This study in HIV-infected children, evaluated the magnitude and duration of immune responses to QHPV. This report describes type-specific serum antibody responses over a 4-to-5year period after either 3 or 4 doses of QHPV. DESIGN/METHODS HIV-infected children, ages 7-to-11years, received 3 doses of QHPV (n=96) or placebo (n=30). At 72weeks QHPV recipients received a fourth dose (n=84), while placebo recipients began the 3-dose QHPV schedule (n=27). HPV serotype-specific antibody was determined, by competitive Luminex immunoassay (cLIA) and IgG Luminex immunoassay, at 2, 3.5, and 4-to-5years after the last dose of QHPV in each treatment arm. RESULTS At 4-to-5years after the last dose of QHPV, antibody titers were significantly higher in 4-dose than in 3-dose group. However, the proportion of vaccinees with a seroresponse in the cLIA assay was not different between the two groups (86-93% for HPV types 6, 11, and 16, and 64% for HPV type 18). These results were very similar to the seroresponse rate in these HIV-infected children at 1month after completing vaccination. CONCLUSIONS Children with well-controlled HIV infection who receive 3 doses of the QHPV vaccine maintain seropositivity and antibody levels that are generally similar to children of the same age who are not HIV-infected. Antibody titer correlated strongly with low log HIV RNA, low CD8%, and high CD4%. Additionally, a fourth dose of vaccine in HIV-infected children produces a marked rise in antibody characteristic of an anamnestic response and persistence of high antibody levels. Study identification: IMPAACT P1085 (V501-021). CLINICALTRIALS.GOV identifier: NCT01206556.
Collapse
Affiliation(s)
- Myron J Levin
- Section of Pediatric Infectious Diseases, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States.
| | - Sharon Huang
- Center for Biostatistics in AIDS Research, Harvard School of Public Health, Boston, MA 02115, United States
| | - Anna-Barbara Moscicki
- Department of Pediatrics, University of California Los Angeles, Los Angeles, CA 19954, United States
| | - Lin-Ye Song
- Formerly Center for Biostatistics in AIDS Research, Harvard School of Public Health, Boston, MA 02115, United States; Office of Science, Food & Drug Administration; Silver Spring, MD 20993, United States
| | - Jennifer S Read
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, United States
| | | | | | - Kelly Richardson
- Section of Pediatric Infectious Diseases, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Adriana Weinberg
- Section of Pediatric Infectious Diseases, Departments of Pediatrics, Medicine, and Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | | |
Collapse
|
43
|
Clifford GM, de Vuyst H, Tenet V, Plummer M, Tully S, Franceschi S. Effect of HIV Infection on Human Papillomavirus Types Causing Invasive Cervical Cancer in Africa. J Acquir Immune Defic Syndr 2016; 73:332-339. [PMID: 27331659 PMCID: PMC5172520 DOI: 10.1097/qai.0000000000001113] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 05/16/2016] [Indexed: 12/31/2022]
Abstract
OBJECTIVES HIV infection is known to worsen the outcome of cervical human papillomavirus (HPV) infection and may do so differentially by HPV type. DESIGN Twenty-one studies were included in a meta-analysis of invasive cervical cancers (ICC) among women infected with HIV in Africa. METHOD Type-specific HPV DNA prevalence was compared with data from a similar meta-analysis of HIV-negative ICC using prevalence ratios (PR). RESULTS HPV detection was similar in 770 HIV-positive (91.2%) and 3846 HIV-negative (89.6%) ICC, but HIV-positive ICC harbored significantly more multiple HPV infections (PR = 1.75, 95% confidence intervals: 1.18 to 2.58), which were significantly more prevalent in ICC tested from cells than from biopsies. HPV16 was the most frequently detected type in HIV-positive ICC (42.5%), followed by HPV18 (22.2%), HPV45 (14.4%), and HPV35 (7.1%). Nevertheless, HIV-positive ICC were significantly less frequently infected with HPV16 than HIV-negative ICC (PR = 0.88, 95% confidence intervals: 0.79 to 0.99). Other high-risk types were significantly more prevalent in HIV-positive ICC, but only for HPV18 was there a significantly higher prevalence of both single and multiple infections in HIV-positive ICC. Increases for other high-risk types were primarily accounted for by multiple infections. The proportion of HPV-positive ICC estimated attributable to HPV16/18 (71.8% in HIV positive, 73.4% in HIV negative) or HPV16/18/31/33/45/52/58 (88.8%, 89.5%) was not affected by HIV. CONCLUSIONS HIV alters the relative carcinogenicity of HPV types, but prophylactic HPV16/18 vaccines may nevertheless prevent a similar proportion of ICC, irrespective of HIV infection.
Collapse
|
44
|
Epidemiology of HPV Genotypes among HIV Positive Women in Kenya: A Systematic Review and Meta-Analysis. PLoS One 2016; 11:e0163965. [PMID: 27764092 PMCID: PMC5072621 DOI: 10.1371/journal.pone.0163965] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 09/16/2016] [Indexed: 12/04/2022] Open
Abstract
Background There is a scarcity of data on the distribution of human papillomavirus (HPV) genotypes in the HIV positive population and in invasive cervical cancer (ICC) in Kenya. This may be different from genotypes found in abnormal cytology. Yet, with the advent of preventive HPV vaccines that target HPV 16 and 18, and the nonavalent vaccine targeting 90% of all ICC cases, such HPV genotype distribution data are indispensable for predicting the impact of vaccination and HPV screening on prevention. Even with a successful vaccination program, vaccinated women will still require screening to detect those who will develop ICC from other High risk (HR) HPV genotypes not prevented by current vaccines. The aim of this review is to report on the prevalence of pHR/HR HPV types and multiple pHR/HR HPV genotypes in Kenya among HIV positive women with normal, abnormal cytology and ICC. Methods PUBMED, EMBASE, SCOPUS, and PROQUEST were searched for articles on HPV infection up to August 2nd 2016. Search terms were HIV, HPV, Cervical Cancer, Incidence or Prevalence, and Kenya. Results The 13 studies included yielded a total of 2116 HIV-infected women, of which 89 had ICC. The overall prevalence of pHR/HR HPV genotypes among HIV-infected women was 64% (95%CI: 50%-77%). There was a borderline significant difference in the prevalence of pHR/HR HPV genotypes between Female Sex workers (FSW) compared to non-FSW in women with both normal and abnormal cytology. Multiple pHR/HR HPV genotypes were highly prominent in both normal cytology/HSIL and ICC. The most prevalent HR HPV genotypes in women with abnormal cytology were HPV 16 with 26%, (95%CI: 23.0%-30.0%) followed by HPV 35 and 52, with 21% (95%CI: 18%-25%) and 18% (95%CI: 15%-21%), respectively. In women with ICC, the most prevalent HPV genotypes were HPV 16 (37%; 95%CI: 28%-47%) and HPV 18 (24%; 95%CI: 16%-33%). Conclusion HPV 16/18 gains prominence as the severity of cervical disease increases, with HPV 16/18 accounting for 61% (95%CI: 50.0%-70.0%) of all ICC cases. A secondary prevention program will be necessary as this population harbors multiple pHR/HR HPV co-infections, which may not be covered by current vaccines. A triage based on FSW as an indicator may be warranted.
Collapse
|
45
|
Leng CY, Low HC, Chua LL, Chong ML, Sulaiman H, Azwa I, Roberts JM, Kamarulzaman A, Rajasuriar R, Woo YL. Human papillomavirus 16 (HPV16) and HPV52 E6-specific immunity in HIV-infected adults on combination antiretroviral therapy. HIV Med 2016; 18:321-331. [PMID: 27649852 DOI: 10.1111/hiv.12432] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2016] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Human papillomavirus (HPV)-associated cancers disproportionately affect those infected with HIV despite effective combination antiretroviral therapy (cART). The primary aim of this study was to quantify HPV16 and HPV52 E6-specific interferon (IFN)-γ enzyme-linked immunospot (ELISPOT) T-cell responses, a correlate of protective immunity, in the first year following cART initiation and subsequently in those patients with suboptimal (sIR) and optimal (oIR) immune reconstitution. METHODS Ninety-four HIV-infected patients were recruited to the study; a longitudinal cohort of patients recruited just prior to commencing cART and followed up for 48 weeks (n = 27), and a cross-sectional cohort (n = 67) consisting of patients with sIR (CD4 T-cell count < 350 cells/μL) and oIR (CD4 T-cell count > 500 cells/μL) after a minimum of 2 years on cART. Controls (n = 29) consisted of HIV-negative individuals. IFN-γ ELISPOT responses against HPV16 and HPV52 E6 were correlated to clinical characteristics, anal and oral HPV carriage, T-cell maturational subsets, markers of activation, senescence and T-regulatory cells. RESULTS HPV16 and HPV52 E6-specific T-cell responses were detected in only one of 27 patients (3.7%) during the initial phase of immune recovery. After at least 2 years of cART, those who achieved oIR had significantly higher E6-specific responses (9 of 34; 26.5%) compared with those with sIR (2 of 32; 6.3%) (P = 0.029). Apart from higher CD4 T-cell counts and lower CD4 T-cell activation, no other immunological correlates were associated with the detection of HPV16 and HPV52 E6-specific responses. CONCLUSIONS HPV16 and HPV52 E6-specific IFN-γ T-cell responses, a correlate of protective immunity, were detected more frequently among HIV-infected patients who achieved optimal immune recovery on cART (26.5%) compared with those with suboptimal recovery (6.3%).
Collapse
Affiliation(s)
- C Y Leng
- Centre of Excellence for Research in AIDS (CERiA), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - H C Low
- Centre of Excellence for Research in AIDS (CERiA), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - L L Chua
- University of Malaya Cancer Research Institute (UMCRI), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - M L Chong
- Centre of Excellence for Research in AIDS (CERiA), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - H Sulaiman
- Centre of Excellence for Research in AIDS (CERiA), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - I Azwa
- Centre of Excellence for Research in AIDS (CERiA), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - J M Roberts
- Douglass Hanly Moir Pathology, Macquarie Park, New South Wales, Australia
| | - A Kamarulzaman
- Centre of Excellence for Research in AIDS (CERiA), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - R Rajasuriar
- Centre of Excellence for Research in AIDS (CERiA), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Y L Woo
- Centre of Excellence for Research in AIDS (CERiA), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,University of Malaya Cancer Research Institute (UMCRI), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
46
|
Brotherton JML, Jit M, Gravitt PE, Brisson M, Kreimer AR, Pai SI, Fakhry C, Monsonego J, Franceschi S. Eurogin Roadmap 2015: How has HPV knowledge changed our practice: Vaccines. Int J Cancer 2016; 139:510-7. [PMID: 26916230 PMCID: PMC7388730 DOI: 10.1002/ijc.30063] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 02/02/2016] [Accepted: 02/11/2016] [Indexed: 12/18/2022]
Abstract
This review is one of two complementary reviews that have been prepared in the framework of the Eurogin Roadmap 2015 to evaluate how knowledge about HPV is changing practices in HPV infection and disease control through vaccination and screening. In this review of HPV vaccine knowledge, we present the most significant findings of the past year which have contributed to our knowledge of the two HPV prophylactic vaccines currently in widespread use and about the recently licensed nonavalent HPV vaccine. Whereas anal cancer is dealt with in the companion mini-review on screening, we also review here the rapidly evolving evidence regarding HPV-associated head and neck cancer and priority research areas.
Collapse
Affiliation(s)
- Julia M L Brotherton
- National HPV Vaccination Program Register, VCS Registries, East Melbourne, Vic, Australia
- School of Population and Global Health, University of Melbourne, Vic, Australia
| | - Mark Jit
- Modelling and Economics Unit, Public Health England, London, United Kingdom
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Patti E Gravitt
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Marc Brisson
- Département de Médecine Sociale et Préventive, Université Laval, Québec, QC, Canada
| | - Aimée R Kreimer
- Infections & Immunoepidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD
| | - Sara I Pai
- Department of Surgery, Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | - Carole Fakhry
- Department of Otolaryngology Head and Neck Surgery, Johns Hopkins School of Medicine, Johns Hopkins Outpatient Center, Baltimore, MD
| | | | - Silvia Franceschi
- International Agency for Research on Cancer, 69372 Lyon cedex 08, France
| |
Collapse
|
47
|
Park LS, Tate JP, Sigel K, Rimland D, Crothers K, Gibert C, Rodriguez-Barradas MC, Goetz MB, Bedimo RJ, Brown ST, Justice AC, Dubrow R. Time trends in cancer incidence in persons living with HIV/AIDS in the antiretroviral therapy era: 1997-2012. AIDS 2016; 30:1795-806. [PMID: 27064994 PMCID: PMC4925286 DOI: 10.1097/qad.0000000000001112] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Utilizing the Veterans Aging Cohort Study, the largest HIV cohort in North America, we conducted one of the few comprehensive comparisons of cancer incidence time trends in HIV-infected (HIV+) versus uninfected persons during the antiretroviral therapy (ART) era. DESIGN Prospective cohort study. METHODS We followed 44 787 HIV+ and 96 852 demographically matched uninfected persons during 1997-2012. We calculated age-, sex-, and race/ethnicity-standardized incidence rates and incidence rate ratios (IRR, HIV+ versus uninfected) over four calendar periods with incidence rate and IRR period trend P values for cancer groupings and specific cancer types. RESULTS We observed 3714 incident cancer diagnoses in HIV+ and 5760 in uninfected persons. The HIV+ all-cancer crude incidence rate increased between 1997-2000 and 2009-2012 (P trend = 0.0019). However, after standardization, we observed highly significant HIV+ incidence rate declines for all cancer (25% decline; P trend <0.0001), AIDS-defining cancers (55% decline; P trend <0.0001), nonAIDS-defining cancers (NADC; 15% decline; P trend = 0.0003), and nonvirus-related NADC (20% decline; P trend <0.0001); significant IRR declines for all cancer (from 2.0 to 1.6; P trend <0.0001), AIDS-defining cancers (from 19 to 5.5; P trend <0.0001), and nonvirus-related NADC (from 1.4 to 1.2; P trend = 0.049); and borderline significant IRR declines for NADC (from 1.6 to 1.4; P trend = 0.078) and virus-related NADC (from 4.9 to 3.5; P trend = 0.071). CONCLUSION Improved HIV care resulting in improved immune function most likely contributed to the HIV+ incidence rate and the IRR declines. Further promotion of early and sustained ART, improved ART regimens, reduction of traditional cancer risk factor (e.g. smoking) prevalence, and evidence-based screening could contribute to future cancer incidence declines among HIV+ persons.
Collapse
Affiliation(s)
| | - Janet P. Tate
- Veterans Affairs Connecticut Healthcare System, West Haven, CT
- Yale School of Medicine, New Haven, CT
| | - Keith Sigel
- Icahn School of Medicine at Mt. Sinai, New York, NY
| | - David Rimland
- Atlanta Veterans Affairs Medical Center, Atlanta, GA; Emory University School of Medicine, Atlanta, GA
| | | | - Cynthia Gibert
- Washington DC Veterans Affairs Medical Center, Washington, DC; George Washington University School of Medicine and Health Sciences, Washington, DC
| | | | - Matthew Bidwell Goetz
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA; David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Roger J. Bedimo
- Veterans Affairs North Texas Healthcare System, Dallas, TX; University of Texas Southwestern Medical Center, Dallas, TX
| | - Sheldon T. Brown
- Icahn School of Medicine at Mt. Sinai, New York, NY
- James J. Peters Veterans Affairs Medical Center, New York, NY
| | - Amy C. Justice
- Veterans Affairs Connecticut Healthcare System, West Haven, CT
- Yale School of Medicine, New Haven, CT
| | - Robert Dubrow
- Yale School of Medicine, New Haven, CT
- Yale School of Public Health, New Haven, CT
| |
Collapse
|
48
|
Zhai L, Tumban E. Gardasil-9: A global survey of projected efficacy. Antiviral Res 2016; 130:101-9. [PMID: 27040313 DOI: 10.1016/j.antiviral.2016.03.016] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/29/2016] [Indexed: 02/07/2023]
Abstract
Human papillomaviruses (HPVs) are the causative agents of human neoplasias such as warts and cancers. There are ∼19 HPV types associated with cancers, which has made it very challenging for first generation HPV vaccines to offer complete protection against all cancer-causing HPV types. Recently, a second generation HPV vaccine, Gardasil-9, has been approved to protect against more HPV types. Worldwide, Gardasil-9 will protect against HPV types associated with ∼90% of cervical cancer case in women and 80-95% of other HPV-associated anogenital cancers in both men and women. However, due to variation in HPV-type specific prevalence and distribution, the vaccine will offer different percentages of protection in different geographical regions; Gardasil-9 will offer protection against HPV types associated with ∼87.7% of cervical cancers in Asia, 91.7% in Africa, 92% in North America, 90.9% in Europe, 89.5% in Latin America & the Caribbean, and 86.5% in Australia. Because of this, Pap smear screening and testing for HPV types not included in Gardasil-9 will need to continue, especially in HIV/AIDS patients. In order to achieve complete protection against all HPV types that cause cervical cancer, a third-generation HPV vaccine is needed.
Collapse
Affiliation(s)
- Lukai Zhai
- Department of Biological Sciences, Michigan Technological University, Houghton, MI 49931, United states
| | - Ebenezer Tumban
- Department of Biological Sciences, Michigan Technological University, Houghton, MI 49931, United states.
| |
Collapse
|
49
|
Gee J, Weinbaum C, Sukumaran L, Markowitz LE. Quadrivalent HPV vaccine safety review and safety monitoring plans for nine-valent HPV vaccine in the United States. Hum Vaccin Immunother 2016; 12:1406-17. [PMID: 27029786 PMCID: PMC4964727 DOI: 10.1080/21645515.2016.1168952] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Quadrivalent human papillomavirus (4vHPV) vaccine was licensed for use in the United States in 2006 and through 2015 was the predominate HPV vaccine used. With the exception of syncope, a known preventable adverse event after any injected vaccination, both pre-licensure and post-licensure 4vHPV safety data have been reassuring with no confirmed safety signals identified. Nine-valent HPV vaccine (9vHPV) was licensed in 2014. This review includes post-licensure 4vHPV safety findings published to date that have informed the US vaccination program; these data will inform US safety monitoring and evaluation for 9vHPV.
Collapse
Affiliation(s)
- Julianne Gee
- a Division of Healthcare and Quality Promotion, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention , Atlanta , GA , USA
| | - Cindy Weinbaum
- a Division of Healthcare and Quality Promotion, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention , Atlanta , GA , USA
| | - Lakshmi Sukumaran
- a Division of Healthcare and Quality Promotion, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention , Atlanta , GA , USA
| | - Lauri E Markowitz
- b Division of Viral Diseases, National Center Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta , GA , USA
| |
Collapse
|
50
|
Faust H, Toft L, Sehr P, Müller M, Bonde J, Forslund O, Østergaard L, Tolstrup M, Dillner J. Human Papillomavirus neutralizing and cross-reactive antibodies induced in HIV-positive subjects after vaccination with quadrivalent and bivalent HPV vaccines. Vaccine 2016; 34:1559-1565. [PMID: 26896686 DOI: 10.1016/j.vaccine.2016.02.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/18/2016] [Accepted: 02/04/2016] [Indexed: 01/10/2023]
Abstract
Ninety-one HIV-infected individuals (61 men and 30 women) were randomized to vaccination either with quadrivalent (Gardasil™) or bivalent (Cervarix™) HPV vaccine. Neutralizing and specific HPV-binding serum antibodies were measured at baseline and 12 months after the first vaccine dose. Presence of neutralizing and binding antibodies had good agreement (average Kappa for HPV types 6, 11, 16, 18, 31, 33 and 45 was 0.65). At baseline, 88% of subjects had antibodies against at least one genital HPV. Following vaccination with Cervarix™, all subjects became seropositive for HPV16 and 18. After Gardasil™ vaccination, 96% of subjects seroconverted for HPV16 and 73% for HPV18. Levels of HPV16-specific antibodies were <1 international unit (IU) in 87% of study subjects before vaccination but >10IU in 85% of study subjects after vaccination. Antibodies against non-vaccine HPV types appeared after Gardasil™ vaccination for >50% of vaccinated females for HPV 31, 35 and 73 and for >50% of Cervarix™-vaccinated females for HPV 31, 33, 35, 45, 56 and 58. Cross-reactivity with non-genital HPV types was also detected. In conclusion, HIV-infected subjects responded to HPV vaccination with induction of neutralizing antibodies against both vaccine and non-vaccine types.
Collapse
Affiliation(s)
- Helena Faust
- Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Lars Toft
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Peter Sehr
- European Molecular Biology Laboratory, Heidelberg, Germany
| | | | - Jesper Bonde
- Molecular Pathology Laboratory, Department of Pathology, Copenhagen University Hospital, Hvidovre, Denmark
| | - Ola Forslund
- Department of Laboratory Medicine, Lund University, Malmö, Sweden
| | - Lars Østergaard
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Martin Tolstrup
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Joakim Dillner
- Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|