1
|
Munk C, Reinholdt K, Kjaer AK, Hemmingsen CH, Ørnskov D, Iftner T, Waldstrøm M, Kjaer SK. Prevalence of Human Papillomavirus (HPV) and HPV Type Distribution in Penile Samples in Young Men in Denmark: Results 10 Years After Implementation of a Girls-Only HPV Vaccination Program. J Infect Dis 2024; 230:949-956. [PMID: 38470214 DOI: 10.1093/infdis/jiae068] [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: 10/13/2023] [Accepted: 01/09/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND In Denmark, a girls-only human papillomavirus (HPV) vaccination program was initiated in 2008-2009. The study aim was to assess the HPV prevalence and type distribution in younger men prior to HPV vaccination in men. METHODS The study population was younger men who attended information days regarding military service. At random days (2019-2020), 280 men were included. We collected questionnaire data regarding risk factors for HPV infection and a penile swab for HPV testing. We compared results in this study with those from a previous study of young men (2006-2007). RESULTS The majority of participants (94%) were 18-20 years old. The median number of lifetime sexual partners was 4. Altogether, 130 men (46.4%) were HPV positive. No infections with HPV types 6, 11, 16, 18, 31, and 45 were detected. The most frequent type was HPV-51 (detected in 11.1%). Comparison showed that the odds of high-risk HPV type infection were higher in 2019-2020 (prevalence odds ratio [POR], 1.7 [95% confidence interval {CI}, 1.1-2.7]) compared with 2006-2007. In contrast, the odds were lower (POR, 0.3 [95% CI, .1-.6]) for HPV types targeted by the 9-valent HPV vaccine. CONCLUSIONS The multicohort girls-only vaccination program has to a large degree protected young men against the HPV types included in the licensed vaccines. This does not speak against gender-neutral vaccination as the HPV prevalence is still high, although consisting largely of less carcinogenic HPV types.
Collapse
Affiliation(s)
- Christian Munk
- Unit of Virus, Lifestyle and Genes, Danish Cancer Institute, Copenhagen
| | | | - Alexander K Kjaer
- Unit of Virus, Lifestyle and Genes, Danish Cancer Institute, Copenhagen
| | | | | | - Thomas Iftner
- Institute of Medical Virology, University of Tübingen, Tübingen, Germany
| | | | - Susanne K Kjaer
- Unit of Virus, Lifestyle and Genes, Danish Cancer Institute, Copenhagen
- Department of Gynecology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
2
|
Shah PT, Wu Z, Ma R, Wu C. Genetic diversity, variation and recombination among the human papillomaviruses (HPVs) genomes isolated in China: a comparative genomic and phylogenetic analysis. Pathog Glob Health 2024; 118:505-518. [PMID: 39263878 PMCID: PMC11441022 DOI: 10.1080/20477724.2024.2401273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024] Open
Abstract
Human papillomaviruses (HPVs) are widespread, sexually transmitted group of viruses that infect most individuals at some stage, causing genital warts and cancers. They are members of the Papillomaviridae family, which contains about 400 HPV types. China is among the high HPV burden countries with reported infections of multiple HPV types, accounting for 17.3% of global deaths and 18.2% of global new cases. Thus, understanding the genetic variation and geographic diversity characteristics of HPVs isolated in China is critical for global HPV prevention strategies. Thus, we analyzed the available HPV genome sequences isolated in China that grouped into two categories (alpha- and gamma-papillomaviruses) based on full-length genomes. The most common were HPV-16, -6, -58, and -52 respectively. In addition, four of the novel strains isolated in China, e.g. TG550, JDFY01, CH2, and L55 clustered with the HPV-mSK 159, 244, 201, and 200 respectively. Our phylogeographic network analysis indicated that the L55, TG550, and CH2 are genetically identical to the mSK 200, 046, and 201 respectively, while JDFY01 appeared separately, connected to the mSK-040 following five mutational steps. Also, we found ten recombination events among HPV-6/11 types within their E1, E2, E7, L1/L2 proteins, and Long Control Region ORFs. We achieved the consensus amino acid sequences of HPV proteins and found a conserved stretch of amino acids within E5A of all HPVs circulating in China. These findings offer valued insights into the genetic relationships, distribution, and evolution of the HPVs in China that may assist in adapting effective HPV preventive measures.
Collapse
Affiliation(s)
- Pir Tariq Shah
- Faculty of Medicine, School of Basic Medical Sciences, Dalian University of Technology, Dalian, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, China
| | - Zhenyong Wu
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Ruilan Ma
- Radiotherapy Oncology Department, The Second Affiliated Hospital of the Dalian Medical University, Dalian, China
| | - Chengjun Wu
- Faculty of Medicine, School of Basic Medical Sciences, Dalian University of Technology, Dalian, China
| |
Collapse
|
3
|
Mirhassani R, Seyedjafari E, Vaziri B. A validated polyclonal antiserum-based immunoassay for assessment of HPV 16 L1 relative potency. Biologicals 2024; 85:101742. [PMID: 38340596 DOI: 10.1016/j.biologicals.2023.101742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 07/28/2022] [Accepted: 12/16/2023] [Indexed: 02/12/2024] Open
Abstract
Vaccine potency is typically evaluated using an assay that acts as a surrogate for biological activity. Although in vivo vaccines better represent human immunological responses, in vitro assays are preferred due to lower variability, higher throughput, easier validation and ethical considerations. In in vitro determination of Human Papillomavirus (HPV), Virus-like particle (VLP) vaccine potency currently depends on monoclonal antibody assays. However, these reagents are hard to obtain and currently are not available commercially. In this work, a polyclonal antiserum-based immunoassay was developed to evaluate the relative potency of Alhydrogel formulated HPV 16 VLPs. The repeatability and specificity were evaluated, and found that the assay was sensitive to small amounts of non-VLP HPV 16 L1 proteins. Finally, the assay was tested in comparison to the mouse effective dose 50 (ED50) assay on a limited number of batches. The agreement between these results suggests this test as a suitable surrogate for the in vivo test.
Collapse
Affiliation(s)
- Reihaneh Mirhassani
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Ehsan Seyedjafari
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran.
| | - Behrouz Vaziri
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
4
|
Shiro R, Tsunoda I. [HPV vaccinations and cervical cancer in Japan: Flaws in alleged experimental evidence for molecular mimicry and animal models of HPV vaccine-induced "adverse reactions "]. Uirusu 2024; 74:17-28. [PMID: 39617449 DOI: 10.2222/jsv.74.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2024]
Abstract
In Japan, around 3,000 females have died of cervical cancer annually. Cervical cancer is caused by sexually transmitted infections of the human papillomavirus (HPV); it is preventable by vaccinations against oncogenic HPV types, such as types 16 and 18. In Japan, the proactive recommendations for HPV vaccinations were suspended in June 2013 due to public concerns about neuropsychological "diverse symptoms" following HPV vaccinations. The suspension resulted in the vaccination rate less than 1% among the eligible population. In April 2022, the Japanese government resumed the proactive HPV vaccination recommendations after 9 years of the suspension. Many Japanese people, however, still have concerns about the "diverse symptoms" as adverse reactions following HPV vaccinations. In this article, we critically evaluated the manuscripts cited in the HPV vaccine lawsuits in Japan, including computational and animal studies, which have been used as the theoretical basis for how HPV vaccinations could induce the "diverse symptoms." We concluded that all the manuscripts had scientific flaws, which should not be used to prove that HPV vaccinations could induce the adverse reactions. We wish that our current evaluation would contribute to eliminating baseless concerns about HPV vaccines, eventually increasing the rate of HPV vaccinations in Japan.
Collapse
Affiliation(s)
- Reona Shiro
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine
| | - Ikuo Tsunoda
- Department of Microbiology, Kindai University Faculty of Medicine
| |
Collapse
|
5
|
Hu YM, Bi ZF, Zheng Y, Zhang L, Zheng FZ, Chu K, Li YF, Chen Q, Quan JL, Hu XW, Huang XC, Zhu KX, Wang-Jiang YH, Jiang HM, Zang X, Liu DL, Yang CL, Pan HX, Zhang QF, Su YY, Huang SJ, Sun G, Huang WJ, Huang Y, Wu T, Zhang J, Xia NS. Immunogenicity and safety of an Escherichia coli-produced human papillomavirus (types 6/11/16/18/31/33/45/52/58) L1 virus-like-particle vaccine: a phase 2 double-blind, randomized, controlled trial. Sci Bull (Beijing) 2023; 68:2448-2455. [PMID: 37743201 DOI: 10.1016/j.scib.2023.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/23/2023] [Accepted: 09/14/2023] [Indexed: 09/26/2023]
Abstract
The Escherichia coli-produced human papillomavirus (HPV) 16/18 bivalent vaccine (Cecolin) has received prequalification by the World Health Organization based on its high efficacy and good safety profile. We aimed to evaluate the immunogenicity and safety of the second-generation nonavalent HPV 6/11/16/18/31/33/45/52/58 vaccine (Cecolin 9) through the randomized, blinded phase 2 clinical trial. Eligible healthy women aged 18-45 years were randomly (1:1) allocated to receive three doses of 1.0 mL (270 µg) of Cecolin 9 or placebo with a 0-1-6-month schedule. The primary endpoint was the seroconversion rate and geometric mean titer of neutralizing antibodies (nAbs) one month after the full vaccination course (month 7). The secondary endpoint was the safety profile including solicited adverse reactions occurring within 7 d, adverse events (AEs) occurring within 30 d after each dose, and serious adverse events (SAEs) occurring during the 7-month follow-up period. In total, 627 volunteers were enrolled and randomly assigned to Cecolin 9 (n = 313) or placebo (n = 314) group in Jiangsu Province, China. Almost all participants in the per-protocol set for immunogenicity (PPS-I) seroconverted for nAbs against all the nine HPV types at month 7, while two failed to seroconvert for HPV 11 and one did not seroconvert for HPV 52. The incidence rates of total AEs in the Cecolin 9 and placebo groups were 80.8% and 72.9%, respectively, with the majority of them being mild and recovering shortly. None of the SAEs were considered related to vaccination. In conclusion, the E. coli-produced 9-valent HPV (9vHPV) vaccine candidate was well tolerated and immunogenic, which warrants further efficacy studies in larger populations.
Collapse
Affiliation(s)
- Yue-Mei Hu
- Jiangsu Provincial Center for Disease Control and Prevention, Public Health Research Institute of Jiangsu Province, Nanjing 210009, China
| | - Zhao-Feng Bi
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Ya Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Li Zhang
- National Institutes for Food and Drug Control, Beijing 102629, China
| | | | - Kai Chu
- Jiangsu Provincial Center for Disease Control and Prevention, Public Health Research Institute of Jiangsu Province, Nanjing 210009, China
| | - Ya-Fei Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Qi Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Jia-Li Quan
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Xiao-Wen Hu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Xing-Cheng Huang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Kong-Xin Zhu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Ya-Hui Wang-Jiang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Han-Min Jiang
- Dongtai Center for Disease Control and Prevention, Yancheng 224200, China
| | - Xia Zang
- Dongtai Center for Disease Control and Prevention, Yancheng 224200, China
| | - Dong-Lin Liu
- Dongtai Center for Disease Control and Prevention, Yancheng 224200, China
| | - Chang-Lin Yang
- Dongtai Center for Disease Control and Prevention, Yancheng 224200, China
| | - Hong-Xing Pan
- Jiangsu Provincial Center for Disease Control and Prevention, Public Health Research Institute of Jiangsu Province, Nanjing 210009, China
| | - Qiu-Fen Zhang
- Xiamen Innovax Biotech Company, Xiamen 361027, China
| | - Ying-Ying Su
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Shou-Jie Huang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Guang Sun
- Xiamen Innovax Biotech Company, Xiamen 361027, China.
| | - Wei-Jin Huang
- National Institutes for Food and Drug Control, Beijing 102629, China.
| | - Yue Huang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China.
| | - Ting Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China.
| | - Jun Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| | - Ning-Shao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, China
| |
Collapse
|
6
|
Chakraborty S, Ramasubbu K, Banerjee M, Balaji MP, Vinayagam Y, V DR. A systematic review on the molecular and clinical association between Human Papillomavirus and Human Immunodeficiency Virus co-infection in Head, Neck and Oral squamous cell carcinoma. Rev Med Virol 2023; 33:e2462. [PMID: 37280764 DOI: 10.1002/rmv.2462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/27/2023] [Accepted: 05/23/2023] [Indexed: 06/08/2023]
Abstract
Head and neck cancer, one of the most commonly prevalent malignancies globally is a complex category of tumours that comprises cancers of the oral cavity, pharynx, and larynx. A specific subgroup of such cancers has been found with some unique chromosomal, therapeutic, and epidemiologic traits with the possibility of affecting via co-infection. About 25% of all head and neck cancers in the population are human papillomavirus infection (HPV)-associated, typically developing in the oropharynx, which comprises the tonsils. In the period of efficient combined antiviral treatment, HPV-positive oral cancers are also becoming a significant contributor to illness and fatality for Human Immunodeficiency Virus (HIV)-infected persons. Although the prevalence and historical background of oral HPV transmission are not thoroughly understood, it seems likely that oral HPV transmission is relatively frequent in HIV-infected people when compared to the overall population. Therefore, there is a need to understand the mechanisms leading to this co-infection, as there is very little research related to that. Hence, this study mainly focus on the therapeutical and biomedical analysis of HPV and HIV co-infection in the above-mentioned cancer, including oral squamous cell carcinoma.
Collapse
Affiliation(s)
- Shreya Chakraborty
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Kanagavalli Ramasubbu
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Manosi Banerjee
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Menaka Priya Balaji
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Yamini Vinayagam
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Devi Rajeswari V
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
7
|
Kannampuzha S, Gopalakrishnan AV, Padinharayil H, Alappat RR, Anilkumar KV, George A, Dey A, Vellingiri B, Madhyastha H, Ganesan R, Ramesh T, Jayaraj R, Prabakaran DS. Onco-Pathogen Mediated Cancer Progression and Associated Signaling Pathways in Cancer Development. Pathogens 2023; 12:770. [PMID: 37375460 DOI: 10.3390/pathogens12060770] [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: 03/11/2023] [Revised: 05/20/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Infection with viruses, bacteria, and parasites are thought to be the underlying cause of about 8-17% of the world's cancer burden, i.e., approximately one in every five malignancies globally is caused by an infectious pathogen. Oncogenesis is thought to be aided by eleven major pathogens. It is crucial to identify microorganisms that potentially act as human carcinogens and to understand how exposure to such pathogens occur as well as the following carcinogenic pathways they induce. Gaining knowledge in this field will give important suggestions for effective pathogen-driven cancer care, control, and, ultimately, prevention. This review will mainly focus on the major onco-pathogens and the types of cancer caused by them. It will also discuss the major pathways which, when altered, lead to the progression of these cancers.
Collapse
Affiliation(s)
- Sandra Kannampuzha
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Hafiza Padinharayil
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur 680596, India
| | - Reema Rose Alappat
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur 680596, India
- Post Graduate and Research Department of Zoology, Maharajas College, Ernakulam 682011, India
| | - Kavya V Anilkumar
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur 680596, India
- Post Graduate and Research Department of Zoology, Maharajas College, Ernakulam 682011, India
| | - Alex George
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur 680596, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata 700073, India
| | - Balachandar Vellingiri
- Stem Cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda 151401, India
| | - Harishkumar Madhyastha
- Department of Cardiovascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Raja Ganesan
- Institute for Liver and Digestive Diseases, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Thiyagarajan Ramesh
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Rama Jayaraj
- Jindal Institute of Behavioral Sciences (JIBS), Jindal Global Institution of Eminence Deemed to Be University, Sonipat 131001, India
- Director of Clinical Sciences, Northern Territory Institute of Research and Training, Darwin, NT 0909, Australia
| | - D S Prabakaran
- Department of Radiation Oncology, College of Medicine, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju 28644, Republic of Korea
- Department of Biotechnology, Ayya Nadar Janaki Ammal College, Srivilliputhur Main Road, Sivakasi 626124, India
| |
Collapse
|
8
|
Nelson CW, Mirabello L. Human papillomavirus genomics: Understanding carcinogenicity. Tumour Virus Res 2023; 15:200258. [PMID: 36812987 PMCID: PMC10063409 DOI: 10.1016/j.tvr.2023.200258] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/01/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Human papillomavirus (HPV) causes virtually all cervical cancers and many cancers at other anatomical sites in both men and women. However, only 12 of 448 known HPV types are currently classified as carcinogens, and even the most carcinogenic type - HPV16 - only rarely leads to cancer. HPV is therefore necessary but insufficient for cervical cancer, with other contributing factors including host and viral genetics. Over the last decade, HPV whole genome sequencing has established that even fine-scale within-type HPV variation influences precancer/cancer risks, and that these risks vary by histology and host race/ethnicity. In this review, we place these findings in the context of the HPV life cycle and evolution at various levels of viral diversity: between-type, within-type, and within-host. We also discuss key concepts necessary for interpreting HPV genomic data, including features of the viral genome; events leading to carcinogenesis; the role of APOBEC3 in HPV infection and evolution; and methodologies that use deep (high-coverage) sequencing to characterize within-host variation, as opposed to relying on a single representative (consensus) sequence. Given the continued high burden of HPV-associated cancers, understanding HPV carcinogenicity remains important for better understanding, preventing, and treating cancers attributable to infection.
Collapse
Affiliation(s)
- Chase W Nelson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, 20850, USA; Institute for Comparative Genomics, American Museum of Natural History, New York, NY, 10024, USA.
| | - Lisa Mirabello
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, 20850, USA.
| |
Collapse
|
9
|
Prabhu PR, Carter JJ, Galloway DA. B Cell Responses upon Human Papillomavirus (HPV) Infection and Vaccination. Vaccines (Basel) 2022; 10:vaccines10060837. [PMID: 35746445 PMCID: PMC9229470 DOI: 10.3390/vaccines10060837] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 02/01/2023] Open
Abstract
Infection with human papillomavirus (HPV) is the necessary cause of cervical cancer. Availability of vaccines against HPV makes it a highly preventable disease. HPV vaccines act through type-specific neutralizing antibodies produced by antigen-specific plasma cells known as long-lived plasma cells (LLPC). However, just as any other vaccine, success of HPV vaccine is attributed to the immunologic memory that it builds, which is largely attained through generation and maintenance of a class of B cells named memory B cells (Bmem). Both LLPCs and Bmems are important in inducing and maintaining immune memory and it is therefore necessary to understand their role after HPV vaccination to better predict outcomes. This review summarizes current knowledge of B-cell responses following HPV vaccination and natural infection, including molecular signatures associated with these responses.
Collapse
|
10
|
Giannella L, Delli Carpini G, Di Giuseppe J, Bogani G, Gardella B, Monti E, Liverani CA, Ghelardi A, Insinga S, Montanari M, Raspagliesi F, Spinillo A, Vercellini P, Roncella E, Ciavattini A. Trend of HPV 16/18 Genotypes in Cervical Intraepithelial Neoplasia Grade 3: Data for 2007-2018. Infect Drug Resist 2021; 14:3763-3771. [PMID: 34557001 PMCID: PMC8453441 DOI: 10.2147/idr.s326851] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/31/2021] [Indexed: 11/23/2022] Open
Abstract
Aim In the post-vaccination era, the starting age and time intervals of cervical screening could change (older age and longer screening intervals). This scenario may be achieved by significantly reducing human papillomavirus (HPV) 16/18 prevalence (genotypes included in the current vaccines). In this regard, assessing the trend over time of these HPV infections in high-grade cervical lesions can provide information on the objective. The present study aimed to evaluate the trend of HPV 16/18 over the years 2007–2018 in women with cervical intraepithelial neoplasia (CIN) grade 3. Methods This is a retrospective multi-institutional study including HPV genotyped and unvaccinated women under 30 with CIN3. The sample was divided into the following periods: 2007–2010, 2011–2014, 2015–2018. HPV genotypes were grouped in genotypes 16/18, genotypes 31/33/35/52/58/67 (genetically related to HPV16), genotypes 39/45/59/68/70 (genetically related to HPV18), genotypes 31/33/45/52/58 (high-risk types included in the nonavalent vaccine), possibly carcinogenic HPV (genotypes 26/30/53/67/70/73/82/85), low-risk HPV (genotypes 6/11/40/42/43/44/54/55/61). The trend between periods and HPV genotypes was measured using the Cochran–Armitage test for trend. Results The final analysis included 474 participants. HPV 16/18 prevalence decreased significantly over the years (77.8% vs 68.9% vs 66.0%, respectively, Ptrend=0.027). Possibly carcinogenic HPV (genotypes 26/30/53/67/70/73/82/85) showed a significant negative prevalence trend over time (4.9% vs 1.1% vs 1.3%, respectively, Ptrend=0.046). Finally, there was a significant positive trend over the years for high-risk HPV genotypes 31/33/45/52/58 in women under 25 (9.9% vs 17.0% vs 24.0%, respectively, Ptrend=0.048). Conclusion The prevalence of CIN3 lesions related to HPV 16/18 genotypes decreased over time from 2007 to 2018. These data highlight a herd effect of the HPV vaccine. However, fifteen years after HPV vaccine introduction, we are still a long way from herd immunity. The increase in high-risk types 31/33/45/52/58 will need to be reassessed when the nonavalent vaccine impact will be more reliable.
Collapse
Affiliation(s)
- Luca Giannella
- Woman's Health Sciences Department, Gynecologic Section, Polytechnic University of Marche, Ancona, Italy
| | - Giovanni Delli Carpini
- Woman's Health Sciences Department, Gynecologic Section, Polytechnic University of Marche, Ancona, Italy
| | - Jacopo Di Giuseppe
- Woman's Health Sciences Department, Gynecologic Section, Polytechnic University of Marche, Ancona, Italy
| | - Giorgio Bogani
- Gynecologic Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Barbara Gardella
- Department of Obstetrics and Gynecology, Fondazione IRCCS Policlinico San Matteo, Università degli Studi di Pavia, Pavia, Italy
| | - Ermelinda Monti
- Gynaecology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Carlo Antonio Liverani
- Gynaecology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Alessandro Ghelardi
- Azienda Usl Toscana Nord-Ovest, UOC Ostetricia e Ginecologia, Ospedale Apuane, Massa, Italy
| | - Salvatore Insinga
- Woman's Health Sciences Department, Gynecologic Section, Polytechnic University of Marche, Ancona, Italy
| | - Michele Montanari
- Woman's Health Sciences Department, Gynecologic Section, Polytechnic University of Marche, Ancona, Italy
| | - Francesco Raspagliesi
- Gynecologic Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Arsenio Spinillo
- Department of Obstetrics and Gynecology, Fondazione IRCCS Policlinico San Matteo, Università degli Studi di Pavia, Pavia, Italy
| | - Paolo Vercellini
- Gynaecology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Elena Roncella
- Gynaecology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Andrea Ciavattini
- Woman's Health Sciences Department, Gynecologic Section, Polytechnic University of Marche, Ancona, Italy
| |
Collapse
|
11
|
Pedroza-Saavedra A, Rodriguez-Ocampo AN, Salazar-Piña A, Perez-Morales AC, Chihu-Amparan L, Maldonado-Gama M, Cruz-Valdez A, Esquivel-Guadarrama F, Gutierrez-Xicotencatl L. Differential Antibody Response against Conformational and Linear Epitopes of the L1 Proteins from Human Papillomavirus Types 16/18 Is Observed in Vaccinated Women or with Uterine Cervical Lesions. Vaccines (Basel) 2021; 9:vaccines9050442. [PMID: 34063178 PMCID: PMC8147477 DOI: 10.3390/vaccines9050442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/19/2021] [Accepted: 04/23/2021] [Indexed: 11/16/2022] Open
Abstract
Antibodies against the Human Papillomavirus (HPV) L1 protein are associated with past infections and related to the evolution of the disease, whereas antibodies against L1 Virus-Like Particles (VLPs) are used to follow the neutralizing antibody response in vaccinated women. In this study, serum antibodies against conformational (VLPs) and linear epitopes of HPV16/18 L1 protein were assessed to distinguish HPV-vaccinated women from those naturally infected or those with uterine cervical lesions. The VLPs-16/18 were generated in baculovirus, and L1 proteins were obtained from denatured VLPs. Serum antibodies against VLPs and L1 proteins were evaluated by ELISA. The ELISA-VLPs and ELISA-L1 16/18 assays were validated with a vaccinated women group by ROC analysis and the regression analysis to distinguish the different populations of female patients. The anti-VLPs-16/18 and anti-L1-16/18 antibodies effectively detect vaccinated women (AUC = 1.0/0.79, and 0.94/0.84, respectively). The regression analysis showed that anti-VLPs-16/18 and anti-L1-16/18 antibodies were associated with the vaccinated group (OR = 2.11 × 108/16.50 and 536.0/49.2, respectively). However, only the anti-L1-16 antibodies were associated with the high-grade lesions and cervical cancer (CIN3/CC) group (OR = 12.18). In conclusion, our results suggest that anti-VLPs-16/18 antibodies are effective and type-specific to detect HPV-vaccinated women, but anti-L1-16 antibodies better differentiate the CIN3/CC group. However, a larger population study is needed to validate these results.
Collapse
Affiliation(s)
- Adolfo Pedroza-Saavedra
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, 62100 Cuernavaca, Mexico; (A.P.-S.); (A.C.P.-M.); (L.C.-A.); (M.M.-G.)
| | | | - Azucena Salazar-Piña
- Facultad de Nutrición, Universidad Autónoma del Estado de Morelos, 62100 Cuernavaca, Mexico;
| | - Aislinn Citlali Perez-Morales
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, 62100 Cuernavaca, Mexico; (A.P.-S.); (A.C.P.-M.); (L.C.-A.); (M.M.-G.)
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, 62100 Cuernavaca, Mexico;
| | - Lilia Chihu-Amparan
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, 62100 Cuernavaca, Mexico; (A.P.-S.); (A.C.P.-M.); (L.C.-A.); (M.M.-G.)
| | - Minerva Maldonado-Gama
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, 62100 Cuernavaca, Mexico; (A.P.-S.); (A.C.P.-M.); (L.C.-A.); (M.M.-G.)
| | - Aurelio Cruz-Valdez
- Centro de Investigación en Salud Poblacional, Instituto Nacional de Salud Pública, 62100 Cuernavaca, Mexico;
| | | | - Lourdes Gutierrez-Xicotencatl
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, 62100 Cuernavaca, Mexico; (A.P.-S.); (A.C.P.-M.); (L.C.-A.); (M.M.-G.)
- Correspondence: ; Tel.: +52-77-7329-3086
| |
Collapse
|
12
|
Carse S, Bergant M, Schäfer G. Advances in Targeting HPV Infection as Potential Alternative Prophylactic Means. Int J Mol Sci 2021; 22:2201. [PMID: 33672181 PMCID: PMC7926419 DOI: 10.3390/ijms22042201] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/15/2021] [Accepted: 02/19/2021] [Indexed: 01/22/2023] Open
Abstract
Infection by oncogenic human papillomavirus (HPV) is the primary cause of cervical cancer and other anogenital cancers. The majority of cervical cancer cases occur in low- and middle- income countries (LMIC). Concurrent infection with Human Immunodeficiency Virus (HIV) further increases the risk of HPV infection and exacerbates disease onset and progression. Highly effective prophylactic vaccines do exist to combat HPV infection with the most common oncogenic types, but the accessibility to these in LMIC is severely limited due to cost, difficulties in accessing the target population, cultural issues, and maintenance of a cold chain. Alternative preventive measures against HPV infection that are more accessible and affordable are therefore also needed to control cervical cancer risk. There are several efforts in identifying such alternative prophylactics which target key molecules involved in early HPV infection events. This review summarizes the current knowledge of the initial steps in HPV infection, from host cell-surface engagement to cellular trafficking of the viral genome before arrival in the nucleus. The key molecules that can be potentially targeted are highlighted, and a discussion on their applicability as alternative preventive means against HPV infection, with a focus on LMIC, is presented.
Collapse
Affiliation(s)
- Sinead Carse
- International Centre for Genetic Engineering and Biotechnology (ICGEB) Cape Town, Observatory 7925, South Africa;
- Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Division of Medical Biochemistry and Structural Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
| | - Martina Bergant
- Laboratory for Environmental and Life Sciences, University of Nova Gorica, Vipavska 13, 5000 Nova Gorica, Slovenia;
| | - Georgia Schäfer
- International Centre for Genetic Engineering and Biotechnology (ICGEB) Cape Town, Observatory 7925, South Africa;
- Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Division of Medical Biochemistry and Structural Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
| |
Collapse
|
13
|
Mugo N, Eckert LO, Odero L, Gakuo S, Ngure K, Celum C, Baeten JM, Barnabas RV, Wald A. Antibody responses to prophylactic quadrivalent human papillomavirus vaccine at 48 months among HIV-infected girls and boys ages 9-14 in Kenya, Africa. Vaccine 2021; 39:4751-4758. [PMID: 33485644 DOI: 10.1016/j.vaccine.2020.12.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 11/05/2020] [Accepted: 12/07/2020] [Indexed: 02/04/2023]
Abstract
OBJECTIVES HIV infected children remain at increased risk of HPV associated malignancies as they initiate sexual activity. Though they mount a vigorous immune response to the quadrivalent human papillomavirus (QHPV-6, -11,-16, and -18; Gardasil®) vaccine, durability of the immune response is uncertain. We assessed antibody responses to HPV 6, -11, -16 and -18 for up to 48 months following administration of quadrivalent human papillomavirus vaccine in HIV-infected girls and boys ages 9-14 years in Kenya. DESIGN Of 178 girls and boys who had previously received three doses of the quadrivalent HPV vaccine, 176 enrolled into extended follow up for 4 years. HPV antibodies to -6, -11, -16 and -18 were measured at 24, 36 and 48 months after the first vaccine dose using the total immunoglobulin G immunoassay (IgG LIA). We evaluated the magnitude and trend in HPV vaccine response and the effect of plasma HIV-1 RNA on HPV vaccine response from month 24 to month 48 of follow up. RESULTS At re-enrollment, 24 months after initial vaccination, median age of participants was 14 years (range 11-17); 167 (95%) were receiving antiretroviral therapy and 110 (66%) had plasma HIV RNA < 40 copies/mL. The rate of HPV seropositivity at 48 months was 83% for HPV-6; 80% for HPV-11; 90% for HPV-16; and 77% for HPV-18. There was a plateau in mean log10 HPV-specific antibody titer between month 24 and 48. The mean log10 HPV-type specific antibody titer for children with undetectable HIV viral load (<40) at the time of vaccination consistently remained higher for the 48 months of follow up compared to children with detectable viral load. CONCLUSION Children with HIV infection may retain long term antibody response following HPV immunization. Further work to define whether HIV-infected children are protected from HPV acquisition with low levels of HPV antibodies is needed.
Collapse
Affiliation(s)
- Nelly Mugo
- Kenya Medical Research Institute, Center for Clinical Research, Kenya; Department of Global Health, University of Washington, Seattle, WA, USA; Partners in Health Research and Development, Kenya.
| | - Linda O Eckert
- Department of Global Health, University of Washington, Seattle, WA, USA; Department of Obstetrics and Gynaecology, University of Washington, WA, USA
| | - Lydia Odero
- Partners in Health Research and Development, Kenya
| | | | - Kenneth Ngure
- Department of Global Health, University of Washington, Seattle, WA, USA; Department of Community Health, Jomo Kenyatta University of Agriculture and Technology, Kenya
| | - Connie Celum
- Department of Global Health, University of Washington, Seattle, WA, USA; Departments of Medicine, University of Washington, Seattle, WA, USA; Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Jared M Baeten
- Department of Global Health, University of Washington, Seattle, WA, USA; Departments of Medicine, University of Washington, Seattle, WA, USA; Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Ruanne V Barnabas
- Department of Global Health, University of Washington, Seattle, WA, USA; Departments of Medicine, University of Washington, Seattle, WA, USA; Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Anna Wald
- Departments of Medicine, University of Washington, Seattle, WA, USA; Department of Epidemiology, University of Washington, Seattle, WA, USA; Department of Laboratory Medicine, University of Washington, Seattle, WA, USA; Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
14
|
Pattyn J, Panicker G, Willhauck‐Fleckenstein M, Van Keer S, Téblick L, Pieters Z, Tjalma WAA, Matheeussen V, Van Damme P, Waterboer T, Unger ER, Vorsters A. Comparison of a VLP-based and GST-L1-based multiplex immunoassay to detect vaccine-induced HPV-specific antibodies in first-void urine. J Med Virol 2020; 92:3774-3783. [PMID: 32266996 PMCID: PMC7687076 DOI: 10.1002/jmv.25841] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/27/2020] [Accepted: 03/28/2020] [Indexed: 12/22/2022]
Abstract
Vaccine-induced human papillomavirus (HPV) antibodies originating from cervicovaginal secretions were recently shown to be detectable in first-void (FV) urine. This presents a novel opportunity for noninvasive sampling to monitor HPV antibody status in women participating in large epidemiological studies and HPV vaccine trials. With a view towards method optimization, this study compared the measurement of HPV antibodies in FV urine using a multiplex L1/L2 virus-like particles (VLP)-based ELISA (M4ELISA) with previously reported results using a glutathione S-transferase (GST)-L1-based immunoassay (GST-L1-MIA). We tested 53 paired FV urine and serum samples from 19- to 26-year-old healthy women, unvaccinated (n = 17) or vaccinated with either the bivalent or quadrivalent HPV-vaccine during adolescence (n = 36). HPV6/11/16/18 antibodies were measured using M4ELISA and compared with GST-L1-MIA results. Inter-assay and inter-specimen correlations were examined using the Spearman's rank test (rs). As expected, lower HPV antibody concentrations were found in FV urine than in serum. Vaccinated women had significantly higher HPV6/11/16/18 antibody levels in both FV urine and serum compared with those unvaccinated (M4ELISA; FV urine P = .0003; serum P ≤ .0001). HPV antibody levels in FV urine and serum showed a significant positive correlation (M4ELISA anti-HPV6/11/16/18, rs = 0.85/0.86/0.91/0.79, P ≤ .001). Despite assay differences, there was moderate to good correlation between M4ELISA and GST-L1-MIA (FV urine anti-HPV6/11/16/18, rs = 0.86/0.83/0.89/0.53, P ≤ .0001; serum anti-HPV6/11/16/18, rs = 0.93/0.89/0.94/0.75, P ≤ .0001). FV urine HPV antibody detection is comparable with both assays, further supporting this noninvasive sampling method as a possible option for HPV vaccine assessment. Approaches to improve the sensitivity and larger studies are warranted to determine the feasibility of FV urine for vaccine-induced HPV antibody detection.
Collapse
Affiliation(s)
- Jade Pattyn
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of AntwerpAntwerpBelgium
| | - Gitika Panicker
- Division of High‐Consequence Pathogens and PathologyNational Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention (CDC)AtlantaGeorgia
| | - Martina Willhauck‐Fleckenstein
- Infections and Cancer Epidemiology Group, Infections, Inflammation and Cancer Research Program, German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Severien Van Keer
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of AntwerpAntwerpBelgium
| | - Laura Téblick
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of AntwerpAntwerpBelgium
| | - Zoë Pieters
- Centre for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of AntwerpAntwerpBelgium
- Centre for Statistics, I‐Biostat, Hasselt UniversityHasseltBelgium
| | - Wiebren A. A. Tjalma
- Multidisciplinary Breast Clinic, Unit Gynaecologic Oncology, Department of Obstetrics and GynaecologyAntwerp University HospitalAntwerpBelgium
- Molecular Imaging, Pathology, Radiotherapy, Oncology (MIPRO), Faculty of Medicine and Health Sciences, University of AntwerpAntwerpBelgium
| | - Veerle Matheeussen
- Department of MicrobiologyAntwerp University Hospital (UZA)AntwerpBelgium
- Department of Medical Microbiology (LMM)Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of AntwerpAntwerpBelgium
- Department of Medical BiochemistryFaculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of AntwerpAntwerpBelgium
| | - Pierre Van Damme
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of AntwerpAntwerpBelgium
| | - Tim Waterboer
- Infections and Cancer Epidemiology Group, Infections, Inflammation and Cancer Research Program, German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Elizabeth R. Unger
- Division of High‐Consequence Pathogens and PathologyNational Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention (CDC)AtlantaGeorgia
| | - Alex Vorsters
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of AntwerpAntwerpBelgium
| |
Collapse
|
15
|
Hu Y, Zhang X, He Y, Ma Z, Xie Y, Lu X, Xu Y, Zhang Y, Jiang Y, Xiao H, Struyf F, Folschweiller N, Jiang J, Poncelet S, Karkada N, Jastorff A, Borys D. Long‐term persistence of immune response to the AS04‐adjuvanted HPV‐16/18 vaccine in Chinese girls aged 9‐17 years: Results from an 8‐9‐year follow‐up phase III open‐label study. Asia Pac J Clin Oncol 2020; 16:392-399. [PMID: 32780946 PMCID: PMC7754390 DOI: 10.1111/ajco.13398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 05/21/2020] [Indexed: 11/29/2022]
Abstract
Aim In 9‐17‐year‐old Chinese girls, the AS04‐adjuvanted HPV‐16/18 vaccine (AS04‐HPV‐16/18) given as three‐dose schedule induced high antibody levels, which were noninferior 1 month after the third dose to those observed in 18‐25‐year‐old Chinese women in a large efficacy study. We assessed the persistence of antibodies 8‐9 years after vaccination in the same subjects. Methods This follow‐up phase III, open‐label study (NCT03355820) included subjects who had received three doses of AS04‐HPV‐16/18 in the initial trial (NCT00996125). Serum antibody concentrations were assessed by ELISA and compared to antibody persistence observed in 18‐25‐year‐old Chinese women 6 years after first vaccination in the efficacy study (NCT00779766). Results Out of the 227 enrolled subjects, 223 were included in the per‐protocol immunogenicity analysis. Mean interval from first AS04‐HPV‐16/18 dose to blood sampling was 101.4 months (8.5 years). For antibodies against HPV‐16 and ‐18, 8.5 years after first vaccine dose all subjects remained seropositive and antibody. Geometric mean concentrations (GMCs) were 1236.3 (95% confidence interval [CI]: 1121.8; 1362.4) and 535.6 (95% CI: 478.6; 599.4) ELISA Units/mL, respectively. These seropositivity rates and antibody GMCs were higher than those observed 6 years after first vaccination of 18‐25‐year‐old women. Conclusion Sustained anti‐HPV‐16 and ‐18 immune responses were observed 8‐9 years after AS04‐HPV‐16/18 vaccination of 9‐17 year‐old Chinese girls that were higher than the ones observed 6 years after first vaccination in Chinese adult women in whom AS04‐HPV‐16/18 efficacy against cervical intraepithelial neoplasia of grade ≥2 was demonstrated.
Collapse
Affiliation(s)
- Yuemei Hu
- Jiangsu Province Center for Disease Control and Prevention Jiangsu China
| | - Xiang Zhang
- Taizhou City Center for Disease Control and Prevention Taizhou China
| | - Yilin He
- Taizhou City Center for Disease Control and Prevention Taizhou China
| | - Zhilong Ma
- Taizhou City Center for Disease Control and Prevention Taizhou China
| | - Yan Xie
- Taizhou City Center for Disease Control and Prevention Taizhou China
| | - Xiangbin Lu
- Taizhou City Center for Disease Control and Prevention Taizhou China
| | - Yabin Xu
- Taizhou City Center for Disease Control and Prevention Taizhou China
| | - Yanqiu Zhang
- Taizhou City Center for Disease Control and Prevention Taizhou China
| | - Yunyu Jiang
- Taizhou City Center for Disease Control and Prevention Taizhou China
| | - Hui Xiao
- Taizhou City Center for Disease Control and Prevention Taizhou China
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Pattinson DJ, Apte SH, Wibowo N, Rivera-Hernandez T, Groves PL, Middelberg APJ, Doolan DL. Chimeric Virus-Like Particles and Capsomeres Induce Similar CD8 + T Cell Responses but Differ in Capacity to Induce CD4 + T Cell Responses and Antibody Responses. Front Immunol 2020; 11:564627. [PMID: 33133076 PMCID: PMC7550421 DOI: 10.3389/fimmu.2020.564627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/25/2020] [Indexed: 12/01/2022] Open
Abstract
Despite extensive research, the development of an effective malaria vaccine remains elusive. The induction of robust and sustained T cell and antibody response by vaccination is an urgent unmet need. Chimeric virus-like particles (VLPs) are a promising vaccine platform. VLPs are composed of multiple subunit capsomeres which can be rapidly produced in a cost-effective manner, but the ability of capsomeres to induce antigen-specific cellular immune responses has not been thoroughly investigated. Accordingly, we have compared chimeric VLPs and their sub-unit capsomeres for capacity to induce CD8+ and CD4+ T cell and antibody responses. We produced chimeric murine polyomavirus VLPs and capsomeres each incorporating defined CD8+ T cell, CD4+ T cell or B cell repeat epitopes derived from Plasmodium yoelii CSP. VLPs and capsomeres were evaluated using both homologous or heterologous DNA prime/boost immunization regimens for T cell and antibody immunogenicity. Chimeric VLP and capsomere vaccine platforms induced robust CD8+ T cell responses at similar levels which was enhanced by a heterologous DNA prime. The capsomere platform was, however, more efficient at inducing CD4+ T cell responses and less efficient at inducing antigen-specific antibody responses. Our data suggest that capsomeres, which have significant manufacturing advantages over VLPs, should be considered for diseases where a T cell response is the desired outcome.
Collapse
Affiliation(s)
- David J Pattinson
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,Centre for Molecular Therapeutics, Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, QLD, Australia
| | - Simon H Apte
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Nani Wibowo
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Tania Rivera-Hernandez
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Penny L Groves
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Anton P J Middelberg
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia.,School of Chemical Engineering, The University of Adelaide, Adelaide, SA, Australia
| | - Denise L Doolan
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,Centre for Molecular Therapeutics, Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
17
|
Taghinezhad-S S, Keyvani H, Bermúdez-Humarán LG, Donders GGG, Fu X, Mohseni AH. Twenty years of research on HPV vaccines based on genetically modified lactic acid bacteria: an overview on the gut-vagina axis. Cell Mol Life Sci 2020; 78:1191-1206. [PMID: 32979054 PMCID: PMC7519697 DOI: 10.1007/s00018-020-03652-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 09/03/2020] [Accepted: 09/16/2020] [Indexed: 10/27/2022]
Abstract
Most cervical cancer (CxCa) are related to persistent infection with high-risk human papillomavirus (HR-HPV) in the cervical mucosa, suggesting that an induction of mucosal cell-mediated immunity against HR-HPV oncoproteins can be a promising strategy to fight HPV-associated CxCa. From this perspective, many pre-clinical and clinical trials have proved the potential of lactic acid bacteria (LAB) genetically modified to deliver recombinant antigens to induce mucosal, humoral and cellular immunity in the host. Altogether, the outcomes of these studies suggest that there are several key factors to consider that may offer guidance on improvement protein yield and improving immune response. Overall, these findings showed that oral LAB-based mucosal HPV vaccines expressing inducible surface-anchored antigens display a higher potential to induce particularly specific systemic and mucosal cytotoxic cellular immune responses. In this review, we describe all LAB-based HPV vaccine investigations by reviewing databases from international studies between 2000 and 2020. Our aim is to promote the therapeutic HPV vaccines knowledge and to complete the gaps in this field to empower scientists worldwide to make proper decisions regarding the best strategies for the development of therapeutic HPV vaccines.
Collapse
Affiliation(s)
- Sedigheh Taghinezhad-S
- Department of Microbiology, Faculty of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, 1477893855, Iran
| | - Hossein Keyvani
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | | | - Gilbert G G Donders
- Department of Obstetrics and Gynaecology, Antwerp University Hospital, Antwerp, Belgium.,Femicare Clinical Research for Women, Tienen, Belgium
| | - Xiangsheng Fu
- Department of Gastroenterology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Amir Hossein Mohseni
- Department of Microbiology, Faculty of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, 1477893855, Iran.
| |
Collapse
|
18
|
Pattyn J, Van Keer S, Téblick L, Van Damme P, Vorsters A. Non-invasive Assessment of Vaccine-Induced HPV Antibodies via First-Void Urine. Front Immunol 2020; 11:1657. [PMID: 32849573 PMCID: PMC7419594 DOI: 10.3389/fimmu.2020.01657] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/22/2020] [Indexed: 12/26/2022] Open
Abstract
The potential of first-void (FV) urine as a non-invasive method to monitor human papillomavirus (HPV) vaccination has been reported, mainly focusing on urine as a sample to assess HPV DNA. Besides HPV DNA, vaccine-induced HPV antibodies originating from cervicovaginal secretions were recently shown to be detectable in FV urine as well. This presents a novel opportunity for non-invasive sampling to monitor HPV antibody status in women participating in large epidemiological studies and HPV vaccine trials. The simultaneous assessment of both HPV infection and immunogenicity on a non-invasive, readily obtained sample is particularly attractive.
Collapse
Affiliation(s)
- Jade Pattyn
- Faculty of Medicine and Health Sciences, Centre for the Evaluation of Vaccination, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Severien Van Keer
- Faculty of Medicine and Health Sciences, Centre for the Evaluation of Vaccination, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Laura Téblick
- Faculty of Medicine and Health Sciences, Centre for the Evaluation of Vaccination, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Pierre Van Damme
- Faculty of Medicine and Health Sciences, Centre for the Evaluation of Vaccination, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Alex Vorsters
- Faculty of Medicine and Health Sciences, Centre for the Evaluation of Vaccination, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
19
|
Watanabe Y, Seto Y, Oikawa R, Nakazawa T, Furuya H, Matsui H, Hosono S, Noike M, Inoue A, Yamamoto H, Itoh F, Wada K. Mouthwash-Based Highly Sensitive Pyro-Genotyping for Nine Sexually Transmitted Human Papilloma Virus Genotypes. Int J Mol Sci 2020; 21:ijms21103697. [PMID: 32456291 PMCID: PMC7279261 DOI: 10.3390/ijms21103697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/20/2020] [Accepted: 05/22/2020] [Indexed: 01/05/2023] Open
Abstract
Human papillomavirus (HPV) is a common sexually transmitted infection worldwide, which spreads via contact with infected genital, anal, and oral/pharyngeal areas (oral sex) owing to diverse manners of sexual intercourse. In this study, we devised an oral HPV detection method using mouthwash waste fluids that causes less psychological resistance to visiting the outpatient otolaryngology departments. We successfully detected only the specific unique reverse sequencing probe (using pyro-genotyping) and identified the nine genotypes of HPV targeted for vaccination by pyrosequencing the mouthwash waste fluids of non-head and neck cancer patient volunteers (n = 52). A relatively large number (11/52) of mouthwash waste fluids tested positive for HPV (21.2%; genotype 6, n = 1; 11, n = 1; 16, n = 1; and 18, n = 8). These results surpassed the sensitivity observed testing the same specimens using the conventional method (1/52, 1.9%). Our method (pyro-genotyping) was developed using nine HPV genotypes targeted for vaccination and the results were highly sensitive compared to those of the conventional method. This less expensive, high-throughput, and simple method can be used for detecting oral HPV infection with fewer socio-psychological barriers.
Collapse
Affiliation(s)
- Yoshiyuki Watanabe
- Department of Internal Medicine, Kawasaki Rinko General Hospital, Kawasaki 210-0806, Japan; (Y.S.); (T.N.); (H.F.); (H.M.); (S.H.); (M.N.); (A.I.)
- Department of Otolaryngology, Toho University Omori Medical Center, Tokyo 143-8540, Japan; (R.O.); (H.Y.); (F.I.)
- Correspondence: ; Tel.: +81-44-977-8111; Fax: +81-44-976-5805
| | - Yukiko Seto
- Department of Internal Medicine, Kawasaki Rinko General Hospital, Kawasaki 210-0806, Japan; (Y.S.); (T.N.); (H.F.); (H.M.); (S.H.); (M.N.); (A.I.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan;
| | - Ritsuko Oikawa
- Department of Otolaryngology, Toho University Omori Medical Center, Tokyo 143-8540, Japan; (R.O.); (H.Y.); (F.I.)
| | - Takara Nakazawa
- Department of Internal Medicine, Kawasaki Rinko General Hospital, Kawasaki 210-0806, Japan; (Y.S.); (T.N.); (H.F.); (H.M.); (S.H.); (M.N.); (A.I.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan;
| | - Hanae Furuya
- Department of Internal Medicine, Kawasaki Rinko General Hospital, Kawasaki 210-0806, Japan; (Y.S.); (T.N.); (H.F.); (H.M.); (S.H.); (M.N.); (A.I.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan;
| | - Hidehito Matsui
- Department of Internal Medicine, Kawasaki Rinko General Hospital, Kawasaki 210-0806, Japan; (Y.S.); (T.N.); (H.F.); (H.M.); (S.H.); (M.N.); (A.I.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan;
| | - Sachiko Hosono
- Department of Internal Medicine, Kawasaki Rinko General Hospital, Kawasaki 210-0806, Japan; (Y.S.); (T.N.); (H.F.); (H.M.); (S.H.); (M.N.); (A.I.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan;
| | - Mika Noike
- Department of Internal Medicine, Kawasaki Rinko General Hospital, Kawasaki 210-0806, Japan; (Y.S.); (T.N.); (H.F.); (H.M.); (S.H.); (M.N.); (A.I.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan;
| | - Akiko Inoue
- Department of Internal Medicine, Kawasaki Rinko General Hospital, Kawasaki 210-0806, Japan; (Y.S.); (T.N.); (H.F.); (H.M.); (S.H.); (M.N.); (A.I.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan;
| | - Hiroyuki Yamamoto
- Department of Otolaryngology, Toho University Omori Medical Center, Tokyo 143-8540, Japan; (R.O.); (H.Y.); (F.I.)
| | - Fumio Itoh
- Department of Otolaryngology, Toho University Omori Medical Center, Tokyo 143-8540, Japan; (R.O.); (H.Y.); (F.I.)
| | - Kota Wada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan;
| |
Collapse
|
20
|
Fernandes Q, Gupta I, Vranic S, Al Moustafa AE. Human Papillomaviruses and Epstein-Barr Virus Interactions in Colorectal Cancer: A Brief Review. Pathogens 2020; 9:pathogens9040300. [PMID: 32325943 PMCID: PMC7238043 DOI: 10.3390/pathogens9040300] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/05/2020] [Accepted: 04/07/2020] [Indexed: 02/06/2023] Open
Abstract
Human papillomaviruses (HPVs) and the Epstein-Barr virus (EBV) are the most common oncoviruses, contributing to approximately 10%-15% of all malignancies. Oncoproteins of high-risk HPVs (E5 and E6/E7), as well as EBV (LMP1, LMP2A and EBNA1), play a principal role in the onset and progression of several human carcinomas, including head and neck, cervical and colorectal. Oncoproteins of high-risk HPVs and EBV can cooperate to initiate and/or enhance epithelial-mesenchymal transition (EMT) events, which represents one of the hallmarks of cancer progression and metastasis. Although the role of these oncoviruses in several cancers is well established, their role in the pathogenesis of colorectal cancer is still nascent. This review presents an overview of the most recent advances related to the presence and role of high-risk HPVs and EBV in colorectal cancer, with an emphasis on their cooperation in colorectal carcinogenesis.
Collapse
Affiliation(s)
- Queenie Fernandes
- College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; (Q.F.); (I.G.)
- Biomedical Research Centre, Qatar University, Doha 2713, Qatar
| | - Ishita Gupta
- College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; (Q.F.); (I.G.)
- Biomedical Research Centre, Qatar University, Doha 2713, Qatar
| | - Semir Vranic
- College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; (Q.F.); (I.G.)
- Correspondence: (S.V.); (A.-E.A.M.); Tel.:+974-4403-7873 (S.V.); +974-4403-7817 (A.-E.A.M.)
| | - Ala-Eddin Al Moustafa
- College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; (Q.F.); (I.G.)
- Biomedical Research Centre, Qatar University, Doha 2713, Qatar
- Correspondence: (S.V.); (A.-E.A.M.); Tel.:+974-4403-7873 (S.V.); +974-4403-7817 (A.-E.A.M.)
| |
Collapse
|
21
|
Pattyn J, Van Keer S, Tjalma W, Matheeussen V, Van Damme P, Vorsters A. Infection and vaccine-induced HPV-specific antibodies in cervicovaginal secretions. A review of the literature. PAPILLOMAVIRUS RESEARCH 2019; 8:100185. [PMID: 31494291 PMCID: PMC6804463 DOI: 10.1016/j.pvr.2019.100185] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/19/2019] [Accepted: 09/04/2019] [Indexed: 02/02/2023]
Abstract
Background Human papillomavirus (HPV) infects and propagates in the cervical mucosal epithelium. Hence, in addition to assessing systemic immunity, the accurate measurement of cervical immunity is important to evaluate local immune responses to HPV infection and vaccination. This review discusses studies that investigated the presence of infection and vaccine-induced HPV-specific antibodies in cervicovaginal secretions (CVS). Methods We searched the two main health sciences databases, PubMed and the ISI Web of Science, from the earliest dates available to March 2019. From the eligible publications, information was extracted regarding: (i) study design, (ii) the reported HPV-specific antibody concentrations in CVS (and the associated serum levels, when provided), (iii) the CVS collection method, and (iv) the immunoassays used. Results The systematic search and selection process yielded 44 articles. The evidence of HPV-specific antibodies in CVS after natural infection (26/44) and HPV vaccination (18/44) is discussed. Many studies indicate that HPV-specific antibody detection in CVS is variable but feasible with a variety of collection methods and immunoassays. Most CVS samples were collected by cervicovaginal washing or wicks, and antibody presence was mostly determined by VLP-based ELISAs. The moderate to strong correlation between vaccine-induced antibody levels in serum and in CVS indicates that HPV vaccines generate antibodies that transudate through the cervical mucosal epithelium. Conclusion Although HPV-specific antibodies have lower titres in CVS than in serum samples, studies have shown that their detection in CVS is feasible. Nevertheless, the high variability of published observations and the lack of a strictly uniform, well-validated method for the collection, isolation and quantification of antibodies indicates a need for specific methods to improve and standardize the detection of HPV-specific antibodies in CVS.
Collapse
Affiliation(s)
- Jade Pattyn
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Belgium.
| | - Severien Van Keer
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Belgium
| | - Wiebren Tjalma
- Multidisciplinary Breast Clinic, Gynaecological Oncology Unit, Department of Obstetrics and Gynaecology, Antwerp University Hospital (UZA) (Belgium), Molecular Imaging, Pathology, Radiotherapy, and Oncology (MIPRO), Faculty of Medicine and Health Sciences, University of Antwerp, Belgium
| | - Veerle Matheeussen
- Department of Microbiology, Antwerp University Hospital (UZA) (Belgium); Department of Medical Microbiology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp (Belgium); Department of Medical Biochemistry, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Belgium
| | - Pierre Van Damme
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Belgium
| | - Alex Vorsters
- Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Belgium
| |
Collapse
|
22
|
Zhu FC, Hu SY, Hong Y, Hu YM, Zhang X, Zhang YJ, Pan QJ, Zhang WH, Zhao FH, Zhang CF, Yang X, Yu JX, Zhu J, Zhu Y, Chen F, Zhang Q, Wang H, Wang C, Bi J, Xue S, Shen L, Zhang YS, He Y, Tang H, Karkada N, Suryakiran P, Bi D, Struyf F. Efficacy, immunogenicity and safety of the AS04-HPV-16/18 vaccine in Chinese women aged 18-25 years: End-of-study results from a phase II/III, randomised, controlled trial. Cancer Med 2019; 8:6195-6211. [PMID: 31305011 PMCID: PMC6797633 DOI: 10.1002/cam4.2399] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 06/18/2019] [Accepted: 06/20/2019] [Indexed: 12/20/2022] Open
Abstract
Background Cervical cancer is a major public health concern in China. We report the end‐of‐study results of a phase II/III trial to assess the efficacy, immunogenicity, and safety of the AS04‐human papillomavirus (HPV)‐16/18 vaccine in Chinese women aged 18‐25 years followed for up to 72 months after first vaccination. Results of approximately 57 months following first vaccination have been previously reported. Methods Healthy 18‐25‐year‐old women (N = 6051) were randomized (1:1) to receive three doses of AS04‐HPV‐16/18 vaccine or Al(OH)3 (control) at Months 0‐1‐6. Vaccine efficacy against HPV‐16/18 infection and cervical intraepithelial neoplasia (CIN), cross‐protective vaccine efficacy against infections and lesions associated with nonvaccine oncogenic HPV types, immunogenicity, and safety were assessed. Efficacy was assessed in the according‐to‐protocol efficacy (ATP‐E) cohort (vaccine N = 2888; control N = 2892), total vaccinated cohort for efficacy (TVC‐E; vaccine N = 2987; control N = 2985) and TVC‐naïve (vaccine N = 1660; control N = 1587). Results In initially HPV‐16/18 seronegative/DNA‐negative women, vaccine efficacy against HPV‐16/18‐associated CIN grade 2 or worse was 87.3% (95% CI: 5.5, 99.7) in the ATP‐E, 88.7% (95% CI: 18.5, 99.7) in the TVC‐E, and 100% (95% CI: 17.9, 100) in the TVC‐naïve. Cross‐protective efficacy against incident infection with HPV‐31, HPV‐33 and HPV‐45 was 59.6% (95% CI: 39.4, 73.5), 42.7% (95% CI: 15.6, 61.6), and 54.8% (95% CI: 19.3, 75.6), respectively (ATP‐E). At Month 72, >95% of initially seronegative women who received HPV vaccine in the ATP cohort for immunogenicity (N = 664) remained seropositive for anti‐HPV‐16/18 antibodies; anti‐HPV‐16 and anti‐HPV‐18 geometric mean titers were 678.1 EU/mL (95% CI: 552.9, 831.5) and 343.7 EU/mL (95% CI: 291.9, 404.8), respectively. Serious adverse events were infrequent (1.9% vaccine group [N = 3026]; 2.7% control group [N = 3025]). Three and zero women died in the control group and the vaccine group respectively. New onset autoimmune disease was reported in two women in the vaccine group and two in the control group. Conclusions This is the first large‐scale randomized clinical trial of HPV vaccination in China. High and sustained vaccine efficacy against HPV‐16/18‐associated infection and cervical lesions was demonstrated up to Month 72. The vaccine had an acceptable safety profile. Combined with screening, prophylactic HPV vaccination could potentially reduce the high burden of HPV infection and cervical cancer in China. Trial registration NCT00779766.
Collapse
Affiliation(s)
- Feng-Cai Zhu
- Jiangsu Province Center for Disease Prevention and Control, Nanjing, China
| | - Shang-Ying Hu
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Ying Hong
- Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yue-Mei Hu
- Jiangsu Province Center for Disease Prevention and Control, Nanjing, China
| | - Xun Zhang
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Yi-Ju Zhang
- Jiangsu Province Center for Disease Prevention and Control, Nanjing, China
| | - Qin-Jing Pan
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Wen-Hua Zhang
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Fang-Hui Zhao
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Cheng-Fu Zhang
- Lianshui Center for Disease Prevention and Control, Lianshui, China
| | - Xiaoping Yang
- Jintan Center for Disease Prevention and Control, Jintan, China
| | - Jia-Xi Yu
- Xuzhou Center for Disease Prevention and Control, Xuzhou, China
| | - Jiahong Zhu
- Lianshui Center for Disease Prevention and Control, Lianshui, China
| | - Yejiang Zhu
- Binhai Center for Disease Prevention and Control, Yancheng, China
| | - Feng Chen
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Qian Zhang
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Hong Wang
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Changrong Wang
- Jintan Center for Disease Prevention and Control, Jintan, China
| | - Jun Bi
- Xuzhou Center for Disease Prevention and Control, Xuzhou, China
| | - Shiyin Xue
- Lianshui Center for Disease Prevention and Control, Lianshui, China
| | - Lingling Shen
- Xuzhou Center for Disease Prevention and Control, Xuzhou, China
| | - Yan-Shu Zhang
- Binhai Center for Disease Prevention and Control, Yancheng, China
| | | | | | | | | | | | | |
Collapse
|
23
|
Immunogenicity noninferiority study of 2 doses and 3 doses of an Escherichia coli-produced HPV bivalent vaccine in girls vs. 3 doses in young women. SCIENCE CHINA-LIFE SCIENCES 2019; 63:582-591. [PMID: 31231780 PMCID: PMC7223315 DOI: 10.1007/s11427-019-9547-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 04/16/2019] [Indexed: 01/26/2023]
Abstract
A new HPV-16/18 bivalent vaccine expressed by the Escherichia coli has been proven to be efficacious in adult women. A randomized, immunogenicity noninferiority study of this candidate vaccine was conducted in December 2015 in China. Girls aged 9–14 years were randomized to receive 2 doses at months 0 and 6 (n=301) or 3 doses at months 0, 1 and 6 (n=304). Girls aged 15–17 years (n=149) and women aged 18–26 years (n=225) received 3 doses. The objectives included noninferiority analysis of the IgG geometric mean concentration (GMC) ratio (95% CI, lower bound>0.5) to HPV-16 and HPV-18 at month 7 in girls compared with women. In the per-protocol set, the GMC ratio of IgG was noninferior for girls aged 9–17 years receiving 3 doses compared with women (1.76 (95% CI, 1.56, 1.99) for HPV-16 and 1.93 (95% CI, 1.69, 2.21) for HPV-18) and noninferior for girls aged 9–14 years receiving 2 doses compared with women (1.45 (95% CI, 1.25, 1.62) for HPV-16 and 1.17 (95% CI, 1.02, 1.33) for HPV-18). Noninferiority was also demonstrated for neutralizing antibodies. The immunogenicity of the HPV vaccine in girls receiving 3 or 2 doses was noninferior compared with that in young adult women.
Collapse
|
24
|
Pattinson DJ, Apte SH, Wibowo N, Chuan YP, Rivera-Hernandez T, Groves PL, Lua LH, Middelberg APJ, Doolan DL. Chimeric Murine Polyomavirus Virus-Like Particles Induce Plasmodium Antigen-Specific CD8 + T Cell and Antibody Responses. Front Cell Infect Microbiol 2019; 9:215. [PMID: 31275867 PMCID: PMC6593135 DOI: 10.3389/fcimb.2019.00215] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 06/03/2019] [Indexed: 12/28/2022] Open
Abstract
An effective vaccine against the Plasmodium parasite is likely to require the induction of robust antibody and T cell responses. Chimeric virus-like particles are an effective vaccine platform for induction of antibody responses, but their capacity to induce robust cellular responses and cell-mediated protection against pathogen challenge has not been established. To evaluate this, we produced chimeric constructs using the murine polyomavirus structural protein with surface-exposed CD8+ or CD4+ T cell or B cell repeat epitopes derived from the Plasmodium yoelii circumsporozoite protein, and assessed immunogenicity and protective capacity in a murine model. Robust CD8+ T cell responses were induced by immunization with the chimeric CD8+ T cell epitope virus-like particles, however CD4+ T cell responses were very low. The B cell chimeric construct induced robust antibody responses but there was no apparent synergy when T cell and B cell constructs were administered as a pool. A heterologous prime/boost regimen using plasmid DNA priming followed by a VLP boost was more effective than homologous VLP immunization for cellular immunity and protection. These data show that chimeric murine polyomavirus virus-like particles are a good platform for induction of CD8+ T cell responses as well as antibody responses.
Collapse
MESH Headings
- Animals
- Antibodies, Protozoan
- Antibody Formation/immunology
- Antigens, Protozoan/immunology
- B-Lymphocytes
- CD4-Positive T-Lymphocytes
- CD8-Positive T-Lymphocytes/immunology
- Disease Models, Animal
- Epitopes, B-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/immunology
- Immunity, Cellular
- Immunization
- Immunization, Secondary
- Malaria Vaccines
- Mice
- Mice, Inbred BALB C
- Plasmodium yoelii
- Polyomavirus/genetics
- Polyomavirus/immunology
- Protozoan Proteins/immunology
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/immunology
Collapse
Affiliation(s)
- David J. Pattinson
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Simon H. Apte
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Nani Wibowo
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Yap P. Chuan
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Tania Rivera-Hernandez
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Penny L. Groves
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Linda H. Lua
- Protein Expression Facility, University of Queensland, Brisbane, QLD, Australia
| | - Anton P. J. Middelberg
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Denise L. Doolan
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
25
|
Lekoane KMB, Kuupiel D, Mashamba-Thompson TP, Ginindza TG. Evidence on the prevalence, incidence, mortality and trends of human papilloma virus-associated cancers in sub-Saharan Africa: systematic scoping review. BMC Cancer 2019; 19:563. [PMID: 31185951 PMCID: PMC6558783 DOI: 10.1186/s12885-019-5781-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 05/31/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Human papilloma virus (HPV) associated cervical cancer remains a global concern particular, in Sub-Saharan Africa (SSA) where the impact is felt most. Evidence show that many other cancers such as vaginal, anal, oropharyngeal, penile are because of persistent infection with HPV especially, high-risk types. AIM We mapped evidence on the incidence, prevalence, mortality, and the trends of human papillomavirus-related cancers in SSA. METHODS A comprehensive literature search was conducted from several databases including PubMed, Google scholar, Science Direct, and CINAHL and MEDLINE via EBSCOhost as well as World Health Organization website for grey literature. Studies reporting HPV-related cancers in SSA outcomes including prevalence, incidence, mortality, and trends were included in this study. The risk of bias of the included studies were assessed using the mixed methods appraisal tool version 2011. We employed PRISMA (preferred reporting items for systematic reviews and meta-analyses) to report the search results. Thematic analysis used to reveal the emerging themes from the included studies. RESULTS Seventy-four (74) studies were retrieved at full article screening, eight of them (six reviews, and two quantitative study) were eligible for data extraction. The degree of agreement between the two independent reviewers following full article screening, was 86.49% agreement versus 64.57% likely by chance which constituted moderate to significant agreement (Kappa statistic = 0.62, p-value< 0.05). Of the eight included studies, four (50%) studies generalized about SSA with no country of interest; two (25%) studies were conducted in Nigeria; one (12.5%) reported about Uganda, Zambia, Guinea, Malawi Tanzania, Mali, Mozambique, Zimbabwe; and one (12.5%) reported about Ethiopia, Senegal, Zimbabwe and Uganda. These eight included studies reported evidence on more than one outcome of interest. Four studies reported about the prevalence of HPV-related cancers, seven studies reported about the incidence, four studies reported about mortality, and four studies reported about the trends of HPV-related cancers. CONCLUSION This study observation highlighted a gap of knowledge regarding the epidemiological data on the recent HPV prevalence in SSA, which will have a potential impact in determining the distribution of HPV on different body sites (cervix, penis, vagina, vulva, anus and oropharynx). Ongoing research projects are recommended in SSA to enhance the value of HPV, and HPV-associated cancers epidemiological data to inform strategies or/and policies on prevention, diagnosis, and treatment of HPV-related conditions.
Collapse
Affiliation(s)
- Kabelo M. B. Lekoane
- Discipline of Public Health Medicine, School of Nursing and Public Health, University of KwaZulu-Natal, 2nd Floor George Campbell Building, Howard College Campus, Durban, 4001 South Africa
| | - Desmond Kuupiel
- Discipline of Public Health Medicine, School of Nursing and Public Health, University of KwaZulu-Natal, 2nd Floor George Campbell Building, Howard College Campus, Durban, 4001 South Africa
- Research for Sustainable Development, Sunyani, Ghana
| | - Tivani P. Mashamba-Thompson
- Discipline of Public Health Medicine, School of Nursing and Public Health, University of KwaZulu-Natal, 2nd Floor George Campbell Building, Howard College Campus, Durban, 4001 South Africa
| | - Themba G. Ginindza
- Discipline of Public Health Medicine, School of Nursing and Public Health, University of KwaZulu-Natal, 2nd Floor George Campbell Building, Howard College Campus, Durban, 4001 South Africa
| |
Collapse
|
26
|
Lieblong BJ, Montgomery BEE, Su LJ, Nakagawa M. Natural history of human papillomavirus and vaccinations in men: A literature review. Health Sci Rep 2019; 2:e118. [PMID: 31139757 PMCID: PMC6529831 DOI: 10.1002/hsr2.118] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 12/07/2018] [Accepted: 02/09/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND AIMS Infection with high-risk (HR) genotypes of the human papillomavirus (HPV) is necessary for and causative of almost all cervical cancers and their precursor condition, cervical intraepithelial neoplasia. These conditions have been sharply reduced by cervical cytology screening, and a further decrease is expected because of the recent introduction of prophylactic HPV vaccinations. While significant attention has been given to gynecologic HPV disease, men can be affected by HPV-related cancers of the anus, penis, and oropharynx. This literature review aims to address disparities in HPV-related disease in men, and certain HR male subpopulations, compared with women. DISCUSSION Overall, immunocompetent men are far less likely than women to develop anogenital HPV-related cancers, despite harboring HR HPV infections at anogenital sites. On the other hand, men who have sex with men and men living with human immunodeficiency virus infection are at considerably higher risk of HPV-related disease. Historic rates of prophylactic HPV vaccination in males have trailed those of females due to numerous multilevel factors, although, in recent years, this sex gap in vaccination coverage has been closing. In the absence of routine HPV screening in males, therapeutic vaccinations have emerged as a potential treatment modality for preinvasive neoplasia and are in various phases of clinical testing. CONCLUSION Successful reductions in HPV disease morbidity at the population level must acknowledge and target HPV infections in men.
Collapse
Affiliation(s)
- Benjamin J. Lieblong
- College of Medicine, Department of PathologyUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Brooke E. E. Montgomery
- Faye W. Boozman College of Public Health, Department of Health Behavior and Health EducationUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - L. Joseph Su
- Faye W. Boozman College of Public Health, Department of EpidemiologyUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Mayumi Nakagawa
- College of Medicine, Department of PathologyUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| |
Collapse
|
27
|
Immunization of mice by the co-administration of codon-optimized HPV16 E7 and lL12 genes against HPV16-associated cervical cancer. Microb Pathog 2019; 132:20-25. [PMID: 31004722 DOI: 10.1016/j.micpath.2019.04.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 04/11/2019] [Accepted: 04/16/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND Various promising procedures have been used to improve the potency of DNA vaccines for the treatment of human papillomavirus type 16 (HPV16) infections. Interleukin-12 (IL12) is a powerful adjuvant that can contribute to T cell-mediated protection against many pathogens, specifically viruses. Considering the important role of T cell-mediated immunity in tumor clearance, the induction of these responses can help control the progression of tumors in animal models. We have demonstrated that the co-administration of codon-optimized E7 (uE7) gene of HPV16 with interleukin-12 is effective in the development of antitumor responses. OBJECTIVES The present study examined the co-administration of codon-optimized HPV16 E7 gene with murine interleukin-12 gene (mIL-12) as a vaccine adjuvant in tumor mice model. MATERIALS AND METHODS C57BL/6 mice were studied for tumor progression after injection of recombinant DNA vaccines. Lactate dehydrogenase (LDH) and IFN-γ were measured to evaluate the activity of cytotoxic T lymphocytes (CTLs). Measurements of tumor volume and MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay were used for assessment of therapeutic antitumor effects of the vaccines. RESULTS Results showed that DNA vaccines, specifically codon-optimized E7/murine interleukin-12 (mIL-12), elicited significant differences in levels of IFN-γ and cytotoxic T lymphocyte (CTLs) responses compared to control groups. Furthermore, higher antitumor response and lower tumor size in the vaccine group was significantly evident compared to control group. CONCLUSION The co-administration of codon-optimized HPV16 E7 gene with IL12 significantly enhances the DNA vaccine potency against HPV16-associated cervical cancer.
Collapse
|
28
|
Non-Vaccine-Type Human Papillomavirus Prevalence After Vaccine Introduction: No Evidence for Type Replacement but Evidence for Cross-Protection. Sex Transm Dis 2019; 45:260-265. [PMID: 29465705 DOI: 10.1097/olq.0000000000000731] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND We examined non-vaccine-type human papillomavirus (HPV) prevalence in a community before and during the first 8 years after vaccine introduction, to assess for (1) type replacement with any non-vaccine-type HPV and (2) cross-protection with non-vaccine types genetically related to vaccine-type HPV. METHODS Sexually experienced 13- to- 26-year-old women were recruited for 3 cross-sectional studies from 2006 to 2014 (N = 1180). Outcome variables were as follows: (1) prevalence of at least 1 of 32 anogenital non-vaccine-type HPVs and (2) prevalence of at least 1 HPV type genetically related to HPV-16 and HPV-18. We determined changes in proportions of non-vaccine-type HPV prevalence across the study waves using logistic regression with propensity score inverse probability weighting. RESULTS Vaccine initiation rates increased from 0% to 71.3%. Logistic regression demonstrated that from 2006 to 2014, there was no increase in non-vaccine-type HPV among vaccinated women (adjusted odds ratio [AOR], 1.02; 95% confidence interval [CI], 0.73-1.42), but an increase among unvaccinated women (AOR, 1.88; 95% CI, 1.16-3.04). Conversely, there was a decrease in types genetically related to HPV-16 among vaccinated (AOR, 0.57; 95% CI, 0.38-0.88) but not unvaccinated women (AOR, 1.33; 95% CI, 0.81-2.17). CONCLUSIONS We did not find evidence of type replacement, but did find evidence of cross-protection against types genetically related to HPV-16. These findings have implications for cost-effectiveness analyses, which may impact vaccine-related policies, and provide information to assess the differential risk for cervical cancer in unvaccinated and vaccinated women, which may influence clinical screening recommendations. The findings also have implications for public health programs, such as health messaging for adolescents, parents, and clinicians about HPV vaccination.
Collapse
|
29
|
Covert C, Ding L, Brown D, Franco EL, Bernstein DI, Kahn JA. Evidence for cross-protection but not type-replacement over the 11 years after human papillomavirus vaccine introduction. Hum Vaccin Immunother 2019; 15:1962-1969. [PMID: 30633598 DOI: 10.1080/21645515.2018.1564438] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Examination of cross-protection and type replacement after human papillomavirus (HPV) vaccine introduction is essential to guide vaccination recommendations and policies. The aims of this study were to examine trends in non-vaccine-type HPV: 1) genetically related to vaccine types (to assess for cross-protection) and 2) genetically unrelated to vaccine types (to assess for type replacement), among young women 13-26 years of age during the 11 years after HPV vaccine introduction. Participants were recruited from a hospital-based teen health center and a community health department for four cross-sectional surveillance studies between 2006 and 2017. Participants completed a survey that assessed sociodemographic characteristics and behaviors, and cervicovaginal swabs were collected and tested for 36 HPV genotypes. We determined changes in proportions of non-vaccine-type HPV prevalence and conducted logistic regression to determine the odds of infection across the surveillance studies, propensity-score adjusted to control for selection bias. Analyses were stratified by vaccination status. Among vaccinated women who received only the 4-valent vaccine (n = 1,540), the adjusted prevalence of HPV types genetically related to HPV16 decreased significantly by 45.8% (adjusted odds ratio [AOR] = 0.48, 95% confidence interval [CI] = 0.31-0.74) from 2006-2017, demonstrating evidence of cross-protection. The adjusted prevalence of HPV types genetically related to HPV18 did not change significantly (14.2% decrease, AOR = 0.83, 95% CI = 0.56-1.21). The adjusted prevalence of HPV types genetically unrelated to vaccine types did not change significantly (4.2% increase, AOR = 1.09, CI = 0.80-1.48), demonstrating no evidence of type replacement. Further studies are needed to monitor for cross-protection and possible type replacement after introduction of the 9-valent HPV vaccine.
Collapse
Affiliation(s)
- Courtney Covert
- a Cincinnati Children's Hospital Medical Center , Cincinnati , OH , USA
| | - Lili Ding
- a Cincinnati Children's Hospital Medical Center , Cincinnati , OH , USA.,b Department of Pediatrics, University of Cincinnati College of Medicine , Cincinnati , OH , USA
| | - Darron Brown
- c Department of Medicine, Indiana University School of Medicine , Indianapolis , IN , USA
| | - Eduardo L Franco
- d Department of Oncology, McGill University , Montreal , QC , Canada, USA
| | - David I Bernstein
- a Cincinnati Children's Hospital Medical Center , Cincinnati , OH , USA.,b Department of Pediatrics, University of Cincinnati College of Medicine , Cincinnati , OH , USA
| | - Jessica A Kahn
- a Cincinnati Children's Hospital Medical Center , Cincinnati , OH , USA.,b Department of Pediatrics, University of Cincinnati College of Medicine , Cincinnati , OH , USA
| |
Collapse
|
30
|
Sherman SM, Nailer E. Attitudes towards and knowledge about Human Papillomavirus (HPV) and the HPV vaccination in parents of teenage boys in the UK. PLoS One 2018; 13:e0195801. [PMID: 29641563 PMCID: PMC5895045 DOI: 10.1371/journal.pone.0195801] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 03/29/2018] [Indexed: 01/27/2023] Open
Abstract
The incidence of cancers attributable to Human Papillomavirus (HPV) that affect males is on the rise. Currently in the UK teenage boys are not vaccinated against HPV while teenage girls are. The rationale for not vaccinating boys is that vaccinating girls should provide herd immunity to boys, however this does not protect men who have sex with men or men who have sex with unvaccinated women. The issue of whether to vaccinate boys or not is a controversial one with considerable lobbying taking place to change the existing policy. On one side of the debate are financial considerations while on the other side health equality is important. One avenue that has not been presented is the parental perspective. The current study uses a self-report questionnaire to explore what parents of teenage boys know about HPV and the vaccine and whether they want the vaccine for their sons. Only half of the parents had heard of HPV prior to completing the survey. Of those who had heard of HPV, knowledge about the health sequelae of HPV for men was poor relative to their knowledge about its impact on female health. Parents who would be willing to vaccinate their sons had higher levels of knowledge about HPV than those parents who would be unwilling or unsure. Irrespective of whether they had previously heard of HPV or not, once provided with a brief description of HPV, the majority of parents thought that boys should be offered the vaccination. There is a pressing need for public education about the potential impact of HPV on male health in order to facilitate uptake of the vaccine in the event of the vaccination programme being extended to men or to facilitate informed decision making about seeking the vaccine privately in the event that it isn't.
Collapse
Affiliation(s)
| | - Emma Nailer
- School of Psychology, Keele University, Keele, Staffs, United Kingdom
| |
Collapse
|
31
|
Stanley M. Tumour virus vaccines: hepatitis B virus and human papillomavirus. Philos Trans R Soc Lond B Biol Sci 2018; 372:rstb.2016.0268. [PMID: 28893935 DOI: 10.1098/rstb.2016.0268] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2017] [Indexed: 12/11/2022] Open
Abstract
Two of the most important human oncogenic viruses are hepatitis B virus (HBV) and human papillomavirus (HPV). HBV infection has been preventable by vaccination since 1982; vaccination of neonates and infants is highly effective, resulting already in decreased rates of new infections, chronic liver disease and hepato-cellular carcinoma. Nonetheless, HBV remains a global public health problem with high rates of vertical transmission from mother to child in some regions. Prophylactic HPV vaccines composed of virus-like particles (VLPs) of the L1 capsid protein have been licensed since 2006/2007. These target infection by the oncogenic HPVs 16 and 18 (the cause of 70% of cervical cancers); a new vaccine licensed in 2014/2015 additionally targets HPVs 31, 33, 45, 52, 58. HPV vaccines are now included in the national immunization programmes in many countries, with young adolescent peri-pubertal girls the usual cohort for immunization. Population effectiveness in women is now being demonstrated in countries with high vaccine coverage with significant reductions in high-grade cervical intra-epithelial neoplasia (a surrogate for cervical cancer), genital warts and vaccine HPV type genoprevalence. Herd effects in young heterosexual men and older women are evident. Cancers caused by HBV and HPV should, in theory, be amenable to immunotherapies and various therapeutic vaccines for HPV in particular are in development and/or in clinical trial.This article is part of the themed issue 'Human oncogenic viruses'.
Collapse
Affiliation(s)
- Margaret Stanley
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| |
Collapse
|
32
|
Quadrivalent human papillomavirus vaccine and autoimmune adverse events: a case-control assessment of the vaccine adverse event reporting system (VAERS) database. Immunol Res 2018; 65:46-54. [PMID: 27406735 PMCID: PMC5406441 DOI: 10.1007/s12026-016-8815-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Gardasil is a quadrivalent human papillomavirus (HPV4) vaccine that was approved for use by the US Food and Drug Administration in June 2006. HPV4 vaccine is routinely recommended for administration to women in the USA who are 11-12 years old by the Advisory Committee on Immunization Practices. Previous studies suggest HPV4 vaccine administration was associated with autoimmune diseases. As a consequence, an epidemiological assessment of the vaccine adverse event reporting system database was undertaken for adverse event reports associated with vaccines administered from 2006 to 2014 to 6-39 year-old recipients with a listed US residence and a specified female gender. Cases with the serious autoimmune adverse event (SAAE) outcomes of gastroenteritis (odds ratio (OR) 4.627, 95 % confidence interval (CI) 1.892-12.389), rheumatoid arthritis (OR 5.629, 95 % CI 2.809-12.039), thrombocytopenia (OR 2.178, 95 % CI 1.222-3.885), systemic lupus erythematosus (OR 7.626, 95 % CI 3.385-19.366), vasculitis (OR 3.420, 95 % CI 1.211-10.408), alopecia (OR 8.894, 95 % CI 6.255-12.914), CNS demyelinating conditions (OR 1.585, 95 % CI 1.129-2.213), ovarian damage (OR 14.961, 95 % CI 6.728-39.199), or irritable bowel syndrome (OR 10.021, 95 % CI 3.725-33.749) were significantly more likely than controls to have received HPV4 vaccine (median onset of initial symptoms ranged from 3 to 37 days post-HPV4 vaccination). Cases with the outcome of Guillain-Barre syndrome (OR 0.839, 95 % CI 0.601-1.145) were no more likely than controls to have received HPV4 vaccine. In addition, cases with the known HPV4-related outcome of syncope were significantly more likely than controls to have received HPV4 vaccine (OR 5.342, 95 % CI 4.942-5.777). Cases with the general health outcomes of infection (OR 0.765, 95 % CI 0.428-1.312), conjunctivitis (OR 1.010, 95 % CI 0.480-2.016), diarrhea (OR 0.927, 95 % CI 0.809-1.059), or pneumonia (OR 0.785, 95 % CI 0.481-1.246) were no more likely than controls to have received HPV4 vaccine. Confirmatory epidemiological studies in other databases should be undertaken and long-term clinical consequences of HPV-linked SAAEs should be examined.
Collapse
|
33
|
Prasetyo RE, Mastutik G, Mustokoweni S. PREVALENCE OF HUMAN PAPILLOMAVIRUS GENOTYPES IN LOW AND HIGH GRADE SQUAMOUS INTRAEPITHELIAL LESIONS AT CERVICAL TISSUE. FOLIA MEDICA INDONESIANA 2017. [DOI: 10.20473/fmi.v53i4.7157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
HPV infection is known to cause cervical cancer. This study aimed to identify the variant of HPV genotypes of cervical precancerous lesions from low grade squamous intraepithelial lesion (LSIL) and high grade squamous intraepithelial lesion (HSIL). This was an explorative study using formalin fix paraffin embedded (FFPE) from cervical precancerous lesions at Dr. Soetomo Hospital, Surabaya. DNA was extracted from FFPE and hybridized for HPV genotyping using Ampliquality HPV Type Express kit (AB ANALITICA) by reverse line blot techniques. The results showed that there were variants of HPV genotype in LSIL. The variants were HPV16 (8/15), HPV18 (3/15), HPV52 (1/15), HPV6+31 (1/15), HPV6+18 (1/15), and HPV72+68 (1/15), and in HSIL which were HPV16 (4/10), HPV18 (2/10), HPV59 (1/10), HPV6+45 (1/10), HPV61+26 (1/10), and HPV16+31 (1/10). The characteristics of infection in LSIL were single infection of high-risk (hr) HPV and multiple infection of low-risk (lr)+hr HPV, and in HSIL were single infection of HPVhr, multiple infection of HPVhr+hr and HPVlr+hr. In conclusion, HPV prevalence in cervical precancerous lesions is single infection by HPV16 (48%), HPV18 (20%), HPV52 (4%), HPV59 (4%), and multiple infection by HPV6+31, HPV6+18, HPV6+45, HPV16+31, HPV61+26, HPV72+68 is 4%.
Collapse
|
34
|
Nilsson L, Brockow K, Alm J, Cardona V, Caubet JC, Gomes E, Jenmalm MC, Lau S, Netterlid E, Schwarze J, Sheikh A, Storsaeter J, Skevaki C, Terreehorst I, Zanoni G. Vaccination and allergy: EAACI position paper, practical aspects. Pediatr Allergy Immunol 2017; 28:628-640. [PMID: 28779496 DOI: 10.1111/pai.12762] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/26/2017] [Indexed: 01/15/2023]
Abstract
Immunization is highly effective in preventing infectious diseases and therefore an indispensable public health measure. Allergic patients deserve access to the same publicly recommended immunizations as non-allergic patients unless risks associated with vaccination outweigh the gains. Whereas the number of reported possible allergic reactions to vaccines is high, confirmed vaccine-triggered allergic reactions are rare. Anaphylaxis following vaccination is rare, affecting <1/100 000, but can occur in any patient. Some patient groups, notably those with a previous allergic reaction to a vaccine or its components, are at heightened risk of allergic reaction and require special precautions. Allergic reactions, however, may occur in patients without known risk factors and cannot be predicted by currently available tools. Unwarranted fear and uncertainty can result in incomplete vaccination coverage for children and adults with or without allergy. In addition to concerns about an allergic reaction to the vaccine itself, there is fear that routine childhood immunization may promote the development of allergic sensitization and disease. Thus, although there is no evidence that routine childhood immunization increases the risk of allergy development, such risks need to be discussed.
Collapse
Affiliation(s)
| | - Knut Brockow
- Department of Dermatology and Allergy Biederstein, Technical University Munich, Munich, Germany
| | - Johan Alm
- Sachs' Children and Youth Hospital and Karolinska Institutet, Södersjukhuset, Stockholm, Sweden
| | - Victoria Cardona
- Allergy Section, Department of Internal Medicine, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | | | | | - Maria C Jenmalm
- Unit of Autoimmunity and Immune Regulation, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Susanne Lau
- Pediatric Pneumology and Immunology, Charité Universitätsmedizin, Berlin, Germany
| | - Eva Netterlid
- Department of Occupational and Environmental Dermatology, Lund University, Malmö, Sweden.,The Public Health Agency of Sweden, Stockholm, Sweden
| | - Jürgen Schwarze
- Child Life & Health and MRC-Centre for Inflammation Research, The University of Edinburgh, Edinburgh, UK
| | - Aziz Sheikh
- Asthma UK Centre for Applied Research, Usher Institute of Population Health Sciences and Informatics, The University of Edinburgh, Edinburgh, UK
| | | | - Chrysanthi Skevaki
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University Marburg, University Hospital Giessen and Marburg GmbH, Marburg, Germany
| | | | | |
Collapse
|
35
|
Barroeta JE, Adhikari-Guragain D, Grotkowski CE. Cervical cancer screening in the era of HPV vaccination: A review of shifting paradigms in cytopathology. Diagn Cytopathol 2017; 45:903-914. [PMID: 28589649 DOI: 10.1002/dc.23737] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 03/16/2017] [Accepted: 04/07/2017] [Indexed: 01/14/2023]
Abstract
Significant changes in cervical cancer screening practice, guidelines, and prevention of cervical cancer have taken place in recent years including the raising of initial cervical cancer screening age, changes in frequency of cytology screening, and the adoption of high risk HPV and cytology co-testing for some patients; the introduction of the bivalent, quadrivalent, and 9-valent HPV vaccines; and the recent approval of high risk HPV testing as primary screening with the use of cytology as triage in positive cases. This review discusses the significance of primary HPV screening, the impact of HPV vaccination in the prevalence of cervical cancer and its precursors, the interplay between high risk HPV testing and vaccination, and the implications for clinical and cytological management. Future strategies for cervical screening in the post-vaccination era are also discussed.
Collapse
Affiliation(s)
- Julieta E Barroeta
- Department of Pathology, Cooper University Hospital, M.D. Anderson Cancer Center at Cooper, Camden, New Jersey
| | - Deepti Adhikari-Guragain
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Carolyn E Grotkowski
- Department of Pathology, Cooper University Hospital, M.D. Anderson Cancer Center at Cooper, Camden, New Jersey
| |
Collapse
|
36
|
Praditpornsilpa K, Kingwatanakul P, Deekajorndej T, Rianthavorn P, Susantitaphong P, Katavetin P, Tiranathanakul K, Srisawat N, Tungsanga K, Eiam-Ong S, Townamchai N. Immunogenicity and safety of quadrivalent human papillomavirus types 6/11/16/18 recombinant vaccine in chronic kidney disease stage IV, V and VD. Nephrol Dial Transplant 2017; 32:132-136. [PMID: 26932687 DOI: 10.1093/ndt/gfv444] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 12/06/2015] [Indexed: 11/12/2022] Open
Abstract
Background Up to >80% of sexually active adults will become infected with human papillomavirus (HPV) during their lifetime. Persistent HPV infection can result in cervical, vulvovaginal, penile and anogenital cancer. Clinical studies have shown the efficacy of three doses of quadrivalent HPV-6/11/16/18 L1 virus-like particle (VLP) vaccination, at Day 0, Month 2 and Month 6, to lower the occurrence of HPV infection and its complications. However, immunogenicity and safety of the HPV vaccine have not been proven in the chronic kidney disease (CKD) population. Methods Sixty CKD stage IV, V and VD patients were enrolled for quadrivalent HPV-6/11/16/18 vaccination. A dose of vaccine was given at Day 0, Month 2 and Month 6. Each dose contained 20 μg HPV-6 L1 VLP, 40 μg HPV-11 L1 VLP, 40 μg HPV-16 L1 VLP and 20 μg HPV-18 L1 VLP, along with 225 μg of amorphous aluminum hydroxyphosphate sulfate adjuvant. HPV type-specific antibody response to neutralizing epitopes on HPV-6/11/16/18 was performed by multiplexed, competitive Luminex® immunoassays (cLIA) at Day 0 and Month 7. Results At Day 0, anti-HPV seropositivity was 0-6.6% depending on HPV genotype. Patients who received three doses of vaccine had 98.2, 100, 100 and 98.2% seropositivity for genotypes 6/11/16/18, respectively. The average cLIA at Month 7 for genotypes 6/11/16/18 were 928.4 ± 231.1, 1136.1 ± 264.6, 6951.0 ± 1872.3 and 2196.3 ± 761.2 milliMerck units (mMu)/mL, respectively. No serious vaccine-related adverse events were observed. Conclusions Quadrivalent HPV vaccine has been well tolerated, is safe and provides excellent immunogenicity in late-stage CKD.
Collapse
Affiliation(s)
- Kearkiat Praditpornsilpa
- Division of Nephrology, Department of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Pornchai Kingwatanakul
- Department of Pediatrics, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Tawatchai Deekajorndej
- Department of Pediatrics, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Pornpimol Rianthavorn
- Department of Pediatrics, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Paweena Susantitaphong
- Division of Nephrology, Department of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Pisut Katavetin
- Division of Nephrology, Department of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Khajohn Tiranathanakul
- Division of Nephrology, Department of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Nattachai Srisawat
- Division of Nephrology, Department of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Kriang Tungsanga
- Division of Nephrology, Department of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Somchai Eiam-Ong
- Division of Nephrology, Department of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Natavudh Townamchai
- Division of Nephrology, Department of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| |
Collapse
|
37
|
Zhu FC, Hu SY, Hong Y, Hu YM, Zhang X, Zhang YJ, Pan QJ, Zhang WH, Zhao FH, Zhang CF, Yang X, Yu JX, Zhu J, Zhu Y, Chen F, Zhang Q, Wang H, Wang C, Bi J, Xue S, Shen L, Zhang YS, He Y, Tang H, Karkada N, Suryakiran P, Bi D, Struyf F. Efficacy, immunogenicity, and safety of the HPV-16/18 AS04-adjuvanted vaccine in Chinese women aged 18-25 years: event-triggered analysis of a randomized controlled trial. Cancer Med 2016; 6:12-25. [PMID: 27998015 PMCID: PMC5269697 DOI: 10.1002/cam4.869] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/15/2016] [Accepted: 07/21/2016] [Indexed: 12/14/2022] Open
Abstract
We previously reported the results of a phase II/III, double‐blind, randomized controlled study in Chinese women (NCT00779766) showing a 94.2% (95% confidence interval: 62.7–99.9) HPV‐16/18 AS04‐adjuvanted vaccine efficacy (VE) against cervical intraepithelial neoplasia grade 1 or higher (CIN1+) and/or 6‐month (M) persistent infection (PI) with a mean follow‐up of <2 years, and immunogenicity until 7 months post‐dose 1. Here, we report efficacy and safety results from an event‐triggered analysis with ~3 years longer follow‐up, and immunogenicity until M24. Healthy 18–25‐year‐old women (N = 6051) were randomized (1:1) to receive three doses of HPV‐16/18 vaccine or Al(OH)3 (control) at M0, 1, 6. VE against HPV‐16/18‐associated CIN2+, and cross‐protective VE against infections with nonvaccine oncogenic HPV types, immunogenicity, and safety were assessed. In the according‐to‐protocol efficacy cohort, in initially seronegative/DNA‐negative women (vaccine group: N = 2524; control group: N = 2535), VE against HPV‐16/18‐associated CIN2+ was 87.3% (5.3–99.7); VE against incident infection or against 6‐month persistent infection associated with HPV‐31/33/45 was 50.1% (34.3–62.3) or 52.6% (24.5–70.9), respectively. At least, 99.6% of HPV‐16/18‐vaccines remained seropositive for anti‐HPV‐16/18 antibodies; anti‐HPV‐16 and ‐18 geometric mean titers were 1271.1 EU/mL (1135.8–1422.6) and 710.0 EU/ml (628.6–801.9), respectively. Serious adverse events were infrequent (1.7% vaccine group [N = 3026]; 2.5% control group [N = 3026]). Of the 1595 reported pregnancies, nine had congenital anomalies (five live infants, three elective terminations, one stillbirth) that were unlikely vaccination‐related (blinded data). VE against HPV‐16/18‐associated CIN2+ was demonstrated and evidence of cross‐protective VE against oncogenic HPV types was shown. The vaccine was immunogenic and had an acceptable safety profile.
Collapse
Affiliation(s)
- Feng-Cai Zhu
- Jiangsu Province Center for Disease Prevention and Control, Nanjing, China
| | - Shang-Ying Hu
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Ying Hong
- Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yue-Mei Hu
- Jiangsu Province Center for Disease Prevention and Control, Nanjing, China
| | - Xun Zhang
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Yi-Ju Zhang
- Jiangsu Province Center for Disease Prevention and Control, Nanjing, China
| | - Qin-Jing Pan
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Wen-Hua Zhang
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Fang-Hui Zhao
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Cheng-Fu Zhang
- Lianshui Center for Disease Prevention and Control, Lianshui, China
| | - Xiaoping Yang
- Jintan Center for Disease Prevention and Control, Jintan, China
| | - Jia-Xi Yu
- Xuzhou Center for Disease Prevention and Control, Xuzhou, China
| | - Jiahong Zhu
- Lianshui Center for Disease Prevention and Control, Lianshui, China
| | - Yejiang Zhu
- Binhai Center for Disease Prevention and Control, Yancheng, China
| | - Feng Chen
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Qian Zhang
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Hong Wang
- National Cancer Center - Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Changrong Wang
- Jintan Center for Disease Prevention and Control, Jintan, China
| | - Jun Bi
- Xuzhou Center for Disease Prevention and Control, Xuzhou, China
| | - Shiyin Xue
- Lianshui Center for Disease Prevention and Control, Lianshui, China
| | - Lingling Shen
- Xuzhou Center for Disease Prevention and Control, Xuzhou, China
| | - Yan-Shu Zhang
- Binhai Center for Disease Prevention and Control, Yancheng, China
| | | | | | | | | | - Dan Bi
- GSK Vaccines, Wavre, Belgium
| | | |
Collapse
|
38
|
Zhang Q, Liu YJ, Hu SY, Zhao FH. Estimating long-term clinical effectiveness and cost-effectiveness of HPV 16/18 vaccine in China. BMC Cancer 2016; 16:848. [PMID: 27814703 PMCID: PMC5097411 DOI: 10.1186/s12885-016-2893-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 10/26/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Human papillomavirus (HPV) 16 and 18 are the two most common HPV oncogenic types that can be prevented by vaccination. This study aimed at assessing the cost-effectiveness of 3 doses of the bivalent HPV vaccine in rural and urban settings in China. METHODS A Markov model was adapted to reflect the lifetime of a modelled 100,000 12-year-old girls cohort in rural and urban settings in China. Input parameters were obtained from published literature, official reports and a two-round expert review panel. Clinical and economic outcomes of vaccination at age 12 with screening was compared to screening only. In the base case analysis, a 3 % discount rate, the vaccine cost of 247 CNY (US$ 39, PAHO vaccine cost in 2013), two rounds of screening in a life time and 70 % coverage for both screening and vaccination were used. One-way, two-way and probabilistic sensitivity analyses were performed. We used different thresholds of cost-effectiveness to reflect the diversity of economic development in China. RESULTS Vaccination in addition to screening could prevent 60 % more cervical cancer cases and deaths than screening only. The incremental cost effectiveness ratio varied largely when changing cost of vaccination and discount in one way analysis. Vaccination was very cost-effective when the vaccine cost ranged 87-630 CNY (US$ 13.8-100) in rural and 87-750 CNY (US$ 13.8-119) in urban; and remained cost-effective when the vaccine cost ranged 630-1,700 CNY (US$ 100-270) in rural and 750-1,900 CNY (US$ 119-302) in urban in two way analysis. Probabilistic sensitivity analyses showed that model results were robust. CONCLUSIONS In both rural and urban, the vaccination cost and discounting are important factors determining the cost-effectiveness of HPV vaccination; policy makers in China should take these into account when making a decision on the introduction of HPV vaccine. In areas with a high burden of cervical cancer and limited screening activities, HPV vaccination should be prioritized. However, the vaccine cost needs to be reduced in order to make it very cost-effective and affordable as well, in particular in poverty areas with high disease burden.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Cancer Epidemiology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yi-Jun Liu
- Department of Cancer Epidemiology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of Preventive Medicine, School of Public Health, Zunyi Medical College, Zunyi, 563099, China
| | - Shang-Ying Hu
- Department of Cancer Epidemiology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Fang-Hui Zhao
- Department of Cancer Epidemiology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
39
|
The Effect of Cryotherapy on Human Papillomavirus Clearance Among HIV-Positive Women in Lusaka, Zambia. J Low Genit Tract Dis 2016; 19:301-6. [PMID: 26125097 DOI: 10.1097/lgt.0000000000000131] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE We sought to investigate the progression of human papillomaviruses (HPV) infection in HIV-positive women after cryotherapy. METHODS We examined changes in detection of high-risk HPV (hrHPV) cervical infections among HIV-infected women over a 12-week period after cryotherapy using stored specimens from a cohort study conducted between June 2009 and March 2011 in Lusaka, Zambia. Samples from visits at baseline and weeks 4, 8, and 12 were tested using the Roche Linear Array assay. RESULTS A total of 89 women were included in the analysis. The median age was 32 years (interquartile range [IQR], 28-36 years). The median CD4+ cell count was 350 cells/μL (IQR, 214-470 cells/μL), and 66% of women were receiving antiretroviral therapy. At baseline, the prevalence of hrHPV was 91% (95% confidence interval [CI], 83%-95%). HPV45 was the most common HPV type, present in (30%) women, followed by HPV16 (27%), HPV18 (27%), HPV51 (20%), and HPV58 (22%). Among women with valid results both at baseline and 12 weeks, 25% (17/67) cleared their initial hrHPV infection within 12 weeks of treatment, although 65% (11/17) had new hrHPV types detected. CONCLUSIONS Cryotherapy led to clearance of 25% of hrHPV infections within 12 weeks of treatment. However, hrHPV infection remained persistent in most women, and new hrHPV types were detected often, explaining the high rate of persistence and recurrence of cervical disease in this population. Continued efforts to scale up HPV vaccination and cervical screening should remain a priority in high HIV burden settings such as Zambia.
Collapse
|
40
|
Guo F, Hirth JM, Berenson AB. Comparison of HPV prevalence between HPV-vaccinated and non-vaccinated young adult women (20-26 years). Hum Vaccin Immunother 2016; 11:2337-44. [PMID: 26376014 DOI: 10.1080/21645515.2015.1066948] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
There is some concern about the effectiveness of the HPV vaccine among young adult women due to the risk of prior HPV infection. This study used National Health and Nutrition Examination Survey (NHANES) 2007-2012 data to evaluate the effectiveness of HPV vaccination among women 20-26 years of age who were vaccinated after 12 years of age. This cross-sectional study examined 878 young adult women (20-26 years) with complete information on HPV prevalence and HPV vaccination status from NHANES 2007-2012. Vaginal swab specimens were analyzed for HPV DNA by L1 consensus polymerase chain reaction followed by type-specific hybridization. Multivariate logistic regression models controlling for sociodemographic characteristics and sexual behaviors were used to compare type-specific HPV prevalence between vaccinated and unvaccinated women. A total of 21.4% of young adult women surveyed through NHANES between 2007 and 2012 received the HPV vaccine. Vaccinated women had a lower prevalence of vaccine types than unvaccinated women (7.4% vs 17.1%, prevalence ratio 0.43, 95% CI 0.21-0.88). The prevalence of high-risk nonvaccine types was higher among vaccinated women than unvaccinated women (52.1% vs 40.4%, prevalence ratio 1.29, 95% CI 1.06-1.57), but this difference was attenuated after adjusting for sexual behavior variables (adjusted prevalence ratio 1.19, 95% CI 0.99-1.43). HPV vaccination was effective against all 4 vaccine types in young women vaccinated after age 12. However, vaccinated women had a higher prevalence of high-risk nonvaccine types, suggesting that they may benefit from newer vaccines covering additional types.
Collapse
Affiliation(s)
- Fangjian Guo
- a Department of Obstetrics & Gynecology ; Center for Interdisciplinary Research in Women's Health; The University of Texas Medical Branch ; Galveston , TX USA
| | - Jacqueline M Hirth
- a Department of Obstetrics & Gynecology ; Center for Interdisciplinary Research in Women's Health; The University of Texas Medical Branch ; Galveston , TX USA
| | - Abbey B Berenson
- a Department of Obstetrics & Gynecology ; Center for Interdisciplinary Research in Women's Health; The University of Texas Medical Branch ; Galveston , TX USA
| |
Collapse
|
41
|
Stanley M. HPV vaccines: how many doses are needed for protection? Future Virol 2016. [DOI: 10.2217/fvl-2016-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
HPV virus-like particle vaccines are highly immunogenic, well tolerated and are in the national immunization programs in more than 62 countries. Genital HPV is a sexually transmitted infection with first infection occurring just after the onset of sexual activity. The routine cohort for immunization in almost all countries are adolescent girls 9–15 years of age with or without catch-up for older adolescents and young women. In countries with vaccine coverage exceeding 50%, reductions in vaccine type HPV geno-prevalence and disease are being shown. The mechanism of protection is assumed to be via neutralizing antibody. Antibody concentration in adolescents less than 14 years of age after two doses of vaccine at 0 and 6 months are noninferior to women after three doses and in whom efficacy was demonstrated in randomized control trials. The original three-dose schedules have already been reduced in many countries, for those 14 years of age and under, to two doses at least 6 months apart for the licensed vaccines Cervarix® and GARDASIL®. There is preliminary evidence that one dose of vaccine is as effective as two or three doses at preventing persistent HPV infection in the cervix in young women and a one-dose schedule may be possible if supported by evidence from randomized controlled trials.
Collapse
Affiliation(s)
- Margaret Stanley
- Department of Pathology, Tennis Court Road, Cambridge, CB2 1QP, UK
| |
Collapse
|
42
|
Mensah FA, Mehta MR, Lewis JS, Lockhart AC. The Human Papillomavirus Vaccine: Current Perspective and Future Role in Prevention and Treatment of Anal Intraepithelial Neoplasia and Anal Cancer. Oncologist 2016; 21:453-60. [PMID: 26961923 DOI: 10.1634/theoncologist.2015-0075] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 01/06/2016] [Indexed: 01/22/2023] Open
Abstract
UNLABELLED The incidences of human papillomavirus (HPV)-related anal cancer and its precursor lesion, anal intraepithelial neoplasia, are rising in the U.S. and globally. Five-year survival rates with current modalities of treatment for anal cancer are generally favorable for localized and regional disease. For metastatic disease, the relative survival rate is poor. Major contributing factors for the increase in anal cancer incidence include increasing receptive anal intercourse (hetero- and homosexual), increasing HPV infections, and longer life expectancy of treated people who are seropositive for human immunodeficiency virus. Because treatment outcomes with systemic therapy in patients with advanced disease are so poor, prevention may be the best approach for reducing disease burden. The association of a major causative agent with anal cancer provides an excellent opportunity for prevention and treatment. The advent of the HPV vaccine for anal cancer prevention and treatment is a significant milestone and has the potential to greatly impact these cancers. The data regarding potential use of the HPV vaccine in anal cancer prevention and treatment are reviewed. IMPLICATIONS FOR PRACTICE The incidences of human papillomavirus (HPV)-related anal cancer and its precursor lesion, anal intraepithelial neoplasia, are on the rise in the U.S. and globally. Based on recent studies, the HPV vaccine is approved for prevention of the infection and development of HPV-related anal cancer. In addition, several small studies have shown that the vaccine may be useful as adjuvant therapy for anal cancer. There is a need for public health strategies aimed at education of both patients and practitioners to improve the use of the vaccine for prevention of HPV-related anal cancer. The development of a therapeutic vaccine is a work in progress.
Collapse
Affiliation(s)
- Felix A Mensah
- Division of Geriatric Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, USA Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Mudresh R Mehta
- Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - James S Lewis
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - A Craig Lockhart
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
43
|
Shirbaghaee Z, Bolhassani A. Different applications of virus-like particles in biology and medicine: Vaccination and delivery systems. Biopolymers 2016; 105:113-32. [PMID: 26509554 PMCID: PMC7161881 DOI: 10.1002/bip.22759] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 10/25/2015] [Accepted: 10/25/2015] [Indexed: 12/17/2022]
Abstract
Virus-like particles (VLPs) mimic the whole construct of virus particles devoid of viral genome as used in subunit vaccine design. VLPs can elicit efficient protective immunity as direct immunogens compared to soluble antigens co-administered with adjuvants in several booster injections. Up to now, several prokaryotic and eukaryotic systems such as insect, yeast, plant, and E. coli were used to express recombinant proteins, especially for VLP production. Recent studies are also generating VLPs in plants using different transient expression vectors for edible vaccines. VLPs and viral particles have been applied for different functions such as gene therapy, vaccination, nanotechnology, and diagnostics. Herein, we describe VLP production in different systems as well as its applications in biology and medicine.
Collapse
Affiliation(s)
- Zeinab Shirbaghaee
- Department of Hepatitis and AIDSPasteur Institute of IranTehranIran
- Department of Immunology, School of Public HealthTehran University of Medical SciencesTehranIran
| | - Azam Bolhassani
- Department of Hepatitis and AIDSPasteur Institute of IranTehranIran
| |
Collapse
|
44
|
Brickman C, Palefsky JM. Human papillomavirus in the HIV-infected host: epidemiology and pathogenesis in the antiretroviral era. Curr HIV/AIDS Rep 2016; 12:6-15. [PMID: 25644977 DOI: 10.1007/s11904-014-0254-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human papillomavirus (HPV) infection is associated with essentially all cervical cancers, 80-90 % of anal cancers, and a high proportion of oropharyngeal, vaginal, penile, and vulvar cancers. Malignancy is preceded by the development of precancerous lesions termed high-grade squamous intraepithelial lesions (HSIL). Men and women with human immunodeficiency virus (HIV) infection are at high risk of HPV-related malignancies. The incidence of anal cancer in particular has markedly risen during the antiretroviral era due to the increased longevity of patients with HIV and the absence of anal malignancy screening programs. HIV infection may facilitate initial HPV infection by disrupting epithelial cell tight junctions. Once infection is established, HIV may promote HSIL development via the up-regulation of HPV oncogene expression and impairment of the immune response needed to clear the lesion. HIV-infected women should be screened for cervical HSIL and cancer, and HIV-infected men and women should be considered for anal screening programs.
Collapse
Affiliation(s)
- Cristina Brickman
- University of California San Francisco, Box 0654 513, Parnassus Ave, Medical Science Room 420E, San Francisco, CA, 94143, USA,
| | | |
Collapse
|
45
|
Klasse PJ. How to assess the binding strength of antibodies elicited by vaccination against HIV and other viruses. Expert Rev Vaccines 2016; 15:295-311. [PMID: 26641943 DOI: 10.1586/14760584.2016.1128831] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Vaccines that protect against viral infections generally induce neutralizing antibodies. When vaccines are evaluated, the need arises to assess the affinity maturation of the antibody responses. Binding titers of polyclonal sera depend not only on the affinities of the constituent antibodies but also on their individual concentrations, which are difficult to ascertain. Therefore an assay based on chaotrope disruption of antibody-antigen complexes was designed for measuring binding strength. This assay works well with many viral antigens but gives differential results depending on the conformational dependence of epitopes on complex antigens such as the envelope glycoprotein of HIV-1. Kinetic binding assays might offer alternatives, since they can measure average off-rate constants for polyclonal antibodies in a serum. Here, potentials and fallacies of these techniques are discussed.
Collapse
Affiliation(s)
- P J Klasse
- a Department of Microbiology and Immunology, Weill Cornell Medical College , Cornell University , New York , NY , USA
| |
Collapse
|
46
|
Shirbaghaee Z, Bolhassani A, Mirshafiey A, Motevalli F, Zohrei N. A Live Vector Expressing HPV16 L1 Generates an Adjuvant-Induced Antibody Response In-vivo. IRANIAN JOURNAL OF CANCER PREVENTION 2015; 8:e3991. [PMID: 26855722 PMCID: PMC4736072 DOI: 10.17795/ijcp-3991] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 10/07/2015] [Indexed: 12/05/2022]
Abstract
Background: The association between human papillomavirus (HPV) infections and cervical cancer has suggested the design of prophylactic and therapeutic vaccines against genital warts. The HPV capsid has made of two L1 and L2 coat proteins that have produced late in viral infections. Regarding to the recent studies, two commercial prophylactic vaccines have based on L1 viral like particles (VLPs) could strongly induce antibody responses, and protect human body from HPV infections. However, the use of these HPV vaccines has hindered due to their high cost and some limitations. Currently, among various vaccination strategies, live vector-based vaccines have attracted a great attention. Objectives: Herein, a non-pathogenic strain of the protozoan organism known as Leishmania tarentolae has utilized to induce potent humoral immunity in mice model. Materials and Methods: At first, cloning of HPV16 L1 gene into Leishmania expression vector has performed and confirmed by PCR and digestion with restriction enzymes. The promastigotes of Leishmania tarentolae (L.tar) have transfected with linearized DNA construct by electroporation. Protein expression has analyzed by SDS-PAGE and western blotting. Then, the immunogenicity of leishmania expressing L1 protein (L.tar-L1) has assessed in mice model. Results: Our data has indicated that subcutaneous immunization of mice with the recombinant L.tar-L1 has led to enhance the levels of IgG1 and lgG2a in comparison with control groups. Furthermore, there was no significant increase in antibody levels between two and three times of immunizations. Conclusions: The recombinant live vector was able to induce humoral immunity in mice without need of any adjuvant. However, further studies have required to increase its efficiency.
Collapse
Affiliation(s)
- Zeinab Shirbaghaee
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, IR Iran; Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, IR Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, IR Iran
| | - Abbas Mirshafiey
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, IR Iran
| | - Fatemeh Motevalli
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, IR Iran
| | - Negar Zohrei
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, IR Iran
| |
Collapse
|
47
|
Lorenzen E, Follmann F, Bøje S, Erneholm K, Olsen AW, Agerholm JS, Jungersen G, Andersen P. Intramuscular Priming and Intranasal Boosting Induce Strong Genital Immunity Through Secretory IgA in Minipigs Infected with Chlamydia trachomatis. Front Immunol 2015; 6:628. [PMID: 26734002 PMCID: PMC4679855 DOI: 10.3389/fimmu.2015.00628] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 11/30/2015] [Indexed: 11/13/2022] Open
Abstract
International efforts in developing a vaccine against Chlamydia trachomatis have highlighted the need for novel immunization strategies for the induction of genital immunity. In this study, we evaluated an intramuscular (IM) prime/intranasal boost vaccination strategy in a Göttingen Minipig model with a reproductive system very similar to humans. The vaccine was composed of C. trachomatis subunit antigens formulated in the Th1/Th17 promoting CAF01 adjuvant. IM priming immunizations with CAF01 induced a significant cell-mediated interferon gamma and interleukin 17A response and a significant systemic high-titered neutralizing IgG response. Following genital challenge, intranasally boosted groups mounted an accelerated, highly significant genital IgA response that correlated with enhanced bacterial clearance on day 3 post infection. By detecting antigen-specific secretory component (SC), we showed that the genital IgA was locally produced in the genital mucosa. The highly significant inverse correlation between the vaginal IgA SC response and the chlamydial load suggests that IgA in the minipig model is involved in protection against C. trachomatis. This is important both for our understanding of protective immunity and future vaccination strategies against C. trachomatis and genital pathogens in general.
Collapse
Affiliation(s)
- Emma Lorenzen
- Section for Veterinary Reproduction and Obstetrics, Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Disease Immunology, Chlamydia Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Frank Follmann
- Department of Infectious Disease Immunology, Chlamydia Vaccine Research, Statens Serum Institut , Copenhagen , Denmark
| | - Sarah Bøje
- Section for Veterinary Reproduction and Obstetrics, Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Disease Immunology, Chlamydia Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Karin Erneholm
- Section for Veterinary Reproduction and Obstetrics, Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Disease Immunology, Chlamydia Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Anja Weinreich Olsen
- Department of Infectious Disease Immunology, Chlamydia Vaccine Research, Statens Serum Institut , Copenhagen , Denmark
| | - Jørgen Steen Agerholm
- Section for Veterinary Reproduction and Obstetrics, Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Gregers Jungersen
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark , Copenhagen , Denmark
| | - Peter Andersen
- Department of Infectious Disease Immunology, Chlamydia Vaccine Research, Statens Serum Institut , Copenhagen , Denmark
| |
Collapse
|
48
|
Kojic EM, Rana AI, Cu-Uvin S. Human papillomavirus vaccination in HIV-infected women: need for increased coverage. Expert Rev Vaccines 2015; 15:105-17. [PMID: 26599305 DOI: 10.1586/14760584.2016.1110025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Human immunodeficiency virus (HIV)-infected women carry a significant burden on human papillomavirus (HPV) infection and associated diseases. As HIV-infected individuals are living longer, the prevalence of HPV infection is rising and HPV-associated cytological abnormalities remain high despite successful treatments of HIV infection. Several HPV vaccines are currently available and recommended for adolescents and adults up to age 26. The vaccines are safe, immunogenic and effective in preventing diseases due to HPV types included in the vaccines, particularly among persons without prior HPV exposure. This review summarizes available data on the use of the HPV vaccines among HIV-infected women. The immunogenicity and safety of the vaccines are highlighted and in particular, barriers to vaccination among HIV-infected women are discussed.
Collapse
Affiliation(s)
- Erna Milunka Kojic
- a Division of Infectious Diseases , Warren Alpert School of Medicine, Brown University , Providence , RI , USA
| | - Aadia I Rana
- a Division of Infectious Diseases , Warren Alpert School of Medicine, Brown University , Providence , RI , USA
| | - Susan Cu-Uvin
- a Division of Infectious Diseases , Warren Alpert School of Medicine, Brown University , Providence , RI , USA
| |
Collapse
|
49
|
Téllez L, Michelli E, Mendoza JA, Vielma S, Noguera ME, Callejas D, Cavazza M, Correnti M. Persistent infection with high-risk human papilloma viruses: cohort study, Mérida, Venezuela. Ecancermedicalscience 2015; 9:579. [PMID: 26557877 PMCID: PMC4631573 DOI: 10.3332/ecancer.2015.579] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Indexed: 11/25/2022] Open
Abstract
Cervical lesions have been associated with infection by high-risk human papilloma virus (high-risk HPV). In 409 women aged >15 years high-risk HPV lesions were identified. In a cohort of this population persistent infection was compared with cytological, colposcopic, and histological lesions. Cervical scrapes were taken and DNA was isolated. HPV was detected by PCR in the E6/E7 region. Genotyping was performed by PCR nested multiple E6/E7. HPV was detected in a 37.40% (153/409), high-risk HPV in 86% (153/178), HPV18 46.64% (83/178), HPV16 34.28% (61/178). Among these 53.93% (96/178) were multiple infections, and HPV18/16 (30/96) was the most frequent 31.25%. The cytology showed changes in 15% of positive patients. A 49.67% in women positive for HPV infection showed abnormalities in the colposcopic study, a relationship that turned out to be statistically significant ( p < 0.0019 test χ2). Among all 85% of the women were younger than 45 years of age. Fifty-seven patients were evaluated 15 months after the base study, with initial prevalence of morbidity 49.12% (28/57) and at the end 10.53% (6/57), showing in 89.29% (25/28) negative for HR-HPV infection, 10.34% (3/28) showed persistence of infection, 17.54% (10/57) presented cytological alterations, with 80% of positivity for HPV, and a regression of 100% (10/10) of the previously identified lesions. With colposcopy, 50% (14/28) presented alterations related to HPV, of these 85.71% (12/14) showed regression of such an alteration. The cumulative incidence for HPV was 10.34% (3/29). The incidence rate was 4.23% (3/71), which is equal to 4.23 new cases of HPV infection per 100 people, per year of follow-up. In conclusion, the present work shows a high frequency of infection by high-risk HPV, with predominance of HPV18 and 16 and in general for multiple infections. Colposcopy was better predictor than the Pap smear for infection. The follow-up study revealed a low percentage of persistent infection, and a high frequency of negativity for viral infection, high regression of cytological and colposcopic lesions, a low cumulative and incidence rate similar to that reported by other Latin American countries and higher than the European countries.
Collapse
Affiliation(s)
- Luis Téllez
- Los Andes University, Department of Microbiology, Mérida CP 5101, Venezuela
| | - Elvia Michelli
- Los Andes University, Department of Microbiology, Mérida CP 5101, Venezuela ; University of Orient, Department of Bioanalysis, Sucre CP 6101, Venezuela
| | | | - Silvana Vielma
- Los Andes University, Department of Microbiology, Mérida CP 5101, Venezuela
| | - María-Eugenia Noguera
- Los Andes University, Department of Gynecology and Obstetrics, Mérida CP 5101, Venezuela
| | - Diana Callejas
- University of Zulia (LUZ), Regional Reference Virology Laboratory, Maracaibo CP 4011, Zulia
| | - María Cavazza
- Institute of Biomedicine, MPPS, Caracas CP 10104, Venezuela
| | - María Correnti
- Institute of Oncology and Haematology, MPPS, Caracas CP 1050, Venezuela
| |
Collapse
|
50
|
Davitt CJ, Lavelle EC. Delivery strategies to enhance oral vaccination against enteric infections. Adv Drug Deliv Rev 2015; 91:52-69. [PMID: 25817337 DOI: 10.1016/j.addr.2015.03.007] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/25/2015] [Accepted: 03/12/2015] [Indexed: 01/22/2023]
Abstract
While the majority of human pathogens infect the body through mucosal sites, most licensed vaccines are injectable. In fact the only mucosal vaccine that has been widely used globally for infant and childhood vaccination programs is the oral polio vaccine (OPV) developed by Albert Sabin in the 1950s. While oral vaccines against Cholera, rotavirus and Salmonella typhi have also been licensed, the development of additional non-living oral vaccines against these and other enteric pathogens has been slow and challenging. Mucosal vaccines can elicit protective immunity at the gut mucosa, in part via antigen-specific secretory immunoglobulin A (SIgA). However, despite their advantages over the injectable route, oral vaccines face many hurdles. A key challenge lies in design of delivery strategies that can protect antigens from degradation in the stomach and intestine, incorporate appropriate immune-stimulatory adjuvants and control release at the appropriate gastrointestinal site. A number of systems including micro and nanoparticles, lipid-based strategies and enteric capsules have significant potential either alone or in advanced combined formulations to enhance intestinal immune responses. In this review we will outline the opportunities, challenges and potential delivery solutions to facilitate the development of improved oral vaccines for infectious enteric diseases.
Collapse
|