951
|
An open-label, randomized study of a 9-valent human papillomavirus vaccine given concomitantly with diphtheria, tetanus, pertussis and poliomyelitis vaccines to healthy adolescents 11-15 years of age. Pediatr Infect Dis J 2015; 34:627-34. [PMID: 25831420 DOI: 10.1097/inf.0000000000000694] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND A 9-valent human papillomavirus (9vHPV) vaccine has recently been reported to be safe and highly efficacious against infection and disease related to HPV6/11/16/18/31/33/45/52/58. We evaluated the immunogenicity and safety of the 9vHPV vaccine administered concomitantly with REPEVAX (diphtheria, tetanus, acellular pertussis and inactivated poliomyelitis vaccine). METHODS This open-label, randomized, multicenter study enrolled 1054 males and females ages 11-15 years. Subjects were randomly assigned to each group in a 1:1 ratio. Subjects received a 0.5 mL dose of 9vHPV vaccine intramuscularly at day 1, months 2 and 6 and a 0.5 mL dose of REPEVAX either on day 1 (concomitant vaccination group; n = 526) or at month 1 (nonconcomitant vaccination group, n = 528). Serologic responses for each vaccine component were tested by 1-sided tests of noninferiority between groups. Systemic and injection-site adverse experiences (AEs) and serious AEs were monitored. RESULTS Noninferiority of anti-HPV geometric mean titers and seroconversion rates for all 9vHPV antigens were demonstrated for the concomitant group compared with the nonconcomitant group. Seroconversion rates for the 9vHPV vaccine types were ≥99.8% in both groups at month 7. For REPEVAX, noninferiority of immune response was established for diphtheria, tetanus, all polio and pertussis antigens for both groups. There were no vaccine-related serious AEs. CONCLUSION Overall, concomitant administration of 9vHPV vaccine and REPEVAX was generally well tolerated and did not interfere with the immune response to either vaccine. This strategy would minimize the number of visits required to deliver each vaccine individually.
Collapse
|
952
|
Frimer M, Sun C, McAndrew T, Smith B, Harari A, Chen Z, Mirabello L, Wentzensen N, Goldberg GL, Rodriguez AC, Schiffman M, Burk RD. HPV16 CpG methyl-haplotypes are associated with cervix precancer and cancer in the Guanacaste natural history study. Gynecol Oncol 2015; 138:94-100. [PMID: 26001326 DOI: 10.1016/j.ygyno.2015.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/02/2015] [Indexed: 01/14/2023]
Abstract
OBJECTIVE To evaluate HPV16 CpG methylation and methyl-haplotypes and their association with cervix precancer and cancer utilizing massively parallel single molecule next-generation sequencing (NGS). METHODS A nested case-control study of HPV16 positive women was performed in a prospective cohort from Guanacaste, Costa Rica designed to study the natural history of HPV and cervical neoplasia. Controls encompassed 31 women with transient infections; there were 44 cases, including 31 women with CIN3 and 13 with cervical cancer. DNA samples from exfoliated cervical cells were treated with bisulfite and four regions (E6, E2, L2 and L1) were amplified with barcoded primers and tested by NGS. CpG methylation was quantified using a bioinformatics pipeline. RESULTS Median methylation levels were significantly different between the CIN3+ cases versus controls in the E2, L2, and L1 regions. Methyl-haplotypes, specifically in 5 CpG sites included in the targeted L2 region, with the pattern "--+-+" had the highest Area Under the Curve value (AUC=88.40%) observed for CIN3 vs. CONTROLS The most significant CpG site, L2 4277, determined by bisulfite NGS had an AUC=78.62%. CONCLUSIONS This study demonstrates that NGS of bisulfite treated HPV DNA is a useful and efficient technique to survey methylation patterns in HPV16. This procedure provides quantitative information on both individual CpG sites and methyl-haplotypes that identify women with elevated present or subsequent risk for HPV16 CIN3 and cancer.
Collapse
Affiliation(s)
- Marina Frimer
- Division of Gynecologic Oncology, Department of Obstetrics & Gynecology and Women's Health, Albert Einstein College of Medicine/Montefiore Medical Center, United States.
| | - Chang Sun
- Department of Pediatrics, Albert Einstein College of Medicine, United States
| | - Thomas McAndrew
- Department of Obstetrics & Gynecology and Women's Health, Albert Einstein College of Medicine, United States
| | - Benjamin Smith
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, United States
| | - Ariana Harari
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, United States
| | - Zigui Chen
- Department of Pediatrics, Albert Einstein College of Medicine, United States
| | - Lisa Mirabello
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, United States
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, United States
| | - Gary L Goldberg
- Division of Gynecologic Oncology, Department of Obstetrics & Gynecology and Women's Health, Albert Einstein College of Medicine/Montefiore Medical Center, United States
| | - Ana C Rodriguez
- Proyecto Epidemiologico Guanacaste, Fundacion INCIENSA, San Jose, Costa Rica
| | - Mark Schiffman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, United States
| | - Robert D Burk
- Department of Pediatrics, Albert Einstein College of Medicine, United States; Department of Obstetrics & Gynecology and Women's Health, Albert Einstein College of Medicine, United States; Department of Microbiology and Immunology, Albert Einstein College of Medicine, United States
| |
Collapse
|
953
|
Bogaards JA, Wallinga J, Brakenhoff RH, Meijer CJLM, Berkhof J. Direct benefit of vaccinating boys along with girls against oncogenic human papillomavirus: bayesian evidence synthesis. BMJ 2015; 350:h2016. [PMID: 25985328 PMCID: PMC4428278 DOI: 10.1136/bmj.h2016] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/16/2015] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To assess the reduction in the vaccine preventable burden of cancer in men if boys are vaccinated along with girls against oncogenic human papillomavirus (HPV). DESIGN Bayesian evidence synthesis approach used to evaluate the impact of vaccination against HPV types 16 and 18 on the burden of anal, penile, and oropharyngeal carcinomas among heterosexual men and men who have sex with men. The reduced transmission of vaccine-type HPV from vaccination of girls was assumed to lower the risk of HPV associated cancer in all men but not to affect the excess risk of HPV associated cancers among men who have sex with men. SETTING General population in the Netherlands. INTERVENTION Inclusion of boys aged 12 into HPV vaccination programmes. MAIN OUTCOME MEASURES Quality adjusted life years (QALYs) and numbers needed to vaccinate. RESULTS Before HPV vaccination, 14.9 (95% credible interval 12.2 to 18.1) QALYs per thousand men were lost to vaccine preventable cancers associated with HPV in the Netherlands. This burden would be reduced by 37% (28% to 48%) if the vaccine uptake among girls remains at the current level of 60%. To prevent one additional case of cancer among men, 795 boys (660 to 987) would need to be vaccinated; with tumour specific numbers for anal, penile, and oropharyngeal cancer of 2162, 3486, and 1975, respectively. The burden of HPV related cancer in men would be reduced by 66% (53% to 805) if vaccine uptake among girls increases to 90%. In that case, 1735 boys (1240 to 2900) would need to be vaccinated to prevent an additional case; with tumour specific numbers for anal, penile, and oropharyngeal cancer of 2593, 29107, and 6484, respectively. CONCLUSIONS Men will benefit indirectly from vaccination of girls but remain at risk of cancers associated with HPV. The incremental benefit of vaccinating boys when vaccine uptake among girls is high is driven by the prevention of anal carcinomas, which underscores the relevance of HPV prevention efforts for men who have sex with men.
Collapse
Affiliation(s)
- Johannes A Bogaards
- Department of Epidemiology and Biostatistics, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, Netherlands Centre for Infectious Disease Control, National Institute for Public Health and the Environment, PO Box 1, 3720 BA Bilthoven, Netherlands
| | - Jacco Wallinga
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, PO Box 1, 3720 BA Bilthoven, Netherlands
| | - Ruud H Brakenhoff
- Department of Otolaryngology/Head and Neck Surgery, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, Netherlands
| | - Chris J L M Meijer
- Department of Pathology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, Netherlands
| | - Johannes Berkhof
- Department of Epidemiology and Biostatistics, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, Netherlands
| |
Collapse
|
954
|
Di Bonito P, Petrone L, Casini G, Francolini I, Ammendolia MG, Accardi L, Piozzi A, D'Ilario L, Martinelli A. Amino-functionalized poly(L-lactide) lamellar single crystals as a valuable substrate for delivery of HPV16-E7 tumor antigen in vaccine development. Int J Nanomedicine 2015; 10:3447-58. [PMID: 26056443 PMCID: PMC4431504 DOI: 10.2147/ijn.s76023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Poly(l-lactide) (PLLA) is a biodegradable polymer currently used in many biomedical applications, including the production of resorbable surgical devices, porous scaffolds for tissue engineering, nanoparticles and microparticles for the controlled release of drugs or antigens. The surfaces of lamellar PLLA single crystals (PLLAsc) were provided with amino groups by reaction with a multifunctional amine and used to adsorb an Escherichia coli-produced human papillomavirus (HPV)16-E7 protein to evaluate its possible use in antigen delivery for vaccine development. Methods PLLA single crystals were made to react with tetraethylenepentamine to obtain amino-functionalized PLLA single crystals (APLLAsc). Pristine and amino-functionalized PLLAsc showed a two-dimensional microsized and one-dimensional nanosized lamellar morphology, with a lateral dimension of about 15–20 μm, a thickness of about 12 nm, and a surface specific area of about 130 m2/g. Both particles were characterized and loaded with HPV16-E7 before being administered to C57BL/6 mice for immunogenicity studies. The E7-specific humoral-mediated and cell-mediated immune response as well as tumor protective immunity were analyzed in mice challenged with TC-1 cancer cells. Results Pristine and amino-functionalized PLLAsc adsorbed similar amounts of E7 protein, but in protein-release experiments E7-PLLAsc released a higher amount of protein than E7-APLLAsc. When the complexes were dried for observation by scanning electron microscopy, both samples showed a compact layer, but E7-APLLAsc showed greater roughness than E7-PLLAsc. Immunization experiments in mice showed that E7-APLLAsc induced a stronger E7-specific immune response when compared with E7-PLLAsc. Immunoglobulin G isotyping and interferon gamma analysis suggested a mixed Th1/Th2 immune response in both E7-PLLAsc-immunized and E7-APLLAsc-immunized mice. However, only the mice receiving E7-APLLAsc were fully protected from TC-1 tumor growth after three doses of vaccine. Conclusion Our results show that APLLA single crystals improve the immunogenicity of HPV16-E7 and indicate that E7-APLLAsc could be used for development of an HPV16 therapeutic vaccine against HPV16-related tumors.
Collapse
Affiliation(s)
- Paola Di Bonito
- Department of Infectious, Parasitic and Immune-mediated Diseases, Italian National Institute of Health, Rome, Italy
| | - Linda Petrone
- Department of Infectious, Parasitic and Immune-mediated Diseases, Italian National Institute of Health, Rome, Italy
| | - Gabriele Casini
- Department of Chemistry, Sapienza University of Rome, Rome, Italy
| | | | | | - Luisa Accardi
- Department of Infectious, Parasitic and Immune-mediated Diseases, Italian National Institute of Health, Rome, Italy
| | - Antonella Piozzi
- Department of Chemistry, Sapienza University of Rome, Rome, Italy
| | - Lucio D'Ilario
- Department of Chemistry, Sapienza University of Rome, Rome, Italy
| | | |
Collapse
|
955
|
Walwyn L, Janusz CB, Clark AD, Prieto E, Waight E, Largaespada N. Cost-effectiveness of HPV vaccination in Belize. Vaccine 2015; 33 Suppl 1:A174-81. [DOI: 10.1016/j.vaccine.2014.12.042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 12/12/2014] [Accepted: 12/15/2014] [Indexed: 11/27/2022]
|
956
|
Herrero R, González P, Markowitz LE. Present status of human papillomavirus vaccine development and implementation. Lancet Oncol 2015; 16:e206-16. [PMID: 25943065 DOI: 10.1016/s1470-2045(14)70481-4] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Oncogenic human papillomavirus (HPV) infection is the cause of nearly all cervical cancers and a proportion of other anogenital and oropharyngeal cancers. A bivalent vaccine containing HPV 16 and 18 and a quadrivalent vaccine containing HPV 6, 11, 16, and 18 antigens are in use in vaccination programmes around the world. In clinical trials, three vaccine doses provided 90-100% protection against cervical infection and pre-cancer related to HPV 16 and 18 in women aged 15-26 years who were not infected at vaccination. Partial cross-protection against other HPV types has been reported but its duration is unknown. The vaccines were also efficacious at the prevention of HPV 16 and 18 infections at other anatomical sites in both sexes. Immunobridging studies allowed licensing of the vaccines for use starting at age 9 years for both sexes. Two-dose schedules elicit high antibody concentrations, leading to the recommendation of two-dose schedules for girls aged 9-14 years. Pre-licensure and post-licensure studies have provided data supporting vaccine safety. In 2014, a nonavalent vaccine containing HPV 6, 11, 16, 18, 31, 33, 45, 52, and 58 antigens was licensed by the US Food and Drug Administration. HPV vaccination was first introduced in high-income countries owing to vaccine cost, logistic challenges, and competing health priorities. Since 2011, vaccine prices have lowered, allowing the introduction of the vaccine in some middle-income countries. Funding of the vaccine by the GAVI Alliance in 2012 led to demonstration projects in some low-income countries. By 2014, more than 57 countries had included the HPV vaccine in their national health programmes. Data from several countries have shown the effect of vaccination on HPV infection and associated disease, and provided evidence of herd immunity. Expansion of programmes to countries with the highest burden of disease is beginning, but further efforts are needed to realise the potential of HPV vaccines.
Collapse
Affiliation(s)
- Rolando Herrero
- Prevention and Implementation Group, Section of Early Detection and Prevention, International Agency for Research on Cancer, Lyon, France.
| | - Paula González
- Prevention and Implementation Group, Section of Early Detection and Prevention, International Agency for Research on Cancer, Lyon, France; Proyecto Epidemiológico Guanacaste, Liberia, Guanacaste, Costa Rica
| | - Lauri E Markowitz
- National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta GA, USA
| |
Collapse
|
957
|
Vacuna frente al virus del papiloma humano. Eficacia y seguridad. Enferm Infecc Microbiol Clin 2015; 33:342-54. [DOI: 10.1016/j.eimc.2015.03.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 03/27/2015] [Indexed: 11/30/2022]
|
958
|
Abstract
Both primary and secondary prevention of cervical cancer are now available. Immunizations against human papillomavirus (HPV) types 16 and 18 have the potential to prevent 70% of cancers of the cervix plus a large percentage of other lower anogenital tract cancers. Screening guidelines were recently changed to recommend cotesting with cytology plus an HPV test. The addition of HPV testing increases the sensitivity and negative predictive value of screening over the Papanicolaou (Pap) test alone.
Collapse
Affiliation(s)
- Lauren Thaxton
- Department of Obstetrics and Gynecology, 1 University of New Mexico, MSC 10 5580, Albuquerque, NM 87131, USA
| | - Alan G Waxman
- Department of Obstetrics and Gynecology, 1 University of New Mexico, MSC 10 5580, Albuquerque, NM 87131, USA.
| |
Collapse
|
959
|
Saraiya M, Unger ER, Thompson TD, Lynch CF, Hernandez BY, Lyu CW, Steinau M, Watson M, Wilkinson EJ, Hopenhayn C, Copeland G, Cozen W, Peters ES, Huang Y, Saber MS, Altekruse S, Goodman MT. US assessment of HPV types in cancers: implications for current and 9-valent HPV vaccines. J Natl Cancer Inst 2015; 107:djv086. [PMID: 25925419 DOI: 10.1093/jnci/djv086] [Citation(s) in RCA: 515] [Impact Index Per Article: 57.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 03/03/2015] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND This study sought to determine the prevaccine type-specific prevalence of human papillomavirus (HPV)-associated cancers in the United States to evaluate the potential impact of the HPV types in the current and newly approved 9-valent HPV vaccines. METHODS The Centers for Disease Control and Prevention partnered with seven US population-based cancer registries to obtain archival tissue for cancers diagnosed from 1993 to 2005. HPV testing was performed on 2670 case patients that were fairly representative of all participating cancer registry cases by age and sex. Demographic and clinical data were evaluated by anatomic site and HPV status. Current US cancer registry data and the detection of HPV types were used to estimate the number of cancers potentially preventable through vaccination. RESULTS HPV DNA was detected in 90.6% of cervical, 91.1% of anal, 75.0% of vaginal, 70.1% of oropharyngeal, 68.8% of vulvar, 63.3% of penile, 32.0% of oral cavity, and 20.9% of laryngeal cancers, as well as in 98.8% of cervical cancer in situ (CCIS). A vaccine targeting HPV 16/18 potentially prevents the majority of invasive cervical (66.2%), anal (79.4%), oropharyngeal (60.2%), and vaginal (55.1%) cancers, as well as many penile (47.9%), vulvar (48.6%) cancers: 24 858 cases annually. The 9-valent vaccine also targeting HPV 31/33/45/52/58 may prevent an additional 4.2% to 18.3% of cancers: 3944 cases annually. For most cancers, younger age at diagnosis was associated with higher HPV 16/18 prevalence. With the exception of oropharyngeal cancers and CCIS, HPV 16/18 prevalence was similar across racial/ethnic groups. CONCLUSIONS In the United States, current vaccines will reduce most HPV-associated cancers; a smaller additional reduction would be contributed by the new 9-valent vaccine.
Collapse
Affiliation(s)
- Mona Saraiya
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion (MSa, TDT, MW) and Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (ERU, MSt), Centers for Disease Control and Prevention, Atlanta, GA; University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI (MTG, BYH); Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA (CFL); Departments of Preventive Medicine (WC) and Pathology (WC, MSS), University of Southern California, Los Angeles, CA; Department of Epidemiology, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA (ESP); Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL (EJW); Michigan Department of Community Health, Lansing, MI (GC); Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY (CH); Florida Department of Health, Tallahassee, FL (YH); Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD (SFA); Battelle Memorial Institute, Durham, NC (CWL); National Cancer Institute (SA).
| | - Elizabeth R Unger
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion (MSa, TDT, MW) and Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (ERU, MSt), Centers for Disease Control and Prevention, Atlanta, GA; University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI (MTG, BYH); Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA (CFL); Departments of Preventive Medicine (WC) and Pathology (WC, MSS), University of Southern California, Los Angeles, CA; Department of Epidemiology, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA (ESP); Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL (EJW); Michigan Department of Community Health, Lansing, MI (GC); Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY (CH); Florida Department of Health, Tallahassee, FL (YH); Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD (SFA); Battelle Memorial Institute, Durham, NC (CWL); National Cancer Institute (SA)
| | - Trevor D Thompson
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion (MSa, TDT, MW) and Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (ERU, MSt), Centers for Disease Control and Prevention, Atlanta, GA; University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI (MTG, BYH); Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA (CFL); Departments of Preventive Medicine (WC) and Pathology (WC, MSS), University of Southern California, Los Angeles, CA; Department of Epidemiology, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA (ESP); Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL (EJW); Michigan Department of Community Health, Lansing, MI (GC); Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY (CH); Florida Department of Health, Tallahassee, FL (YH); Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD (SFA); Battelle Memorial Institute, Durham, NC (CWL); National Cancer Institute (SA)
| | - Charles F Lynch
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion (MSa, TDT, MW) and Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (ERU, MSt), Centers for Disease Control and Prevention, Atlanta, GA; University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI (MTG, BYH); Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA (CFL); Departments of Preventive Medicine (WC) and Pathology (WC, MSS), University of Southern California, Los Angeles, CA; Department of Epidemiology, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA (ESP); Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL (EJW); Michigan Department of Community Health, Lansing, MI (GC); Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY (CH); Florida Department of Health, Tallahassee, FL (YH); Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD (SFA); Battelle Memorial Institute, Durham, NC (CWL); National Cancer Institute (SA)
| | - Brenda Y Hernandez
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion (MSa, TDT, MW) and Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (ERU, MSt), Centers for Disease Control and Prevention, Atlanta, GA; University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI (MTG, BYH); Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA (CFL); Departments of Preventive Medicine (WC) and Pathology (WC, MSS), University of Southern California, Los Angeles, CA; Department of Epidemiology, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA (ESP); Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL (EJW); Michigan Department of Community Health, Lansing, MI (GC); Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY (CH); Florida Department of Health, Tallahassee, FL (YH); Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD (SFA); Battelle Memorial Institute, Durham, NC (CWL); National Cancer Institute (SA)
| | - Christopher W Lyu
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion (MSa, TDT, MW) and Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (ERU, MSt), Centers for Disease Control and Prevention, Atlanta, GA; University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI (MTG, BYH); Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA (CFL); Departments of Preventive Medicine (WC) and Pathology (WC, MSS), University of Southern California, Los Angeles, CA; Department of Epidemiology, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA (ESP); Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL (EJW); Michigan Department of Community Health, Lansing, MI (GC); Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY (CH); Florida Department of Health, Tallahassee, FL (YH); Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD (SFA); Battelle Memorial Institute, Durham, NC (CWL); National Cancer Institute (SA)
| | - Martin Steinau
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion (MSa, TDT, MW) and Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (ERU, MSt), Centers for Disease Control and Prevention, Atlanta, GA; University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI (MTG, BYH); Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA (CFL); Departments of Preventive Medicine (WC) and Pathology (WC, MSS), University of Southern California, Los Angeles, CA; Department of Epidemiology, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA (ESP); Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL (EJW); Michigan Department of Community Health, Lansing, MI (GC); Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY (CH); Florida Department of Health, Tallahassee, FL (YH); Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD (SFA); Battelle Memorial Institute, Durham, NC (CWL); National Cancer Institute (SA)
| | - Meg Watson
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion (MSa, TDT, MW) and Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (ERU, MSt), Centers for Disease Control and Prevention, Atlanta, GA; University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI (MTG, BYH); Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA (CFL); Departments of Preventive Medicine (WC) and Pathology (WC, MSS), University of Southern California, Los Angeles, CA; Department of Epidemiology, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA (ESP); Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL (EJW); Michigan Department of Community Health, Lansing, MI (GC); Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY (CH); Florida Department of Health, Tallahassee, FL (YH); Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD (SFA); Battelle Memorial Institute, Durham, NC (CWL); National Cancer Institute (SA)
| | - Edward J Wilkinson
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion (MSa, TDT, MW) and Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (ERU, MSt), Centers for Disease Control and Prevention, Atlanta, GA; University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI (MTG, BYH); Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA (CFL); Departments of Preventive Medicine (WC) and Pathology (WC, MSS), University of Southern California, Los Angeles, CA; Department of Epidemiology, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA (ESP); Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL (EJW); Michigan Department of Community Health, Lansing, MI (GC); Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY (CH); Florida Department of Health, Tallahassee, FL (YH); Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD (SFA); Battelle Memorial Institute, Durham, NC (CWL); National Cancer Institute (SA)
| | - Claudia Hopenhayn
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion (MSa, TDT, MW) and Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (ERU, MSt), Centers for Disease Control and Prevention, Atlanta, GA; University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI (MTG, BYH); Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA (CFL); Departments of Preventive Medicine (WC) and Pathology (WC, MSS), University of Southern California, Los Angeles, CA; Department of Epidemiology, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA (ESP); Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL (EJW); Michigan Department of Community Health, Lansing, MI (GC); Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY (CH); Florida Department of Health, Tallahassee, FL (YH); Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD (SFA); Battelle Memorial Institute, Durham, NC (CWL); National Cancer Institute (SA)
| | - Glenn Copeland
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion (MSa, TDT, MW) and Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (ERU, MSt), Centers for Disease Control and Prevention, Atlanta, GA; University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI (MTG, BYH); Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA (CFL); Departments of Preventive Medicine (WC) and Pathology (WC, MSS), University of Southern California, Los Angeles, CA; Department of Epidemiology, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA (ESP); Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL (EJW); Michigan Department of Community Health, Lansing, MI (GC); Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY (CH); Florida Department of Health, Tallahassee, FL (YH); Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD (SFA); Battelle Memorial Institute, Durham, NC (CWL); National Cancer Institute (SA)
| | - Wendy Cozen
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion (MSa, TDT, MW) and Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (ERU, MSt), Centers for Disease Control and Prevention, Atlanta, GA; University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI (MTG, BYH); Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA (CFL); Departments of Preventive Medicine (WC) and Pathology (WC, MSS), University of Southern California, Los Angeles, CA; Department of Epidemiology, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA (ESP); Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL (EJW); Michigan Department of Community Health, Lansing, MI (GC); Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY (CH); Florida Department of Health, Tallahassee, FL (YH); Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD (SFA); Battelle Memorial Institute, Durham, NC (CWL); National Cancer Institute (SA)
| | - Edward S Peters
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion (MSa, TDT, MW) and Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (ERU, MSt), Centers for Disease Control and Prevention, Atlanta, GA; University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI (MTG, BYH); Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA (CFL); Departments of Preventive Medicine (WC) and Pathology (WC, MSS), University of Southern California, Los Angeles, CA; Department of Epidemiology, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA (ESP); Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL (EJW); Michigan Department of Community Health, Lansing, MI (GC); Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY (CH); Florida Department of Health, Tallahassee, FL (YH); Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD (SFA); Battelle Memorial Institute, Durham, NC (CWL); National Cancer Institute (SA)
| | - Youjie Huang
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion (MSa, TDT, MW) and Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (ERU, MSt), Centers for Disease Control and Prevention, Atlanta, GA; University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI (MTG, BYH); Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA (CFL); Departments of Preventive Medicine (WC) and Pathology (WC, MSS), University of Southern California, Los Angeles, CA; Department of Epidemiology, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA (ESP); Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL (EJW); Michigan Department of Community Health, Lansing, MI (GC); Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY (CH); Florida Department of Health, Tallahassee, FL (YH); Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD (SFA); Battelle Memorial Institute, Durham, NC (CWL); National Cancer Institute (SA)
| | - Maria Sibug Saber
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion (MSa, TDT, MW) and Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (ERU, MSt), Centers for Disease Control and Prevention, Atlanta, GA; University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI (MTG, BYH); Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA (CFL); Departments of Preventive Medicine (WC) and Pathology (WC, MSS), University of Southern California, Los Angeles, CA; Department of Epidemiology, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA (ESP); Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL (EJW); Michigan Department of Community Health, Lansing, MI (GC); Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY (CH); Florida Department of Health, Tallahassee, FL (YH); Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD (SFA); Battelle Memorial Institute, Durham, NC (CWL); National Cancer Institute (SA)
| | - Sean Altekruse
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion (MSa, TDT, MW) and Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (ERU, MSt), Centers for Disease Control and Prevention, Atlanta, GA; University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI (MTG, BYH); Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA (CFL); Departments of Preventive Medicine (WC) and Pathology (WC, MSS), University of Southern California, Los Angeles, CA; Department of Epidemiology, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA (ESP); Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL (EJW); Michigan Department of Community Health, Lansing, MI (GC); Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY (CH); Florida Department of Health, Tallahassee, FL (YH); Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD (SFA); Battelle Memorial Institute, Durham, NC (CWL); National Cancer Institute (SA)
| | - Marc T Goodman
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion (MSa, TDT, MW) and Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (ERU, MSt), Centers for Disease Control and Prevention, Atlanta, GA; University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI (MTG, BYH); Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA (CFL); Departments of Preventive Medicine (WC) and Pathology (WC, MSS), University of Southern California, Los Angeles, CA; Department of Epidemiology, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA (ESP); Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL (EJW); Michigan Department of Community Health, Lansing, MI (GC); Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY (CH); Florida Department of Health, Tallahassee, FL (YH); Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD (SFA); Battelle Memorial Institute, Durham, NC (CWL); National Cancer Institute (SA)
| | | |
Collapse
|
960
|
Kollipara R, Ekhlassi E, Downing C, Guidry J, Lee M, Tyring SK. Advancements in Pharmacotherapy for Noncancerous Manifestations of HPV. J Clin Med 2015; 4:832-46. [PMID: 26239450 PMCID: PMC4470201 DOI: 10.3390/jcm4050832] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 04/07/2015] [Accepted: 04/14/2015] [Indexed: 12/30/2022] Open
Abstract
Human papillomavirus (HPV) is the most common sexually transmitted disease. Via infection of the basal epithelial cells, HPV causes numerous malignancies and noncancerous cutaneous manifestations. Noncancerous cutaneous manifestations of HPV, including common, plantar, plane, and anogenital warts, are among the most common reasons for an office visit. Although there are various therapies available, they are notoriously difficult to treat. HPV treatments can be grouped into destructive (cantharidin, salicylic acid), virucidal (cidofovir, interferon-α), antimitotic (bleomycin, podophyllotoxin, 5-fluorouracil), immunotherapy (Candida antigen, contact allergen immunotherapy, imiquimod) or miscellaneous (trichloroacetic acid, polyphenon E). The mechanism of action, recent efficacy data, safety profile and recommended regimen for each of these treatment modalities is discussed.
Collapse
Affiliation(s)
| | - Erfon Ekhlassi
- Department of Dermatology, the University of Texas Health Science Center at Houston, Houston 77030, TX, USA.
| | | | | | - Michael Lee
- Center for Clinical Studies, Houston 77004, TX, USA.
| | - Stephen K Tyring
- Center for Clinical Studies, Houston 77004, TX, USA.
- Department of Dermatology, the University of Texas Health Science Center at Houston, Houston 77030, TX, USA.
| |
Collapse
|
961
|
Wang D, Li Z, Xiao J, Wang J, Zhang L, Liu Y, Fan F, Xin L, Wei M, Kong Z, Yu H, Gu Y, Zhang J, Li S, Xia N. Identification of Broad-Genotype HPV L2 Neutralization Site for Pan-HPV Vaccine Development by a Cross-Neutralizing Antibody. PLoS One 2015; 10:e0123944. [PMID: 25905781 PMCID: PMC4408011 DOI: 10.1371/journal.pone.0123944] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 03/09/2015] [Indexed: 02/01/2023] Open
Abstract
Human Papillomavirus (HPV), a non-enveloped, double-stranded DNA virus, is responsible for 5% of human cancers. The HPV capsid consists of major and minor structural proteins, L1 and L2. L1 proteins form an icosahedral shell with building blocks of the pentameric capsomere, and one L2 molecule extends outward from the central hole of the capsid. Thus, L2 is concealed within L1 and only becomes exposed when the capsid interacts with host cells. The low antigenic variation of L2 means that this protein could offer a target for the development of a pan-HPV vaccine. Toward this goal, here we describe an anti-L2 monoclonal antibody, 14H6, which broadly neutralizes at least 11 types of HPV, covering types 6, 11, 16, 18, 31, 33, 35, 45, 52, 58 and 59, in pseudovirion--based cell neutralization assay. The mAb 14H6 recognizes a minimal linear epitope located on amino acids 21 to 30 of the L2 protein. Alanine scanning mutagenesis and sequence alignment identified several conserved residues (Cys22, Lys23, Thr27, Cys28 and Pro29) that are involved in the 14H6 binding with L2. The epitope was grafted to several scaffolding proteins, including HPV16 L1 virus-like particles, HBV 149 core antigen and CRM197. The resultant chimeric constructs were expressed in Escherichia coli and purified with high efficiency. Immunization with these pan-HPV vaccine candidates elicited high titers of the L2-specific antibody in mice and conferred robust (3-log) titers of cross-genotype neutralization, including against HPV11, 16, 18, 45, 52, 58 and 59. These findings will help in the development of an L2-based, pan-HPV vaccine.
Collapse
Affiliation(s)
- Daning Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361005, China
| | - Zhihai Li
- National Institute of Diagnostics and Vaccine Development in Infectious Disease, School of Life Sciences, Xiamen University, Xiamen, 361005, China
| | - Jieqiong Xiao
- National Institute of Diagnostics and Vaccine Development in Infectious Disease, School of Life Sciences, Xiamen University, Xiamen, 361005, China
| | - Junqi Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361005, China
| | - Li Zhang
- National Institute of Diagnostics and Vaccine Development in Infectious Disease, School of Life Sciences, Xiamen University, Xiamen, 361005, China
| | - Yajing Liu
- National Institute of Diagnostics and Vaccine Development in Infectious Disease, School of Life Sciences, Xiamen University, Xiamen, 361005, China
| | - Fei Fan
- National Institute of Diagnostics and Vaccine Development in Infectious Disease, School of Life Sciences, Xiamen University, Xiamen, 361005, China
| | - Lu Xin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361005, China
| | - Minxi Wei
- National Institute of Diagnostics and Vaccine Development in Infectious Disease, School of Life Sciences, Xiamen University, Xiamen, 361005, China
| | - Zhibo Kong
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361005, China
| | - Hai Yu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361005, China
| | - Ying Gu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361005, China
- National Institute of Diagnostics and Vaccine Development in Infectious Disease, School of Life Sciences, Xiamen University, Xiamen, 361005, China
| | - Jun Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361005, China
| | - Shaowei Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361005, China
- National Institute of Diagnostics and Vaccine Development in Infectious Disease, School of Life Sciences, Xiamen University, Xiamen, 361005, China
- * E-mail: (SL); (NX)
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361005, China
- National Institute of Diagnostics and Vaccine Development in Infectious Disease, School of Life Sciences, Xiamen University, Xiamen, 361005, China
- * E-mail: (SL); (NX)
| |
Collapse
|
962
|
Butsashvili M, Abzianidze T, Kajaia M, Agladze D, Kldiashvili E, Bednarczyk R, McNutt LA, Kamkamidze G. Seroprevalence and awareness of human papillomavirus infection and cervical cancer screening results among reproductive-aged Georgian women. THE JOURNAL OF FAMILY PLANNING AND REPRODUCTIVE HEALTH CARE 2015; 41:265-71. [PMID: 25900523 DOI: 10.1136/jfprhc-2013-100833] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 03/06/2015] [Indexed: 11/04/2022]
Abstract
INTRODUCTION As is the case in many developing countries, more than half of the new cervical cancer cases in Georgia are late-stage diagnoses, thus reducing the opportunity for effective treatment. A state cancer screening programme was launched in Tbilisi in 2006; 5 years later the programme had expanded to other regions in Georgia. METHODS This study was designed to estimate awareness about human papillomavirus (HPV), cervical cancer screening, the HPV vaccine, and the seroprevalence of HPV infection among reproductive-aged Georgian women. Study participants were recruited from four women's consultation centres in different regions of Georgia. Data were collected through interviewer-administered questionnaires and HPV seroprevalence was assessed for HPV types 6/11/16/18. RESULTS Of the 500 study participants, 52.0% were aware of HPV and 36.4% stated that the main cause of cervical cancer is HPV. Of those aware of HPV, 78% reported attending for cervical cancer screening at least once during their lifetime. Half (50.8%) of all respondents were unaware of the HPV vaccine. Of the women who agreed to be tested for anti-HPV antibodies (n=317), 21.1% were positive. Women reporting no condom use were more likely to have HPV antibodies (prevalence ratio 2.77; 95% confidence interval 1.79-4.27). Awareness of cervical cancer screening was significantly associated with HPV seropositivity. With multivariate analysis, both absence of condom use and lack of knowledge about cervical cancer screening were independently associated with HPV seropositivity. CONCLUSION More comprehensive public awareness campaigns should be developed to raise awareness about HPV screening and prevention.
Collapse
Affiliation(s)
| | | | - Maia Kajaia
- Head of Epidemiologic Department, Health Research Union (HRU), Tbilisi, Georgia
| | - Dodo Agladze
- Laboratory Scientist, Health Research Union (HRU), Tbilisi, Georgia
| | | | - Robert Bednarczyk
- Associate Professor, Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Louise-Anne McNutt
- Associate Professor of Epidemiology and Biostatistics, School of Public Health, University at Albany, State University of New York, Albany, NY, USA
| | - George Kamkamidze
- Head of Laboratory Department, Health Research Union (HRU), Tbilisi, Georgia
| |
Collapse
|
963
|
Prediction of high-risk types of human papillomaviruses using statistical model of protein "sequence space". COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2015; 2015:756345. [PMID: 25972913 PMCID: PMC4418008 DOI: 10.1155/2015/756345] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 03/31/2015] [Indexed: 11/29/2022]
Abstract
Discrimination of high-risk types of human papillomaviruses plays an important role in the diagnosis and remedy of cervical cancer. Recently, several computational methods have been proposed based on protein sequence-based and structure-based information, but the information of their related proteins has not been used until now. In this paper, we proposed using protein “sequence space” to explore this information and used it to predict high-risk types of HPVs. The proposed method was tested on 68 samples with known HPV types and 4 samples without HPV types and further compared with the available approaches. The results show that the proposed method achieved the best performance among all the evaluated methods with accuracy 95.59% and F1-score 90.91%, which indicates that protein “sequence space” could potentially be used to improve prediction of high-risk types of HPVs.
Collapse
|
964
|
Clowry J, Callanan I, Eustace K, Collins P, Kirby B, Lally A. Cervical screening uptake in a dermatology cohort on immunosuppressive agents. Br J Dermatol 2015; 172:1682-1683. [DOI: 10.1111/bjd.13609] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- J. Clowry
- Department of Dermatology; St Vincent's University Hospital; Elm Park Dublin 4 Ireland
| | - I. Callanan
- Clinical Audit Department; St Vincent's University Hospital; Elm Park Dublin 4 Ireland
| | - K. Eustace
- Department of Dermatology; St Vincent's University Hospital; Elm Park Dublin 4 Ireland
| | - P. Collins
- Department of Dermatology; St Vincent's University Hospital; Elm Park Dublin 4 Ireland
| | - B. Kirby
- Department of Dermatology; St Vincent's University Hospital; Elm Park Dublin 4 Ireland
| | - A. Lally
- Department of Dermatology; St Vincent's University Hospital; Elm Park Dublin 4 Ireland
| |
Collapse
|
965
|
Nowakowski A, Cybulski M, Śliwczyński A, Chil A, Teter Z, Seroczyński P, Arbyn M, Anttila A. The implementation of an organised cervical screening programme in Poland: an analysis of the adherence to European guidelines. BMC Cancer 2015; 15:279. [PMID: 25879466 PMCID: PMC4417537 DOI: 10.1186/s12885-015-1242-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 03/21/2015] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Well-organised quality-controlled screening can substantially reduce the burden of cervical cancer (CC). European guidelines (EuG) for quality assurance in CC screening provide guidance on all aspects of an organised screening programme. Organised CC screening in Poland was introduced in 2007. The purpose of our study was to analyse: (i) adherence of the programme to EuG; (ii) programme process and performance indicators; (iii) impact of the programme on the incidence of and mortality from CC. METHODS Available data on the policy, structure and functioning of the Polish programme were compared with the major points of the EuG. Data on the process, and available performance indicators were drawn from the screening database and other National Health Fund (NHF) systems. Joinpoint regression was used to assess changes in CC incidence and mortality trends. RESULTS The Polish programme adheres partially to EuG in terms of policy and organisation. Only a limited set of performance indicators can be calculated due to screening database incompleteness or lack of linkage between existing databases. The screening database does not include opportunistic smears collected within NHF-reimbursed or private care. The organised programme coverage rate fluctuated from 21% to 27% between 2007-2013. In 2012 the coverage reached 35% after combining both organised and opportunistic smears reimbursed by the NHF. In 2012 the number of smears reimbursed by NHF was 60% higher in opportunistic than in organised screening with significant overlap. Data from the private sector are not recorded. Depending on years, 30-50% of women referred for colposcopy/biopsy because of abnormal Pap smears were managed within the programme. The age-standardised CC incidence and mortality dropped linearly between 1999 and 2011 without evidence of a period effect. CONCLUSIONS The Polish organised cervical screening programme is only partially adherent to evidence-based EuG. Its implementation has not influenced the burden of CC in the country so far. Changes with special focus on increasing coverage, development of information systems and assessment of quality are required to increase programme adherence to EuG and to measure its effectiveness. Our findings may be useful to improve the Polish programme and those implemented or planned in other countries.
Collapse
Affiliation(s)
- Andrzej Nowakowski
- Department of Gynaecology and Oncologic Gynaecology, Military Institute of Medicine, ul. Szaserów 128, 04-141, Warsaw 44, Poland.
| | - Marek Cybulski
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, ul. Chodźki 1, 20-093, Lublin, Poland.
| | - Andrzej Śliwczyński
- National Health Fund, Central Office, ul. Grójecka 186, 02-390, Warsaw, Poland.
| | - Arkadiusz Chil
- Department of Oncologic Gynaecology, Regional Coordinating Office for Cervical and Breast Cancer Prevention Programmes, Świętokrzyskie Cancer Centre, ul. Artwińskiego 3, 25-734, Kielce, Poland.
| | - Zbigniew Teter
- National Health Fund, Central Office, ul. Grójecka 186, 02-390, Warsaw, Poland.
| | | | - Marc Arbyn
- Unit of Cancer Epidemiology & Belgian Cancer Centre, Scientific Institute of Public Health, Juliette Wytsman Street, 14, B1050, Brussels, Belgium.
| | - Ahti Anttila
- Finnish Cancer Registry, Unioninkatu 22, FI-00130, Helsinki, Finland.
| |
Collapse
|
966
|
Abstract
Viruses are considered intracellular obligates with a nucleic acid, either RNA or DNA. They have the ability to encode proteins involved in viral replication and production of the protective coat within the host cells but require host cell ribosomes and mitochondria for translation. The members of the families Herpesviridae, Poxviridae, Papovaviridae, and Picornaviridae are the most commonly known agents for the cutaneous viral diseases, but other virus families, such as Adenoviridae, Togaviridae, Parvoviridae, Paramyxoviridae, Flaviviridae, and Hepadnaviridae, can also infect the skin. Though the cutaneous manifestations of viral infections are closely related to the type and the transmission route of the virus, viral skin diseases may occur in almost any part of the body. In addition to friction caused by skin-to-skin touch, skin folds are warm and moist areas of the skin that have limited air circulation. These features provide a fertile breeding ground for many kinds of microorganisms, including bacteria and fungi. In contrast to specific bacterial and fungal agents that have an affinity for the skin folds, except for viral diseases of the anogenital area, which have well-known presentations, viral skin infections that have a special affinity to the skin folds are not known. Many viral exanthems may affect the skin folds during the course of the infection, but here we focus only on the ones that usually affect the fold areas and also on the less well-known conditions or recently described associations.
Collapse
|
967
|
Safety and Efficacy Data on Vaccines and Immunization to Human Papillomavirus. J Clin Med 2015; 4:614-33. [PMID: 26239350 PMCID: PMC4470159 DOI: 10.3390/jcm4040614] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 02/09/2015] [Accepted: 02/17/2015] [Indexed: 12/21/2022] Open
Abstract
Since the discovery of the causal association between human papillomavirus (HPV) and cervical cancer, efforts to develop an effective prophylactic vaccine to prevent high-risk HPV infections have been at the forefront of modern medical research. HPV causes 530,000 cervical cancer cases worldwide, which is the second most common cause of cancer deaths in women; a worldwide collaboration among epidemiologists, molecular biologists, vaccinologists, virologists, and clinicians helped lead to the development of two highly effective prophylactive HPV vaccines. The first, Gardasil, is a quadrivalent vaccine made up of recombinant HPV L1 capsid proteins from the two high-risk HPV types (16/18) responsible for 70% of cervical cancer cases as well as two low-risk HPV types (6/11) which are the causative agent for genital warts. The second, Cervarix, is a bivalent vaccine that was FDA approved three years after Gardasil and is also composed of L1 capsid proteins from HPV types 16/18. This review article focuses on the safety and efficacy data of both FDA-approved vaccines, as well as highlighting a few advances in future HPV vaccines that show promise in becoming additional treatment options for this worldwide disease.
Collapse
|
968
|
Artificial Recruitment of UAF1-USP Complexes by a PHLPP1-E1 Chimeric Helicase Enhances Human Papillomavirus DNA Replication. J Virol 2015; 89:6227-39. [PMID: 25833051 DOI: 10.1128/jvi.00560-15] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 03/18/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED The E1 helicase from anogenital human papillomavirus (HPV) types interacts with the cellular WD repeat-containing protein UAF1 in complex with the deubiquitinating enzyme USP1, USP12, or USP46. This interaction stimulates viral DNA replication and is required for maintenance of the viral episome in keratinocytes. E1 associates with UAF1 through a short UAF1-binding site (UBS) located within the N-terminal 40 residues of the protein. Here, we investigated if the E1 UBS could be replaced by the analogous domain from an unrelated protein, the pleckstrin homology domain and leucine-rich repeat protein phosphatase 1 (PHLPP1). We found that PHLPP1 and E1 interact with UAF1 in a mutually exclusive manner and mapped the minimal PHLPP1 UBS (PUBS) to a 100-amino-acid region sufficient for assembly into UAF1-USP complexes. Similarly to the E1 UBS, overexpression of PUBS in trans inhibited HPV DNA replication, albeit less efficiently. Characterization of a PHLPP1-E1 chimeric helicase revealed that PUBS could partially substitute for the E1 UBS in enhancing viral DNA replication and that the stimulatory effect of PUBS likely involves recruitment of UAF1-USP complexes, as it was abolished by mutations that weaken UAF1-binding and by overexpression of catalytically inactive USPs. Although functionally similar to the E1 UBS, PUBS is larger in size and requires both the WD repeat region and C-terminal ubiquitin-like domain of UAF1 for interaction, in contrast to E1, which does not contact the latter. Overall, this comparison of two heterologous UBSs indicates that these domains function as transferable protein interaction modules and provide further evidence that the association of E1 with UAF1-containing deubiquitinating complexes stimulates HPV DNA replication. IMPORTANCE The E1 protein from anogenital HPV types interacts with the UAF1-associated deubiquitinating enzymes USP1, USP12, and USP46 to stimulate replication of the viral genome. Little is known about the molecular nature of the E1-UAF1 interaction and, more generally, how UAF1-USP complexes recognize their substrate proteins. To address this question, we characterized the UAF1-binding site (UBS) of PHLPP1, a protein unrelated to E1. Using a PHLPP1-E1 chimeric helicase, we show that the PHLPP1 UBS (PUBS) can partially substitute for the E1 UBS in stimulating HPV DNA replication. This stimulation required conserved sequences in PUBS that meditate its interaction with UAF1, including a motif common to the E1 UBS. These results indicate that UAF1-binding sequences function as transferable protein interaction modules and provide further evidence that UAF1-USP complexes stimulate HPV DNA replication.
Collapse
|
969
|
Slåttelid Schreiber SM, Juul KE, Dehlendorff C, Kjær SK. Socioeconomic predictors of human papillomavirus vaccination among girls in the Danish childhood immunization program. J Adolesc Health 2015; 56:402-7. [PMID: 25659994 DOI: 10.1016/j.jadohealth.2014.12.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 12/11/2014] [Accepted: 12/11/2014] [Indexed: 12/01/2022]
Abstract
PURPOSE In 2009, human papillomavirus (HPV) vaccination was introduced in the Danish national childhood immunization program targeting all 12-year-old girls. Previous findings suggest that 10%-13% of girls born in 1996-1997 have not initiated vaccination despite free access. This study aims to identify socioeconomic predictors of initiation and completion of HPV vaccination. METHODS Girls born in 1996-1997 and their guardians were identified through the Danish Civil Registration System. Information on socioeconomic variables and HPV vaccination status was obtained by linkage to Statistics Denmark and the Danish National Health Insurance Service Register. Through logistic regression, we examined associations between socioeconomic variables and HPV vaccine initiation (N = 65,926) and completion (N = 61,162). RESULTS Girls with immigrant ethnicity (odds ratio [OR] = .49; 95% confidence interval [CI], .42-.57) had lower HPV vaccine initiation than Danish girls. Girls of mothers with basic education (OR = .75; 95% CI, .69-.82) or low disposable income (OR = .67; 95% CI, .61-.73) had decreased initiation compared with girls of mothers with higher education/income. Girls of unemployed mothers (OR = .75; 95% CI, .69-.82) or mothers being unmarried (OR = .70; 95% CI, .65-.76) had lower initiation than girls of employed or married mothers. Finally, vaccine initiation varied depending on place of residence. The predictors of HPV vaccine completion were similar to those of initiation. CONCLUSIONS We found social inequality in the initiation and completion of HPV vaccination despite free access. As socioeconomic risk factors identified for cervical cancer also are associated with decreased HPV vaccination, social inequalities in cervical cancer have the potential to increase.
Collapse
Affiliation(s)
| | - Kirsten Egebjerg Juul
- Unit of Virus, Lifestyle, and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Christian Dehlendorff
- Unit of Statistics, Bioinformatics, and Registries, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Susanne Krüger Kjær
- Unit of Virus, Lifestyle, and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark; Department of Gynecology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.
| |
Collapse
|
970
|
Bown E, Shah V, Sridhar T, Boyle K, Hemingway D, Yeung JM. Cancers of the anal canal: diagnosis, treatment and future strategies. Future Oncol 2015; 10:1427-41. [PMID: 25052753 DOI: 10.2217/fon.14.23] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Anal cancer is an uncommon cancer; however, it is rising in incidence. There is confusion regarding nomenclature and the distinction between anal canal cancer and anal margin cancer. This article discusses the modern definition, etiology and staging of anal canal and anal margin cancers. Modern chemotherapy and radiotherapy regimens are discussed, in addition to modern imaging and radiotherapy techniques. Future preventative strategies and potential novel treatments are discussed.
Collapse
Affiliation(s)
- Emma Bown
- Department of Colorectal Surgery, Leicester Royal Infirmary, Infirmary Square, Leicester, LE1 5WW, UK
| | | | | | | | | | | |
Collapse
|
971
|
Cambou MC, Luz PM, Lake JE, Levi JE, Coutinho JR, de Andrade A, Heinke T, Derrico M, Veloso VG, Friedman RK, Grinsztejn B. Anal human papillomavirus (HPV) prevalences and factors associated with abnormal anal cytology in HIV-infected women in an urban cohort from Rio de Janeiro, Brazil. AIDS Patient Care STDS 2015; 29:4-12. [PMID: 25361401 DOI: 10.1089/apc.2014.0166] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Identifying factors, including human papillomavirus (HPV) genotypes, associated with abnormal anal cytology in HIV-infected women have implications for anal squamous cell cancer (SCC) prevention in HIV-infected women. Anal and cervical samples were collected for cytology, and tested for high-(HR-HPV) and low-risk HPV (LR-HPV) genotypes in a cross-sectional analysis of the IPEC Women's HIV Cohort (Rio de Janeiro, Brazil). Multivariate log-binomial regression models estimated prevalence ratios for factors associated with abnormal anal cytology [≥ atypical squamous cells of undetermined significance, (ASC-US)]. Characteristics of the 863 participants included: median age 42 years, 57% non-white, 79% current CD4+ T-cell count >350 cells/mm(3), 53% HIV-1 viral load <50 copies/mL, median ART duration 5.8 years. Fifty-one percent of anal specimens contained ≥ 1 HR-HPV genotype; 31% had abnormal anal cytology [14% ASC-US, 11% low-grade squamous intra-epithelial lesion, (LSIL); 2% atypical squamous cells-cannot exclude high-grade SIL (ASC-H); 4% high-grade SIL/cancer (HSIL+)]. In multivariate analysis, cervical LSIL+, nadir CD4+ T-cell count ≤ 50 cells/mm(3), HIV-1 viral load ≥ 50 copies/mL, and anal HPV 6, 11, 16, 18, 33, 45, 52, 56, and 58 were associated with ≥ anal ASC-US (p<0.05). Abnormal anal cytology and HR-HPV prevalences were high. HIV-infected women with cervical LSIL+, low nadir CD4+ counts, or detectable HIV-1 viral loads should be a particular focus for enhanced anal SCC screening efforts.
Collapse
Affiliation(s)
- Mary C. Cambou
- Division of Infectious Diseases and Program in Global Health, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Paula M. Luz
- Oswaldo Cruz Foundation, Evandro Chagas Clinical Research Institute, Rio de Janeiro, Brazil
| | - Jordan E. Lake
- Division of Infectious Diseases and Program in Global Health, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - José Eduardo Levi
- Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo, Brazil
| | - José Ricardo Coutinho
- Oswaldo Cruz Foundation, Evandro Chagas Clinical Research Institute, Rio de Janeiro, Brazil
| | - Angela de Andrade
- Oswaldo Cruz Foundation, Evandro Chagas Clinical Research Institute, Rio de Janeiro, Brazil
| | | | - Mônica Derrico
- Oswaldo Cruz Foundation, Evandro Chagas Clinical Research Institute, Rio de Janeiro, Brazil
| | - Valdilea G. Veloso
- Oswaldo Cruz Foundation, Evandro Chagas Clinical Research Institute, Rio de Janeiro, Brazil
| | - Ruth K. Friedman
- Oswaldo Cruz Foundation, Evandro Chagas Clinical Research Institute, Rio de Janeiro, Brazil
| | - Beatriz Grinsztejn
- Oswaldo Cruz Foundation, Evandro Chagas Clinical Research Institute, Rio de Janeiro, Brazil
| |
Collapse
|
972
|
Rusan M, Li YY, Hammerman PS. Genomic landscape of human papillomavirus-associated cancers. Clin Cancer Res 2015; 21:2009-19. [PMID: 25779941 DOI: 10.1158/1078-0432.ccr-14-1101] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 01/28/2015] [Indexed: 11/16/2022]
Abstract
Recent next-generation sequencing studies have generated a comprehensive overview of the genomic landscape of human papillomavirus (HPV)-associated cancers. This review summarizes these findings to provide insight into the tumor biology of these cancers and potential therapeutic opportunities for HPV-driven malignancies. In addition to the tumorigenic properties of the HPV oncoproteins, integration of HPV DNA into the host genome is suggested to be a driver of the neoplastic process. Integration may confer a growth and survival advantage via enhanced expression of viral oncoproteins, alteration of critical cellular genes, and changes in global promoter methylation and transcription. Alteration of cellular genes may lead to loss of function of tumor suppressor genes, enhanced oncogene expression, loss of function of DNA repair genes, or other vital cellular functions. Recurrent integrations in RAD51B, NR4A2, and TP63, leading to aberrant forms of these proteins, are observed in both HPV-positive head and neck squamous cell carcinoma (HNSCC) and cervical carcinoma. Additional genomic alterations, independent of integration events, include recurrent PIK3CA mutations (and aberrations in other members of the PI3K pathway), alterations in receptor tyrosine kinases (primarily FGFR2 and FGFR3 in HPV-positive HNSCC, and ERBB2 in cervical squamous cell carcinoma), and genes in pathways related to squamous cell differentiation and immune responses. A number of the alterations identified are potentially targetable, which may lead to advances in the treatment of HPV-associated cancers.
Collapse
Affiliation(s)
- Maria Rusan
- Department of Clinical Medicine, Aarhus University, Denmark. Department of Otorhinolaryngology, Aarhus University, Denmark. Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Cancer Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Yvonne Y Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Cancer Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Peter S Hammerman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Cancer Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts.
| |
Collapse
|
973
|
A link between cold environment and cancer. Tumour Biol 2015; 36:5953-64. [PMID: 25736923 DOI: 10.1007/s13277-015-3270-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 02/17/2015] [Indexed: 12/22/2022] Open
Abstract
Many risk factors such as smoking and change of life style have been shown to promote genetic and adaptive epigenetic changes responsible for tumorigenesis. This study brings environmental temperature as a cancer causing factor to light. The cancer mortality rate (CMR) of a country was correlated with 17 different variables. Multivariate analysis of a total of 188 countries found that the average annual temperature (AAT) of a country might have a significant contribution to cancer death when compared with other factors such as alcohol and meat consumption. Univariate analysis found a negative correlation between AAT and CMR. All these countries were categorized into three temperature zones (zone I, -2 to 11.5 °C; number of countries, 38; zone II, 11.6 to 18.6 °C; number of countries, 32; and zone III, 18.7 to 30 °C; number of countries, 118). Out of the top-most 50 countries having the highest CMR, 26 (68.42 %), 10 (31.25 %), and 14 (11.66 %) belong to zone I, zone II, and zone III, respectively. Out of the least 50 countries having the lowest CMR, 1 (2.63 %), 4 (12.5 %), and 45 (37.5 %) belong to zone I, zone II, and zone III, respectively. CMR is low in those countries situated near to the Torrid zone (33(°) N to 23.5(°)S), but it is high for those countries situated away from these two latitudes. These data indicate that cold temperature may have a contribution in increasing tumorigenesis. High metabolic stress, which is the result of maintaining our body temperature against a cold environment, could be the possible cause for the higher cancer mortality.
Collapse
|
974
|
Design of a large outcome trial for a multivalent human papillomavirus L1 virus-like particle vaccine. Contemp Clin Trials 2015; 42:18-25. [PMID: 25749310 DOI: 10.1016/j.cct.2015.02.009] [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] [Received: 12/11/2014] [Revised: 02/23/2015] [Accepted: 02/24/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND The 9-valent human papillomavirus (HPV) (9vHPV) vaccine targets the four HPV types (6/11/16/18) covered by the licensed quadrivalent HPV (qHPV) vaccine and five additional types (31/33/45/52/58). A large outcome trial of 9vHPV vaccine was conducted. METHODS An active control (qHPV vaccine) was used because a placebo is not ethically acceptable. Since qHPV vaccine is (and 9vHPV vaccine was anticipated to be) highly efficacious against HPV 6/11/16/18, low incidence of HPV 6/11/16/18-associated disease was expected. Consequently, an efficacy comparison of 9vHPV versus qHPV vaccine for HPV 6/11/16/18 would have been prohibitively large in size. Moreover, no minimum antibody level predicting protection against infection or disease is defined for HPV vaccination. As an alternative approach, the two vaccines were compared using immunogenicity bridging for HPV 6/11/16/18 and clinical efficacy for HPV 31/33/45/52/58. RESULTS The two co-primary objectives were to demonstrate: (1) non-inferior anti-HPV 6/11/16/18 antibody response; and (2) superior efficacy in HPV 31/33/45/52/58-related clinical outcome, for 9vHPV vaccine versus qHPV vaccine. For HPV 6/11/16/18, supportive analyses included a non-inferiority assessment of the percent risk reduction (compared to historical placebo) for 9vHPV versus qHPV vaccine. CONCLUSIONS A Phase III study of 9vHPV vaccine was successfully implemented. Experience from this study design may be applicable when developing a multivalent vaccine covering the same serotypes as an existing vaccine plus additional serotypes and there is no immune correlate of protection. Also, this study established that efficacy of a new HPV vaccine may be demonstrated using immunogenicity endpoints, which may open new options in HPV vaccine development.
Collapse
|
975
|
Meeuwis KAP, Hilbrands LB, IntHout J, Slangen BFM, Hendriks IMP, Hinten F, Christiaans MHL, Quint WGV, van de Kerkhof PCM, Massuger LFAG, Hoitsma AJ, van Rossum MM, Melchers WJG, de Hullu JA. Cervicovaginal HPV infection in female renal transplant recipients: an observational, self-sampling based, cohort study. Am J Transplant 2015; 15:723-33. [PMID: 25675976 DOI: 10.1111/ajt.13053] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 09/21/2014] [Accepted: 10/04/2014] [Indexed: 01/25/2023]
Abstract
Immunosuppressive treatment of organ transplant recipients is associated with an increase in the occurrence of human papillomavirus (HPV) related anogenital (pre)malignancies. This cohort study investigated the genotype-specific prevalence of HPV infections in a large cohort of female renal transplant recipients (RTRs). Participants self-collected a cervicovaginal sample for detection and genotyping of HPV. Besides, they completed a questionnaire regarding sociodemographic variables, medical data and sexual behavior. Anogenital screening was offered to all HPV-positive participants. A total number of 218 female RTRs was included. The prevalence of mucosal HPV infections was 27.1% and 17.4% for high risk HPV in particular. The studied cohort showed a broad range of HPV genotypes and multiple HPV genotypes were found in 27.1% of HPV-positive patients. Seven participants were identified with occult premalignant anogenital lesions. In conclusion, this study shows a high point-prevalence of HPV in female RTRs (age-matched West-European general population: 9-10%) with a shift in the distribution of genotypes as compared with the general population. Moreover, a substantial number of patients with occult premalignancies was identified. The introduction of self-sampling for HPV positivity can help in early detection of (pre)malignant anogenital lesions in this vulnerable population.
Collapse
Affiliation(s)
- K A P Meeuwis
- Department of Obstetrics and Gynaecology, Radboud university medical center, Nijmegen, The Netherlands; Department of Dermatology, Radboud university medical center, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
976
|
Abstract
Cancer constitutes an enormous burden on society in more and less economically developed countries alike. The occurrence of cancer is increasing because of the growth and aging of the population, as well as an increasing prevalence of established risk factors such as smoking, overweight, physical inactivity, and changing reproductive patterns associated with urbanization and economic development. Based on GLOBOCAN estimates, about 14.1 million new cancer cases and 8.2 million deaths occurred in 2012 worldwide. Over the years, the burden has shifted to less developed countries, which currently account for about 57% of cases and 65% of cancer deaths worldwide. Lung cancer is the leading cause of cancer death among males in both more and less developed countries, and has surpassed breast cancer as the leading cause of cancer death among females in more developed countries; breast cancer remains the leading cause of cancer death among females in less developed countries. Other leading causes of cancer death in more developed countries include colorectal cancer among males and females and prostate cancer among males. In less developed countries, liver and stomach cancer among males and cervical cancer among females are also leading causes of cancer death. Although incidence rates for all cancers combined are nearly twice as high in more developed than in less developed countries in both males and females, mortality rates are only 8% to 15% higher in more developed countries. This disparity reflects regional differences in the mix of cancers, which is affected by risk factors and detection practices, and/or the availability of treatment. Risk factors associated with the leading causes of cancer death include tobacco use (lung, colorectal, stomach, and liver cancer), overweight/obesity and physical inactivity (breast and colorectal cancer), and infection (liver, stomach, and cervical cancer). A substantial portion of cancer cases and deaths could be prevented by broadly applying effective prevention measures, such as tobacco control, vaccination, and the use of early detection tests.
Collapse
Affiliation(s)
- Lindsey A Torre
- Epidemiologist, Surveillance and Health Services Research, American Cancer Society, Atlanta, GA
| | | | | | | | | | | |
Collapse
|
977
|
Georgantis G, Syrakos T, Agorastos T, Miliaras S, Gagalis A, Tsoulfas G, Spanos K, Marakis G. Detection of human papillomavirus DNA in esophageal carcinoma in Greece. World J Gastroenterol 2015; 21:2352-2357. [PMID: 25741141 PMCID: PMC4342910 DOI: 10.3748/wjg.v21.i8.2352] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 09/15/2014] [Accepted: 11/19/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To detect human papillomavirus (HPV) in the esophageal mucosa and the possible relationship with esophageal cancer in Greece.
METHODS: Forty-nine patients underwent esophagogastroduodenoscopy (EGD) and esophageal biopsy at a university hospital that acts as a referral center for Northern Greece. Nineteen of these patients (14 male and 5 female) had esophageal squamous cell carcinoma (ESCC) and 30 (15 male and 15 female) did not have any reported esophageal malignancy. Histopathological assessment was followed by polymerase chain reaction analysis of all the samples. Patient demographic data (age, sex, and place of birth) and information regarding smoking habits, alcohol consumption or sexual habits were collected. A method of statistical interference, verification of hypotheses based on homogeneity and independent χ2 test, was used.
RESULTS: From the 49 patients that underwent EGD and biopsy, 19 had ESCC and 30 had normal esophageal mucosa, with a mean age of 65.2 years. Regarding the prevalence of oncogenic risk factors for esophageal carcinoma, an interesting conclusion was that 78% of the patients used tobacco and almost one-third had multiple sexual partners, whereas only 20% of the patients consumed alcohol, which was not statistically significant, when compared to the control group. In the ESCC group, the only two positive samples were among the male patients (2/14 male patients with ESCC, 14.5%). No HPV was identified in the control group. The predominant HPV types identified were 11 and 31, which have a low malignancy potential. The presence of HPV DNA in the ESCC group was not statistically significant, 95% confidence interval (χ2 = 3.292, P = 0.07).
CONCLUSION: This is the first relevant study in Greece, and despite the lack of statistical significance, the issue of HPV infection and ESCC does merit further investigation.
Collapse
|
978
|
Ghosn M, Kourie HR, Abdayem P, Antoun J, Nasr D. Anal cancer treatment: Current status and future perspectives. World J Gastroenterol 2015; 21:2294-2302. [PMID: 25741135 PMCID: PMC4342904 DOI: 10.3748/wjg.v21.i8.2294] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 10/24/2014] [Accepted: 12/22/2014] [Indexed: 02/07/2023] Open
Abstract
Anal cancers (AC) are relatively rare tumors. Their incidence is increasing, particularly among men who have sex with other men due to widespread infection by human papilloma virus. The majority of anal cancers are squamous cell carcinomas, and they are treated according to stage. In local and locally advanced AC, concomitant chemoradiation therapy based on mitomycin C and 5-Fluorouracil (5-FU) is the current best treatment, while metastatic AC, chemotherapy with 5-FU and cisplatin remains the gold standard. There are no indications for induction or maintenance therapies in locally advanced tumors. Many novel strategies, such as targeted therapies, vaccination, immunotherapy and photodynamic therapy are in clinical trials for the treatment of AC, with promising results in some indications.
Collapse
|
979
|
McClure CA, MacSwain MA, Morrison H, Sanford CJ. Human papillomavirus vaccine uptake in boys and girls in a school-based vaccine delivery program in Prince Edward Island, Canada. Vaccine 2015; 33:1786-90. [PMID: 25731789 DOI: 10.1016/j.vaccine.2015.02.047] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 02/10/2015] [Accepted: 02/17/2015] [Indexed: 01/11/2023]
Abstract
BACKGROUND In 2013, Prince Edward Island was the first province to introduce HPV vaccine universally to grade six boys in a school-based program. Because uptake rates in boys are unknown in this type of vaccination program, uptake of HPV vaccination in boys was measured and compared with uptake rates in girls and then analyzed with factors such as county, urban-rural location of the school, and school board to identify where the vaccine program could be improved. METHODS HPV vaccination records from the provincial childhood immunization registry in PEI were merged with Department of Education data containing all grade six girls and boys in PEI. Vaccine uptakes between years and between sexes were compared using two sample tests of proportions. Logistic regression modeling which accounted for the hierarchical nature of the data was used to analyze associations between factors and uptake rates. RESULTS Although uptake was high in boys and girls, a significantly greater proportion of girls (85%) received all three doses of the HPV vaccine compared to boys (79%; p=0.004). The odds of grade six girls being fully vaccinated for HPV were 1.5 times greater than of grade six boys, and the odds of students in the English Language School Board receiving all three doses were more than twice as great as the odds of French Language School Board students. CONCLUSIONS HPV vaccination for boys in PEI has had a successful launch, almost reaching the Canadian Immunization Committee recommendations of >80% for the early years of a program. PEI has a highly organized Public Health Nursing program that is involved in all childhood and school-based vaccinations in PEI and in this context very high coverage rates were obtained. Areas to target for improving uptake include the boys and the students in the French Language School Board.
Collapse
Affiliation(s)
- Carol A McClure
- Chief Public Health Office, Department of Health and Wellness, Government of Prince Edward Island, 16 Fitzroy Street, 2nd Floor Sullivan Building, Charlottetown, PE, Canada C1A 7N8.
| | - Mary-Ann MacSwain
- Chief Public Health Office, Department of Health and Wellness, Government of Prince Edward Island, 16 Fitzroy Street, 2nd Floor Sullivan Building, Charlottetown, PE, Canada C1A 7N8.
| | - Heather Morrison
- Chief Public Health Office, Department of Health and Wellness, Government of Prince Edward Island, 16 Fitzroy Street, 2nd Floor Sullivan Building, Charlottetown, PE, Canada C1A 7N8
| | - Carolyn J Sanford
- Chief Public Health Office, Department of Health and Wellness, Government of Prince Edward Island, 16 Fitzroy Street, 2nd Floor Sullivan Building, Charlottetown, PE, Canada C1A 7N8.
| |
Collapse
|
980
|
Aguilar-Lemarroy A, Vallejo-Ruiz V, Cortés-Gutiérrez EI, Salgado-Bernabé ME, Ramos-González NP, Ortega-Cervantes L, Arias-Flores R, Medina-Díaz IM, Hernández-Garza F, Santos-López G, Piña-Sánchez P. Human papillomavirus infections in Mexican women with normal cytology, precancerous lesions, and cervical cancer: Type-specific prevalence and HPV coinfections. J Med Virol 2015; 87:871-84. [DOI: 10.1002/jmv.24099] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2014] [Indexed: 11/12/2022]
Affiliation(s)
| | | | | | | | | | | | - Rafael Arias-Flores
- Molecular Oncology Laboratory; Oncology Research Unit (UIMEO)-IMSS; Mexico City Mexico
| | | | | | | | - Patricia Piña-Sánchez
- Molecular Oncology Laboratory; Oncology Research Unit (UIMEO)-IMSS; Mexico City Mexico
| | | |
Collapse
|
981
|
Woodham AW, Taylor JR, Jimenez AI, Skeate JG, Schmidt T, Brand HE, Da Silva DM, Kast WM. Small molecule inhibitors of the annexin A2 heterotetramer prevent human papillomavirus type 16 infection. J Antimicrob Chemother 2015; 70:1686-90. [PMID: 25712315 DOI: 10.1093/jac/dkv045] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 02/03/2015] [Indexed: 01/29/2023] Open
Abstract
OBJECTIVES High-risk human papillomavirus (HPV) infection leads to the development of several human cancers that cause significant morbidity and mortality worldwide. HPV type 16 (HPV16) is the most common of the cancer-causing genotypes and gains entry to the basal cells of the epithelium through a non-canonical endocytic pathway that involves the annexin A2/S100A10 heterotetramer (A2t). A2t is composed of two annexin A2 monomers bound to an S100A10 dimer and this interaction is a potential target to block HPV16 infection. Here, recently identified small molecule inhibitors of A2t (A2ti) were investigated for their ability to prevent HPV16 infection in vitro. METHODS A2ti were added to HeLa cells in increasing concentrations prior to the addition of HPV16. Cytotoxicity was evaluated via trypan blue exclusion. HPV16 pseudovirion infection and fluorescently labelled HPV16 capsid internalization was measured with flow cytometry. RESULTS A2ti blocked HPV16 infection by 100% without substantial cellular toxicity or reduction in cell growth. Furthermore, A2ti blocked HPV16 entry into epithelial cells by 65%, indicating that the observed inhibition of HPV16 infection is in part due to a block in entry and that non-infectious entry may occur in the absence of A2t binding. CONCLUSIONS These results demonstrate that targeting A2t may be an effective strategy to prevent HPV16 infection.
Collapse
Affiliation(s)
- Andrew W Woodham
- Department of Molecular Microbiology & Immunology, University of Southern California, 2011 Zonal Avenue HMR 401, Los Angeles, CA, USA
| | - Julia R Taylor
- Department of Molecular Microbiology & Immunology, University of Southern California, 2011 Zonal Avenue HMR 401, Los Angeles, CA, USA
| | - Andrew I Jimenez
- Department of Molecular Microbiology & Immunology, University of Southern California, 2011 Zonal Avenue HMR 401, Los Angeles, CA, USA
| | - Joseph G Skeate
- Norris Comprehensive Cancer Center, University of Southern California, 1441 Eastlake Avenue, Los Angeles, CA, USA
| | - Thomas Schmidt
- Laboratories of Chemical Physics and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Heike E Brand
- Norris Comprehensive Cancer Center, University of Southern California, 1441 Eastlake Avenue, Los Angeles, CA, USA
| | - Diane M Da Silva
- Norris Comprehensive Cancer Center, University of Southern California, 1441 Eastlake Avenue, Los Angeles, CA, USA Department of Obstetrics & Gynecology, University of Southern California, 2020 Zonal Avenue Room 220, Los Angeles, CA, USA
| | - W Martin Kast
- Department of Molecular Microbiology & Immunology, University of Southern California, 2011 Zonal Avenue HMR 401, Los Angeles, CA, USA Norris Comprehensive Cancer Center, University of Southern California, 1441 Eastlake Avenue, Los Angeles, CA, USA Department of Obstetrics & Gynecology, University of Southern California, 2020 Zonal Avenue Room 220, Los Angeles, CA, USA
| |
Collapse
|
982
|
[HPV (Human Papilloma Virus) implication in other cancers than gynaecological]. Rev Med Interne 2015; 36:540-7. [PMID: 25661671 DOI: 10.1016/j.revmed.2015.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 01/07/2015] [Accepted: 01/09/2015] [Indexed: 01/26/2023]
Abstract
Worldwide, approximately 5 to 10% of the population is infected by a Human Papilloma Virus (HPV). Some of these viruses, with a high oncogenic risk (HPV HR), are responsible for about 5% of cancer. It is now accepted that almost all carcinomas of the cervix and the vulva are due to an HPV HR (HPV16 and 18) infection. However, these viruses are known to be involved in the carcinogenesis of many other cancers (head and neck [SCCHN], penis, anus). For head and neck cancer, HPV infection is considered as a good prognostic factor. The role of HPV HR in anal cancer is also extensively studied in high-risk patient's population. The role of HPV infection in the carcinogenesis of esophageal, bladder, lung, breast or skin cancers is still debated. Given the multiple possible locations of HPV HR infection, the question of optimizing the management of patients with a HPV+ cancer arises in the implementation of a comprehensive clinical and biological monitoring. It is the same in therapeutics with the existence of a preventive vaccination, for example.
Collapse
|
983
|
Kreimer AR, Sherman ME, Sahasrabuddhe VV, Safaeian M. The case for conducting a randomized clinical trial to assess the efficacy of a single dose of prophylactic HPV vaccines among adolescents. J Natl Cancer Inst 2015; 107:dju436. [PMID: 25650316 DOI: 10.1093/jnci/dju436] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Aimée R Kreimer
- Division of Cancer Epidemiology and Genetics, Infections and Immunoepidemiology Branch (ARK, MS), Division of Cancer Prevention, Breast and Gynecologic Cancer Research Group (MES, VVS), Division of Cancer Epidemiology and Genetics, Human Genetics Program (MES), and Division of Cancer Epidemiology and Genetics, Hormonal and Reproductive Epidemiology Branch (VVS), National Cancer Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN (VVS).
| | - Mark E Sherman
- Division of Cancer Epidemiology and Genetics, Infections and Immunoepidemiology Branch (ARK, MS), Division of Cancer Prevention, Breast and Gynecologic Cancer Research Group (MES, VVS), Division of Cancer Epidemiology and Genetics, Human Genetics Program (MES), and Division of Cancer Epidemiology and Genetics, Hormonal and Reproductive Epidemiology Branch (VVS), National Cancer Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN (VVS)
| | - Vikrant V Sahasrabuddhe
- Division of Cancer Epidemiology and Genetics, Infections and Immunoepidemiology Branch (ARK, MS), Division of Cancer Prevention, Breast and Gynecologic Cancer Research Group (MES, VVS), Division of Cancer Epidemiology and Genetics, Human Genetics Program (MES), and Division of Cancer Epidemiology and Genetics, Hormonal and Reproductive Epidemiology Branch (VVS), National Cancer Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN (VVS)
| | - Mahboobeh Safaeian
- Division of Cancer Epidemiology and Genetics, Infections and Immunoepidemiology Branch (ARK, MS), Division of Cancer Prevention, Breast and Gynecologic Cancer Research Group (MES, VVS), Division of Cancer Epidemiology and Genetics, Human Genetics Program (MES), and Division of Cancer Epidemiology and Genetics, Hormonal and Reproductive Epidemiology Branch (VVS), National Cancer Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN (VVS)
| |
Collapse
|
984
|
Bravo IG, Félez-Sánchez M. Papillomaviruses: Viral evolution, cancer and evolutionary medicine. EVOLUTION MEDICINE AND PUBLIC HEALTH 2015; 2015:32-51. [PMID: 25634317 PMCID: PMC4356112 DOI: 10.1093/emph/eov003] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Papillomaviruses (PVs) are a numerous family of small dsDNA viruses infecting virtually all mammals. PVs cause infections without triggering a strong immune response, and natural infection provides only limited protection against reinfection. Most PVs are part and parcel of the skin microbiota. In some cases, infections by certain PVs take diverse clinical presentations from highly productive self-limited warts to invasive cancers. We propose PVs as an excellent model system to study the evolutionary interactions between the immune system and pathogens causing chronic infections: genotypically, PVs are very diverse, with hundreds of different genotypes infecting skin and mucosa; phenotypically, they display extremely broad gradients and trade-offs between key phenotypic traits, namely productivity, immunogenicity, prevalence, oncogenicity and clinical presentation. Public health interventions have been launched to decrease the burden of PV-associated cancers, including massive vaccination against the most oncogenic human PVs, as well as systematic screening for PV chronic anogenital infections. Anti-PVs vaccines elicit protection against infection, induce cross-protection against closely related viruses and result in herd immunity. However, our knowledge on the ecological and intrapatient dynamics of PV infections remains fragmentary. We still need to understand how the novel anthropogenic selection pressures posed by vaccination and screening will affect viral circulation and epidemiology. We present here an overview of PV evolution and the connection between PV genotypes and the phenotypic, clinical manifestations of the diseases they cause. This differential link between viral evolution and the gradient cancer-warts-asymptomatic infections makes PVs a privileged playground for evolutionary medicine research.
Collapse
Affiliation(s)
- Ignacio G Bravo
- Infections and Cancer Laboratory, Catalan Institute of Oncology (ICO), Barcelona, Spain; Bellvitge Institute of Biomedical Research (IDIBELL), Barcelona, Spain Infections and Cancer Laboratory, Catalan Institute of Oncology (ICO), Barcelona, Spain; Bellvitge Institute of Biomedical Research (IDIBELL), Barcelona, Spain Infections and Cancer Laboratory, Catalan Institute of Oncology (ICO), Barcelona, Spain; Bellvitge Institute of Biomedical Research (IDIBELL), Barcelona, Spain
| | - Marta Félez-Sánchez
- Infections and Cancer Laboratory, Catalan Institute of Oncology (ICO), Barcelona, Spain; Bellvitge Institute of Biomedical Research (IDIBELL), Barcelona, Spain Infections and Cancer Laboratory, Catalan Institute of Oncology (ICO), Barcelona, Spain; Bellvitge Institute of Biomedical Research (IDIBELL), Barcelona, Spain
| |
Collapse
|
985
|
Groves IJ, Coleman N. Pathogenesis of human papillomavirus-associated mucosal disease. J Pathol 2015; 235:527-38. [PMID: 25604863 DOI: 10.1002/path.4496] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 12/03/2014] [Indexed: 12/15/2022]
Abstract
Human papillomaviruses (HPVs) are a necessary cause of carcinoma of the cervix and other mucosal epithelia. Key events in high-risk HPV (HRHPV)-associated neoplastic progression include persistent infection, deregulated expression of virus early genes in basal epithelial cells and genomic instability causing secondary host genomic imbalances. There are multiple mechanisms by which deregulated virus early gene expression may be achieved. Integration of virus DNA into host chromosomes is observed in the majority of cervical squamous cell carcinomas (SCCs), although in ∼15% of cases the virus remains extrachromosomal (episomal). Interestingly, not all integration events provide a growth advantage to basal cervical epithelial cells or lead to increased levels of the virus oncogenes E6 and E7, when compared with episome-containing basal cells. The factors that provide a competitive advantage to some integrants, but not others, are complex and include virus and host contributions. Gene expression from integrated and episomal HRHPV is regulated through host epigenetic mechanisms affecting the virus long control region (LCR), which appear to be of functional importance. New approaches to treating HRHPV-associated mucosal neoplasia include knockout of integrated HRHPV DNA, depletion of virus transcripts and inhibition of virus early gene transcription through targeting or use of epigenetic modifiers. Copyright © 2014 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Ian J Groves
- University of Cambridge, Department of Pathology, UK
| | | |
Collapse
|
986
|
Dykens A, Hedrick C, Ndiaye Y, Linn A. Peace corps partnered health services implementation research in global health: opportunity for impact. Glob Adv Health Med 2015; 3:8-15. [PMID: 25568819 PMCID: PMC4268604 DOI: 10.7453/gahmj.2014.031] [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] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND There is abundant evidence of the affordable, life-saving interventions effective at the local primary health care level in low- and middle-income countries (LMICs). However, the understanding of how to deliver those interventions in diverse settings is limited. Primary healthcare services implementation research is needed to elucidate the contextual factors that can influence the outcomes of interventions, especially at the local level. US universities commonly collaborate with LMIC universities, communities, and health system partners for health services research but common barriers exist. Current challenges include the capacity to establish an ongoing presence in local settings in order to facilitate close collaboration and communication. The Peace Corps is an established development organization currently aligned with local health services in many LMICs and is well-positioned to facilitate research partnerships. This article explores the potential of a community-Peace Corps-academic partnership approach to conduct local primary healthcare services implementation research. DISCUSSION The Peace Corps is well positioned to offer insights into local contextual factors because volunteers work closely with local leaders, have extensive trust within local communities, and have an ongoing, constant, well-integrated presence. However, the Peace Corps does not routinely conduct primary healthcare services implementation research. Universities, within the United States and locally, could benefit from the established resources and trust of the Peace Corps to conduct health services implementation research to advance access to local health services and further the knowledge of real world application of local health services in a diversity of settings. The proposed partnership would consist of (1) a local community advisory board and local health system leaders, (2) Peace Corps volunteers, and (3) a US-LMIC academic institutional collaboration. Within the proposed partnership approach, the contributions of each partner are as follows: the local community and health system leadership guides the work in consideration of local priorities and context; the Peace Corps provides logistical support, community expertise, and local trust; and the academic institutions offer professional technical and public health educational and training resources and research support. CONCLUSION The Peace Corps offers the opportunity to enhance a community-academic partnership in LMICs through community-level guidance, logistical assistance, and research support for community based participatory primary health-care services implementation research that addresses local primary healthcare priorities.
Collapse
Affiliation(s)
- Andrew Dykens
- University of Illinois at Chicago (Dr Dykens), United States
| | | | | | - Annē Linn
- Peace Corps, Senegal (Dr Linn), Senegal
| |
Collapse
|
987
|
Abstract
The oncogenic, anogenital types of human papillomavirus (HPV) are established as causing about 4.8% of all human cancers worldwide, particularly cervical, anal, vulvar, vaginal, penile, and oropharyngeal cancers. Quantitative knowledge of the HPV type-specific risks for these cancers, as well as for the different cervical cancer precursors (cervical intraepithelial neoplasias, CINs), is useful for estimating the effect of elimination of specific HPV types and clinical benefits of screening for specific HPV types. The present review summarizes both the worldwide presence of specific HPV types in cervical cancer precursors and in invasive cervical cancers, and also the long-term follow-up data from a large randomized clinical trial of HPV-based cervical cancer screening. All 12 HPV types classified as class I (established) carcinogens (HPV types 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59) were more common in cervical cancers than among women without cervical lesions. A few rare HPV types also were more common in cervical cancers (eg, HPV26, 67, 68, 69, 73, 82). The follow-up studies found increased long-term risks particularly for HPV types 16, 18, 31, and 33, which had 14-year cumulative incidences for CIN3+above 28%, while HPV35, 45, 52, and 58 had 14-year risks between 14%-18% and HPV39, 51, 56, 59, 66, and 68 had risks<10%. HPV16 contributed to the greatest proportion of CIN2+(first-round population attributable proportion [PAR] 36%), followed by types 31, 52, 45, and 58 (7%-11%). HPV16, 18, 31, 33, 45, 52, and 58 together contributed 73.9% of CIN2+lesions and all high-risk types contributed 86.9%.In summary, the different oncogenic HPV types have substantial differences in their oncogenic potential. These differences are relevant for the design and evaluation of cervical screening tests and programs, as well as for studying the effect of vaccination programs using different HPV vaccines.
Collapse
Affiliation(s)
- Joakim Dillner
- International HPV Reference Center, Department of Laboratory Medicine, and Department of Medical Epidemiology & Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
988
|
Bonanni P, Bechini A, Donato R, Capei R, Sacco C, Levi M, Boccalini S. Human papilloma virus vaccination: impact and recommendations across the world. THERAPEUTIC ADVANCES IN VACCINES 2015; 3:3-12. [PMID: 25553242 PMCID: PMC4266686 DOI: 10.1177/2051013614557476] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Human papilloma virus (HPV) vaccination has been implemented in several countries for about the past 7 years, mainly in the adolescent female population, with varying coverage results. Although the impact of immunization on cervical and other HPV-related cancers will be evident in the next decades, a marked decrease of prevalent HPV infections, precancerous lesions and genital warts is already dramatic in the vaccinated cohorts, and also in their sexual partners, thus providing clear evidence of the effectiveness of HPV vaccination, including a herd-protection effect. Today, recommendations and implementation of universal HPV vaccination for adolescent girls are a public-health priority in all countries of the world. Countries with limited resources are presently involved in demonstration projects and, in some cases, have launched national programmes with the help of international agencies and alliances. Extension of immunization offer to young women and to adolescent male subjects has become an important additional opportunity for several countries, with a special focus needed on homosexual men with HIV infection who are at particularly increased risk of HPV-related diseases. Public-health authorities are confronted with the need to enlarge HPV-vaccination offer to all target groups, especially pre-adolescent girls, so that they can be saved from dreadful cancers by reaching high immunization coverage.
Collapse
Affiliation(s)
- Paolo Bonanni
- Department of Health Sciences, University of Florence, Viale G.B. Morgagni 48, 50134 Florence, Italy
| | - Angela Bechini
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Rosa Donato
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Raffaella Capei
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Cristiana Sacco
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Miriam Levi
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Sara Boccalini
- Department of Health Sciences, University of Florence, Florence, Italy
| |
Collapse
|
989
|
Kabekkodu SP, Bhat S, Pandey D, Varghese VK, Shukla V, Ghosh S, Kushtagi P, Bhat P, Gopinath PM, Satyamoorthy K. Prevalence of human papillomavirus types and phylogenetic analysis of HPV-16 L1 variants from Southern India. Asian Pac J Cancer Prev 2015; 16:2073-2080. [PMID: 25773853 DOI: 10.7314/apjcp.2015.16.5.2073] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND The human papillomavirus (HPV) and its variants show wide geographical distribution and have been reported to cause cervical lesions. With cervical neoplasia as the leading cancer in Indian women, the aim of the present study was to evaluate the multiple infection HPV type distribution and variant genotypes in cervical samples from the coastal Karnataka region, India. MATERIALS AND METHODS A total of 212 samples were screened by nested polymerase chain reaction using PGMY9/11 and GP5+/6+ primers. HPV positive samples were sequenced to identify the types and a phylogenetic tree was constructed using the neighbor-joining method. RESULTS Sequence analysis identified a total of 14 HPV types distributed in 20%, 73.3% and 82.5% of non-malignant, pre-malignant [low grade squamous intraepithelial lesion (LSIL) and high grade squamous intraepithelial lesion (HSIL)] and cervical cancer samples. The distribution of high risk HPV in cancer samples was HPV 16, 76.4%, HPV18, 11.7%, HPV81, 2.9%, HPV31, 1.4%, HPV35, 1.4% and HPV 45, 1.4%. Multiple infections were observed in 11.8% of tumor samples with HPV 16 contributing to 62.5% of cases. In non-malignant samples, 20% of HPV positive samples were detected with HPV16, 82.3%, HPV33, 5.8% and HPV58, 5.8% and very low incidence of multiple infections. Comparative phylogenetic analysis of HPV variants identified 9 HPV sequences as new papillomavirus species, predominantly classified as European lineage type. CONCLUSIONS The findings for HPV infections associated with progression of cervical cancer in coastal Karnataka region and HPV variant analysis provide baseline data for prevention and HPV vaccination programs.
Collapse
Affiliation(s)
- Shama Prasada Kabekkodu
- Division of Biotechnology, School of Life Sciences, Manipal University, Karnataka, India E-mail :
| | | | | | | | | | | | | | | | | | | |
Collapse
|
990
|
Pack AE, Voskuhl GW. Anal Cancer Prevention in a High-risk Population. J Nurse Pract 2015. [DOI: 10.1016/j.nurpra.2014.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
991
|
Lang Kuhs KA, Gonzalez P, Rodriguez AC, van Doorn LJ, Schiffman M, Struijk L, Chen S, Quint W, Lowy DR, Porras C, DelVecchio C, Jimenez S, Safaeian M, Schiller JT, Wacholder S, Herrero R, Hildesheim A, Kreimer AR. Reduced prevalence of vulvar HPV16/18 infection among women who received the HPV16/18 bivalent vaccine: a nested analysis within the Costa Rica Vaccine Trial. J Infect Dis 2014; 210:1890-9. [PMID: 24958910 PMCID: PMC4271025 DOI: 10.1093/infdis/jiu357] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 06/18/2014] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Vaccine efficacy (VE) against vulvar human papillomavirus (HPV) infection has not been reported and data regarding its epidemiology are sparse. METHODS Women (n = 5404) age 22-29 present at the 4-year study visit of the Costa Rica Vaccine Trial provided vulvar and cervical samples. A subset (n = 1044) was tested for HPV DNA (SPF10/LiPA25 version 1). VE against 1-time detection of vulvar HPV16/18 among HPV vaccinated versus unvaccinated women was calculated and compared to the cervix. Prevalence of and risk factors for HPV were evaluated in the control arm (n = 536). RESULTS Vulvar HPV16/18 VE (54.1%; 95% confidence interval [CI], 4.9%-79.1%) was comparable to cervix (45.8%; 95% CI, 6.4%-69.4%). Vulvar and cervical HPV16 prevalence within the control arm was 3.0% and 4.7%, respectively. Independent risk factors for vulvar HPV were similar to cervix and included: age (adjusted odds ratio [aOR] 0.5 [95% CI, .3-.9] ≥28 vs 22-23]); marital status (aOR 2.3 [95% CI, 1.5-3.5] single vs married/living-as-married); and number of sexual partners (aOR 3.6 [95% CI, 1.9-7.0] ≥6 vs 1). CONCLUSIONS In this intention-to-treat analysis, VE against vulvar and cervical HPV16/18 were comparable 4 years following vaccination. Risk factors for HPV were similar by anatomic site. CLINICAL TRIALS REGISTRATION NCT00128661.
Collapse
Affiliation(s)
| | - Paula Gonzalez
- Proyecto Epidemiológico Guanacaste, Fundación INCIENSA, Costa Rica
- Prevention and Implementation Group, International Agency for Research on Cancer, Lyon, France
| | | | | | | | - Linda Struijk
- DDL Diagnostic Laboratory, Rijswijk, The Netherlands
| | - Sabrina Chen
- Information Management Services, Calverton, Maryland
| | - Wim Quint
- DDL Diagnostic Laboratory, Rijswijk, The Netherlands
| | | | - Carolina Porras
- Proyecto Epidemiológico Guanacaste, Fundación INCIENSA, Costa Rica
| | | | - Silvia Jimenez
- Proyecto Epidemiológico Guanacaste, Fundación INCIENSA, Costa Rica
| | | | | | | | - Rolando Herrero
- Prevention and Implementation Group, International Agency for Research on Cancer, Lyon, France
| | | | | |
Collapse
|
992
|
Wadhera P, Evans JL, Stein E, Gandhi M, Couture MC, Sansothy N, Sichan K, Maher L, Kaldor J, Page K, Kien. Human papillomavirus knowledge, vaccine acceptance, and vaccine series completion among female entertainment and sex workers in Phnom Penh, Cambodia: the Young Women's Health Study. Int J STD AIDS 2014; 26:893-902. [PMID: 25505042 DOI: 10.1177/0956462414563626] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 11/04/2014] [Indexed: 11/17/2022]
Abstract
Human papillomavirus is a common sexually transmitted infection and the causative agent for cervical cancer, a frequently occurring malignant disease among women in developing countries. We assessed human papillomavirus awareness prior to the delivery of a brief information and education intervention, and human papillomavirus vaccine provision to female entertainment and sex workers (N = 220). At baseline, only 23.6% of women had heard of human papillomavirus. Following the educational intervention, 90% answered all the human papillomavirus knowledge questions correctly. Of 192 participants attending the first quarterly cohort visit where vaccine was offered, 149 (78%) were eligible for vaccination; HIV-positive (n = 32) and pregnant (n = 11) women were excluded. Acceptance of vaccine among eligible women was universal, and 79.2% completed the three-dose vaccination series. Women who reported use of amphetamine-type stimulants had significantly and independently lower odds of vaccine completion (adjusted odds ratio [AOR] 0.24; 95% confidence interval [CI] 0.08, 0.69). New pregnancies also had an impact on vaccine completion: 5.4% (8/149 5.4%) who started the series had to stop due to new pregnancy. Results demonstrate the effectiveness of a simple education intervention designed to increase human papillomavirus knowledge and the feasibility of successful human papillomavirus vaccine in a population that is often difficult to engage in preventive health care.
Collapse
Affiliation(s)
- Priya Wadhera
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, and Global Health Sciences, San Francisco, CA, USA
| | - Jennifer L Evans
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, and Global Health Sciences, San Francisco, CA, USA
| | - Ellen Stein
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, and Global Health Sciences, San Francisco, CA, USA
| | - Monica Gandhi
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Marie-Claude Couture
- School of Nursing and Health Professions, University of San Francisco, San Francisco, CA, USA
| | - Neth Sansothy
- National Center for HIV/AIDS, Dermatology and STDs (NCHADS), Phnom Penh, Cambodia
| | - Keo Sichan
- Cambodian Women's Development Association (CWDA), Phnom Penh, Cambodia
| | - Lisa Maher
- The Kirby Institute for Infection and Immunity (formerly the National Centre in HIV Epidemiology and Clinical Research); University of New South Wales, Sydney, NSW, Australia
| | - John Kaldor
- The Kirby Institute for Infection and Immunity (formerly the National Centre in HIV Epidemiology and Clinical Research); University of New South Wales, Sydney, NSW, Australia
| | - Kimberly Page
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, and Global Health Sciences, San Francisco, CA, USA Division of Epidemiology, Biostatistics and Preventive Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | | | - Kien
- Cambodian Women's Development Association (CWDA), Phnom Penh, Cambodia
| |
Collapse
|
993
|
Rosales R, Rosales C. Immune therapy for human papillomaviruses-related cancers. World J Clin Oncol 2014; 5:1002-1019. [PMID: 25493236 PMCID: PMC4259927 DOI: 10.5306/wjco.v5.i5.1002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 04/08/2014] [Accepted: 05/29/2014] [Indexed: 02/06/2023] Open
Abstract
Human papillomaviruses (HPVs) are a large family of double strand DNA viruses comprising more than 180 types. Infection with HPV is very common and it is associated with benign and malignant proliferation of skin and squamous mucosae. Many HPVs, considered low-risk such as HPV 6 and 11, produce warts; while high-risk viruses, such as HPVs 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, and 58, induce tumors. About 5% of all cancers in men and women are associated with HPV infection. Because there are not antiviral drugs for HPV infection, current therapies for low-risk HPV infections involve physical removal of the lesion by cryotherapy, trichloracetic acid, laser, or surgical removal. Surgical procedures are effective in the treatment of pre-cancerous lesions, however after these procedures, many recurrences appear due to new re-infections, or to failure of the procedure to eliminate the HPV. In addition, HPV can inhibit recognition of malignant cells by the immune system, leading to the development of cancer lesions. When this occurs, radiotherapy and chemotherapy are then used. Unfortunately, about 50% of the HPV-cancer patients still die. In the past decade, a better knowledge of the natural history of the virus-host interaction and of the immune response against this viral infection has brought new therapeutic strategies geared to modulate the immune system to generate an efficient virus-specific cytotoxic response. Novel HPV protein-expressing vaccines have shown some significant clinical efficacy and systemic HPV-specific cytotoxic T cell responses. This review will describe the current status of the several therapeutic strategies used to treat HPV-induced lesions, and discuss the various new therapies now being tested.
Collapse
|
994
|
Tsakogiannis D, Kyriakopoulou Z, Ruether IGA, Amoutzias GD, Dimitriou TG, Diamantidou V, Kotsovassilis C, Markoulatos P. Determination of human papillomavirus 16 physical status through E1/E6 and E2/E6 ratio analysis. J Med Microbiol 2014; 63:1716-1723. [DOI: 10.1099/jmm.0.076810-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Human papillomavirus (HPV) 16 genome integration into the host chromosome is a crucial event during the life cycle of the virus and a major step towards carcinogenesis. The integration of HPV16 DNA promotes a constitutive high expression level of E6 and E7 oncoproteins, resulting in the extensive proliferation of the infected epithelial cells. In the present report the physical status of the HPV16 genome was studied, through determination of E1/E6 and E2/E6 DNA copy number ratios in 61 cervical samples of low- and high-grade malignancy and 8 cervical cancer samples, all of them associated with HPV16 infection. The selection of E1, E2 and E6 amplification target regions was performed according to the most prevalent deleted/disrupted sites of E1 and E2 genes. For this target selection we also considered the most conserved regions of E1, E2 and E6 genes among the same HPV16 isolates that were recently reported by our group. The analysis of HPV16 DNA form revealed a significant association among the mixed DNA forms in low-grade and high-grade malignancies, (χ2, P<0.01). The comparative analysis of E1/E6 and E2/E6 in the same cervical samples provides an accurate picture of HPV16 DNA form and may reveal whether different HPV16 DNA integrants coexist in the same cervical sample or not. This study proposes that E1/E6 and E2/E6 ratios determine with accuracy the HPV16 DNA integration pattern and may predict multiple integration events in the examined sample, thus providing significant information about the progression of cervical dysplasia.
Collapse
Affiliation(s)
- Dimitris Tsakogiannis
- Microbiology–Virology Laboratory, Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Zaharoula Kyriakopoulou
- Microbiology–Virology Laboratory, Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Irina Georgia Anna Ruether
- Microbiology–Virology Laboratory, Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Grigoris D. Amoutzias
- Microbiology–Virology Laboratory, Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Tilemachos G. Dimitriou
- Microbiology–Virology Laboratory, Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Valentina Diamantidou
- Microbiology–Virology Laboratory, Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, Larissa, Greece
| | | | - Panayotis Markoulatos
- Microbiology–Virology Laboratory, Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, Larissa, Greece
| |
Collapse
|
995
|
Abstract
The development of efficacious prophylactic human papillomavirus vaccines provided an opportunity for the primary prevention of related infections and diseases. Certain oncogenic human papillomaviruses that preferentially infect the genital epithelium cause cervical cancer and a substantial proportion of anal, penile, vaginal, vulvar and oropharyngeal cancers. Following extensive clinical trials demonstrating their efficacy and safety, two vaccines have been in global use for over 6 years. This review summarises the accumulated evidence regarding their high level of efficacy, safety in population usage, reductions in genital warts, infections and cervical disease following their adoption, and facilitators and barriers to achieving high vaccination coverage. The review also discusses practical issues and frequently asked questions regarding duration of effect, vaccination of women treated for cervical disease and alternate vaccination schedules, as well as the need to review cervical screening strategies in the post- vaccination environment.
Collapse
Affiliation(s)
- Julia M L Brotherton
- National HPV Vaccination Program Register, VCS Inc, East Melbourne, Victoria, Australia
| |
Collapse
|
996
|
HPV vaccine acceptability in Africa: a systematic review. Prev Med 2014; 69:274-9. [PMID: 25451327 DOI: 10.1016/j.ypmed.2014.08.035] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 08/14/2014] [Accepted: 08/30/2014] [Indexed: 02/08/2023]
Abstract
OBJECTIVE The objective of this study was to provide a systematic review of peer-reviewed literature on the factors associated with HPV vaccine acceptability among adults in African countries. METHODS A systematic search was conducted across five electronic databases: EMBASE, PsychINFO, CINAHL, Global Health and Ovid MEDLINE, to identify studies related to HPV vaccination acceptability in African countries (August 2013). The Health Belief Model was used to guide data abstraction and synthesis. RESULTS Fourteen unique studies representing ten sub-Saharan African countries were identified, with more than half published within the last two years. Acceptability of the HPV vaccine for daughters was high (range 59-100%); however, vaccine-related awareness and knowledge were low. Perceived barriers including accessibility and cost concerns were important for acceptance, as were cues to action from healthcare providers and governments. CONCLUSIONS This review suggests that acceptability of the HPV vaccine in countries in this region will be high. Broad knowledge gaps were highlighted regarding HPV and cervical cancer and these should be addressed. Education on the vaccine's effectiveness and reducing perceived barriers to vaccination would also be useful. Public endorsement by governments and healthcare providers will likely also increase acceptance.
Collapse
|
997
|
Eng C, Chang GJ, Nancy You Y, Das P, Rodriguez-Bigas M, Xing Y, Vauthey JN, Rogers JE, Ohinata A, Pathak P, Sethi S, Phillips JK, Crane CH, Wolff RA. The role of systemic chemotherapy and multidisciplinary management in improving the overall survival of patients with metastatic squamous cell carcinoma of the anal canal. Oncotarget 2014; 5:11133-42. [PMID: 25373735 PMCID: PMC4294384 DOI: 10.18632/oncotarget.2563] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 10/02/2014] [Indexed: 01/30/2023] Open
Abstract
Metastatic squamous cell carcinoma (SCCA) of the anal canal is a rare malignancy for which no standard treatment algorithm exists. To determine the best approach, all patients diagnosed with metastatic SCCA of the anal canal treated at a single institution were evaluated for choice of chemotherapy and treatment outcome. A retrospective study from January 2000 to May 2012 was conducted. Electronic medical records were reviewed for diagnosis of metastatic SCCA of the anal canal. All patients were treatment naïve for metastatic disease and completed all radiographic imaging at our institution. The purpose of this study was to evaluate outcomes among patients who received systemic chemotherapy and if appropriate were referred for multidisciplinary intervention (e.g., surgery, radiofrequency ablation, etc.). Seventy-seven patients fulfilled eligibility criteria. Forty-two patients (55%) received 5-fluorouracil (5-FU) + cisplatin (PF); 24 patients (31%) received carboplatin + paclitaxel (CP); 11 patients (14%) received an alternative regimen. After a median follow-up of 42 months, the median progression-free survival (PFS) for all patients was 7 months; the median overall survival (OS) was 22 months. Thirty-three patients (43%) underwent multidisciplinary management for metastatic disease resulting in a median PFS of 16 months (95% CI: 9.2 -22.8) and median OS of 53 months (95% CI: 28.3 - 77.6). Systemic chemotherapy provides durable survival for patients with surgically unresectable metastatic SCCA of the anal canal. Multidisciplinary management for select patients with metastatic disease effectively improves survival and should be considered whenever possible.
Collapse
Affiliation(s)
- Cathy Eng
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030
| | - George J. Chang
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030
| | - Y. Nancy You
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030
| | - Prajnan Das
- Department of Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030
| | - Miguel Rodriguez-Bigas
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030
| | - Yan Xing
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030
| | - Jean-Nicolas Vauthey
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030
| | - Jane E. Rogers
- Division of Pharmacy, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030
| | - Aki Ohinata
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030
| | - Priyanka Pathak
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030
| | - Salil Sethi
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030
| | - Jonathan K. Phillips
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030
| | - Christopher H. Crane
- Department of Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030
| | - Robert A. Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030
| |
Collapse
|
998
|
Hariri S, Unger ER, Schafer S, Niccolai LM, Park IU, Bloch KC, Bennett NM, Steinau M, Johnson ML, Markowitz LE. HPV type attribution in high-grade cervical lesions: assessing the potential benefits of vaccines in a population-based evaluation in the United States. Cancer Epidemiol Biomarkers Prev 2014; 24:393-9. [PMID: 25416715 DOI: 10.1158/1055-9965.epi-14-0649] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Two currently available vaccines targeting human papillomavirus (HPV) types 16 and 18 could prevent 70% of cervical cancers and 50% of high-grade cervical lesions. Next-generation vaccines against additional types, such as a candidate 9-valent vaccine against HPV6/11/16/18/31/33/45/52/58, could further reduce HPV-associated disease burden. METHODS HPV was typed in archived tissues from women ages 21 to 39 years residing in five catchment areas in the United States with cervical intraepithelial neoplasia 2/3 and adenocarcinoma in situ (CIN2+) using L1 consensus PCR and type-specific hybridization. Type attribution was estimated using weights to account for lesions with multiple types detected. RESULTS From 2008 to 2011, 5,498 of 6,306 (87.2%) specimens obtained from 8,469 women with CIN2+ had valid typing results; HPV DNA was detected in 97.3%. Overall, 50.1% of lesions were attributable to HPV16/18, ranging from 50.3% to 52.4% among those ages 21 to 34 years, and significantly declined in 35 to 39 year-olds (43.5%). HPV16/18 attribution was higher in non-Hispanic whites (56.4%) versus racial/ethnic minorities (range, 41.8%-45.9%; P < 0.001). HPV31/33/45/52/58 attribution was 25.0% overall and increased with age (P < 0.001). A higher proportion of CIN2+ was attributable to HPV31/33/45/52/58 in non-Hispanic black (29.9%), Hispanic (29.2%), and Asian (33.1%) women compared with non-Hispanic whites (22.8%; P < 0.001). CONCLUSIONS Overall, 75% of lesions were attributable to 7 oncogenic HPV types: 50% to HPV16/18 and 25% to HPV31/33/45/52/58. HPV16/18 had the largest attributable fraction in CIN2+ across all subpopulations, although to a lesser extent in older women and racial/ethnic minorities. IMPACT Vaccines targeting additional oncogenic HPV types could prevent more high-grade cervical lesions, especially among racial/ethnic minorities.
Collapse
Affiliation(s)
- Susan Hariri
- Division of STD Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia.
| | - Elizabeth R Unger
- Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Sean Schafer
- HIV/STD/TB Program, Center for Public Health Practice, Oregon Public Health Division, Portland, Oregon
| | - Linda M Niccolai
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut
| | - Ina U Park
- California Department of Public Health, STD Control Branch, Richmond, California
| | - Karen C Bloch
- Departments of Medicine and Health Policy, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Nancy M Bennett
- Center for Community Health and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Martin Steinau
- Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Michelle L Johnson
- Division of STD Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Lauri E Markowitz
- Division of STD Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
999
|
Brotherton JM. Human papillomavirus vaccination. Br J Hosp Med (Lond) 2014; 75 Suppl 11:C165-8. [PMID: 25381861 DOI: 10.12968/hmed.2014.75.sup11.c165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Julia Ml Brotherton
- Medical Director of the National HPV Vaccination Program Register, Victorian Cytology Service, East Melbourne, Victoria 8002, Australia and Honorary Senior Fellow, School of Global and Population Health, University of Melbourne, Victoria, Australia
| |
Collapse
|
1000
|
Langers I, Renoux V, Reschner A, Touzé A, Coursaget P, Boniver J, Koch J, Delvenne P, Jacobs N. Natural killer and dendritic cells collaborate in the immune response induced by the vaccine against uterine cervical cancer. Eur J Immunol 2014; 44:3585-95. [PMID: 25229656 DOI: 10.1002/eji.201444594] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 08/14/2014] [Accepted: 09/11/2014] [Indexed: 11/09/2022]
Abstract
Virus-like particles (VLPs) of human papillomavirus (HPV) are used as a vaccine against HPV-induced cancer, and recently we have shown that these VLPs are able to activate natural killer (NK) cells. Since NK cells collaborate with dendritic cells (DCs) to induce an immune response against viral infections and tumors, we studied the impact of this crosstalk in the context of HPV vaccination. NK cells in the presence of HPV-VLPs enhanced DC-maturation as shown by an upregulation of CD86 and HLA-DR and an increased production of IL-12p70, but not of the immunosuppressive cytokine IL-10. This activation was bidirectional. Indeed, in the presence of HPV-VLPs, DCs further activated NK cells by inducing the upregulation of cell surface activation markers (CD69 and HLA-DR). The function of NK cells was also improved as shown by an increase in IFN-γ secretion and cytotoxic activity against an HPV(+) cell line. This crosstalk between NK cells and DCs needed CD40 interaction and IL-12p70 secretion, whereas NKG2D was not implicated. Our results provide insight into how VLPs interact with innate immune cells and how NK cells and DCs play a role in the immune response induced by this vaccine agent.
Collapse
Affiliation(s)
- Inge Langers
- Cellular and Molecular Immunology, GIGA-Research, University of Liège, Liège, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|