1
|
Abstract
Preclinical infection model systems are extremely valuable tools to aid in our understanding of Human Papillomavirus (HPV) biology, disease progression, prevention, and treatments. In this context, rodent papillomaviruses and their respective infection models are useful tools but remain underutilized resources in the field of papillomavirus biology. Two rodent papillomaviruses, MnPV1, which infects the Mastomys species of multimammate rats, and MmuPV1, which infects laboratory mice, are currently the most studied rodent PVs. Both of these viruses cause malignancy in the skin and can provide attractive infection models to study the lesser understood cutaneous papillomaviruses that have been frequently associated with HPV-related skin cancers. Of these, MmuPV1 is the first reported rodent papillomavirus that can naturally infect the laboratory strain of mice. MmuPV1 is an attractive model virus to study papillomavirus pathogenesis because of the ubiquitous availability of lab mice and the fact that this mouse species is genetically modifiable. In this review, we have summarized the knowledge we have gained about PV biology from the study of rodent papillomaviruses and point out the remaining gaps that can provide new research opportunities.
Collapse
|
2
|
Yu HT, Wang JY, Tian D, Wang MX, Li Y, Yuan L, Chen WJ, Li D, Zhuang M, Ling H. Comparison of the patterns of antibody recall responses to HIV-1 gp120 and hepatitis B surface antigen in immunized mice. Vaccine 2016; 34:6276-6284. [PMID: 27843002 DOI: 10.1016/j.vaccine.2016.10.063] [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: 01/22/2016] [Revised: 08/10/2016] [Accepted: 10/24/2016] [Indexed: 12/23/2022]
Abstract
To date, we still lack an ideal strategy for designing envelope glycoprotein (Env) vaccines to elicit potent protective antibodies against HIV-1 infection. Since the human hepatitis B virus surface antigen (HBsAg) is representative of effective vaccines that can induce ideal humoral immune responses, knowledge of how it elicits antibody responses and T helper cells would be an useful reference for HIV vaccine development. We compared the characteristics of the HIV-1 Env gp120 trimer and HBsAg in antibody elicitation and induction of T follicular helper (Tfh) and memory B cells in immunized Balb/c mice. Using the strategy of protein prime-protein boost, we found that HIV-1 gp120 induced slower recall antibody responses but redundant non-specific IgG responses at early time after boosting compared to HBsAg. The higher frequency of PD-1hiCD4+ T cells and Tfh cells that appeared at the early time point after gp120 boosting is likely to limit the development of memory B cells, memory T cells, and specific antibody recall responses. These findings regarding the different features of HIV envelope and HBsAg in T helper cell responses may provide a direction to improve HIV envelope immunogenicity.
Collapse
Affiliation(s)
- Hao-Tong Yu
- Department of Microbiology, Harbin Medical University, Harbin, China
| | - Jia-Ye Wang
- Department of Microbiology, Harbin Medical University, Harbin, China; Heilongjiang Provincial Key Laboratory of Infection and Immunity, Key Laboratory of Pathogen Biology, Harbin, China; Wu Lien-Teh Institute, Harbin Medical University, Harbin, China
| | - Dan Tian
- Department of Microbiology, Harbin Medical University, Harbin, China
| | - Ming-Xia Wang
- Department of Microbiology, Harbin Medical University, Harbin, China
| | - Yan Li
- Department of Microbiology, Harbin Medical University, Harbin, China; Heilongjiang Provincial Key Laboratory of Infection and Immunity, Key Laboratory of Pathogen Biology, Harbin, China; Wu Lien-Teh Institute, Harbin Medical University, Harbin, China
| | - Li Yuan
- Department of Microbiology, Harbin Medical University, Harbin, China
| | - Wen-Jiang Chen
- Department of Microbiology, Harbin Medical University, Harbin, China
| | - Di Li
- Department of Microbiology, Harbin Medical University, Harbin, China; Heilongjiang Provincial Key Laboratory of Infection and Immunity, Key Laboratory of Pathogen Biology, Harbin, China
| | - Min Zhuang
- Department of Microbiology, Harbin Medical University, Harbin, China; Heilongjiang Provincial Key Laboratory of Infection and Immunity, Key Laboratory of Pathogen Biology, Harbin, China; Wu Lien-Teh Institute, Harbin Medical University, Harbin, China.
| | - Hong Ling
- Department of Microbiology, Harbin Medical University, Harbin, China; Heilongjiang Provincial Key Laboratory of Infection and Immunity, Key Laboratory of Pathogen Biology, Harbin, China; Wu Lien-Teh Institute, Harbin Medical University, Harbin, China; Department of Parasitology, Harbin Medical University, Harbin, China.
| |
Collapse
|
3
|
Sabah SN, Gazi MA, Sthity RA, Husain AB, Quyyum SA, Rahman M, Islam MR. Designing of Epitope-Focused Vaccine by Targeting E6 and E7 Conserved Protein Sequences: An Immuno-Informatics Approach in Human Papillomavirus 58 Isolates. Interdiscip Sci 2016; 10:251-260. [PMID: 27640170 DOI: 10.1007/s12539-016-0184-5] [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: 12/10/2015] [Revised: 08/02/2016] [Accepted: 09/02/2016] [Indexed: 10/21/2022]
Abstract
Human papillomavirus (HPV) is a DNA virus that belongs to the papillomavirus family and is capable of infecting humans. Currently, few vaccines are available to prevent infection by HPV. However, they are not so much effective and provide little benefit to women who have already been infected with HPV. The aim of this study was to design epitope-based vaccines of HPV58 by targeting E6 and E7 proteins of HPV58. Proteomic sequences were retrieved from different isolates at different time periods and later analyzed by performing alignment of these sequences. To ensure the capacity of humoral and cell-mediated immunity, both B cell and T cell immunity were checked for the peptides. For E6 protein, the peptide sequence from 48 to 54 amino acids and one 9-m epitope ETSVHEIEL were the most potential B cell and T cell epitopes, respectively. This peptide could interact with as many as eight MHC-1 alleles and showed high population coverage up to 90.31 %. On the other hand, the peptide region for the E7 protein ranged from 27 to 33 amino acids and two 9-m epitopes QAQPATANY, SSDEDEIGL were found as the most potential B cell and T cell epitopes, respectively. The peptide sequences could interact with as many as seven MHC-1 alleles and showed population coverage up to 90.31 %. Furthermore, conservancy analysis was also performed using in silico tools and showed a conservancy of 100 % for all the selected epitopes. In addition to this, the allergenicity of the epitopes was also evaluated. Although the study requires further in vitro and in vivo screening, this epitope-focused peptide vaccine designing opens up a new skyline that holds a prospective future in HPV research.
Collapse
Affiliation(s)
| | - Md Amran Gazi
- Nutrition and Clinical Services Division, Centre for Nutrition and Food Security, International Centre for Diarrhoeal Disease Research, GPO Box 128, Dhaka, 1000, Bangladesh.
| | - Rahvia Alam Sthity
- Immunobiology, Nutrition and Toxicology Laboratory, International Centre for Diarrheal Disease Research, Dhaka, Bangladesh
| | - Amena Binte Husain
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - Salwa Abdul Quyyum
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - Mustafizur Rahman
- Center for Bio-Medical Research, Manarat University, Dhaka, Bangladesh
| | - Md Rezaul Islam
- International Max Planck Research School, University of Göttingen, 37075, Göttingen, Germany.
| |
Collapse
|
4
|
Dillner J, Arbyn M, Unger E, Dillner L. Monitoring of human papillomavirus vaccination. Clin Exp Immunol 2010; 163:17-25. [PMID: 21062269 DOI: 10.1111/j.1365-2249.2010.04268.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Persistent infection with oncogenic human papillomavirus (HPV) is a necessary causal factor in the development of cervical cancer. Moreover, HPV, predominately type 16 and to a lesser degree type 18, is linked causally to varying proportions of other anogenital cancers (vulva, vagina, penis, anus) as well as cancers elsewhere in the body (oropharynx, larynx, conjunctiva). HPV types 6 and 11 cause most of genital warts and recurrent respiratory papillomatosis. Effective prophylactic vaccines have been developed. In this review, we address briefly the immunological aspects of HPV infection and the results of HPV vaccination trials. Internationally standardized monitoring and evaluation of prophylactic HPV vaccination programmes will be essential for arriving at the most cost-effective strategies for cancer control.
Collapse
Affiliation(s)
- J Dillner
- WHO HPV LabNet Global Reference Laboratory at Department of Clinical Microbiology, University Hospital, Malmö,
| | | | | | | |
Collapse
|
5
|
Zhou L, Zhu T, Ye X, Yang L, Wang B, Liang X, Lu L, Tsao YP, Chen SL, Li J, Xiao X. Long-term protection against human papillomavirus e7-positive tumor by a single vaccination of adeno-associated virus vectors encoding a fusion protein of inactivated e7 of human papillomavirus 16/18 and heat shock protein 70. Hum Gene Ther 2010; 21:109-19. [PMID: 19715402 DOI: 10.1089/hum.2009.139] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
We investigated a gene vaccine strategy against human papillomavirus (HPV)-induced cancer and premalignant diseases, using adeno-associated virus (AAV) vector encoding the viral E7 oncoproteins as the tumor antigens from HPV serotypes 16 (HPV16) and 18 (HPV18). Genetically inactivated E7 proteins were fused with a heat shock protein 70 (hsp70) to minimize the risk of cell transformation and enhance immune responses. The fusion protein gene was packaged in AAV serotype 1 or 2 (AAV1 or 2) for efficient in vivo gene expression. Our results showed that after a single intramuscular injection, the AAV1 vector elicited stronger HPV-specific cytotoxic T lymphocyte (CTL) responses and interferon-gamma secretion when compared with the AAV2 vector. Prophylactic immunization with AAV1 protected 100% of the mice from tumor growth for more than 1 year, whereas all the control mice immunized with either a LacZ vector or saline grew large tumors and died within 6 weeks after inoculation of E7-positive tumor cell line TC-1. In addition, this single-dose AAV1 vaccination completely protected the mice against second and third challenges with higher numbers of TC-1 cells. Despite lower CTL responses against the E7 antigens, AAV2 vector prophylactic immunization was also sufficient to protect 100% of the mice against the initial and second tumor challenges and 70% of the mice against the third challenge. In addition, therapeutic immunization with AAV1 after palpable tumor formation inhibited tumor growth and caused tumor regression in some mice. Thus, our studies support the potential of AAV vectors as a genetic vaccine for the prevention and treatment of HPV-induced malignancies.
Collapse
Affiliation(s)
- Liqiao Zhou
- Molecular Therapy Laboratory, Department of Orthopedic Surgery, University of Pittsburgh School of Medicine , Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Christensen ND, Bounds CE. Cross-protective responses to human papillomavirus infection. Future Virol 2010. [DOI: 10.2217/fvl.10.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Human papillomavirus (HPV) infections with oncogenic types account for approximately 500,000 deaths per year worldwide, predominantly in underdeveloped countries. The major cause of death is cervical cancer in women, but some additional cancers of the head and neck and anogenital sites also have an HPV etiology. Current virus-like particle-based vaccines are in clinical trials, and show very strong, long-lasting protection against vaccine-matched HPV types. These vaccines currently contain virus-like particles for the HPV types 6, 11, 16 and 18 (Gardasil®) and HPV16 and -18 (Cervarix®). Although type-specific neutralizing antibodies develop from immunizations with these virus-like particle vaccines, promising evidence for cross-protection against related but nonvaccine HPV types is emerging. Strategies to increase cross-protection to cover all oncogenic HPV types (currently approximately 20 types) are underway. These strategies include increasing the number of HPV types in the virus-like particle vaccine, and to the development of second-generation HPV vaccines that include the minor coat protein.
Collapse
Affiliation(s)
- ND Christensen
- Penn State University, College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - CE Bounds
- Penn State University, College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| |
Collapse
|
7
|
Samara RN, Khleif SN. HPV as a model for the development of prophylactic and therapeutic cancer vaccines. Curr Mol Med 2009; 9:766-73. [PMID: 19689303 PMCID: PMC6948838 DOI: 10.2174/156652409788970625] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
HPV has been linked to many human malignancies and, as such, represents a major public health crisis. The understanding of HPV biology, however, has helped tremendously in developing prophylactic vaccines, which should help in decreasing mortality due to HPV infections. Understanding HPV biology has allowed researchers to use the virus as a model for the development of not only prophylactic vaccines, but also therapeutic ones. The advantages of HPV as a model stem from the limited number of proteins encoded by the HPV genome that can be targeted by vaccines, and also from the restricted expression of certain viral proteins during different stages of infection. In this review, we discuss how HPV can be used as a model for the development of both prophylactic and therapeutic vaccines.
Collapse
Affiliation(s)
- Raed N. Samara
- Cancer Vaccine Section, National Cancer Institute, NCI-NNMC, Bldg 10, Rm. 5101, Bethesda, MD 20889, USA
| | - Samir N. Khleif
- Cancer Vaccine Section, National Cancer Institute, NCI-NNMC, Bldg 10, Rm. 5101, Bethesda, MD 20889, USA
| |
Collapse
|
8
|
Prophylactic HPV vaccines: new interventions for cancer control. Vaccine 2008; 26:6244-57. [PMID: 18694795 DOI: 10.1016/j.vaccine.2008.07.056] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2008] [Revised: 07/01/2008] [Accepted: 07/13/2008] [Indexed: 12/28/2022]
Abstract
Human Papillomavirus (HPV) infection causes cervical cancer, a significant portion of anal, vulvar, vaginal, and oropharyngeal cancers, genital warts, and recurrent respiratory papillomatosis (RRP). HPV 16 and 18 cause 70-90% of HPV-related cancers whereas HPV 6 and 11 cause 90% of RRP and genital wart cases. Together these four types cause 30-50% of all cervical intraepithelial neoplasia such as those detected by Papinicalou screening. In June 2006, a quadrivalent HPV (6, 11, 16, 18) vaccine was licensed in the United States, and subsequently in the European Union (September 2006), both following expedited review. We describe the primary objectives of the quadrivalent HPV vaccine clinical trial program including studies in females aged 9-45 and males aged 9-26. Planned long-term efficacy and safety evaluations, as well as programs to evaluate vaccine impact on oropharyngeal cancer are also described.
Collapse
|
9
|
Hu J, Budgeon LR, Cladel NM, Culp TD, Balogh KK, Christensen ND. Detection of L1, infectious virions and anti-L1 antibody in domestic rabbits infected with cottontail rabbit papillomavirus. J Gen Virol 2008; 88:3286-3293. [PMID: 18024897 DOI: 10.1099/vir.0.82879-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Shope papillomavirus or cottontail rabbit papillomavirus (CRPV) is one of the first small DNA tumour viruses to be characterized. Although the natural host for CRPV is the cottontail rabbit (Sylvilagus floridanus), CRPV can infect domestic laboratory rabbits (Oryctolagus cuniculus) and induce tumour outgrowth and cancer development. In previous studies, investigators attempted to passage CRPV in domestic rabbits, but achieved very limited success, leading to the suggestion that CRPV infection in domestic rabbits was abortive. The persistence of specific anti-L1 antibody in sera from rabbits infected with either virus or viral DNA led us to revisit the questions as to whether L1 and infectious CRPV can be produced in domestic rabbit tissues. We detected various levels of L1 protein in most papillomas from CRPV-infected rabbits using recently developed monoclonal antibodies. Sensitive in vitro infectivity assays additionally confirmed that extracts from these papillomas were infectious. These studies demonstrated that the CRPV/New Zealand White rabbit model could be used as an in vivo model to study natural virus infection and viral life cycle of CRPV and not be limited to studies on abortive infections.
Collapse
Affiliation(s)
- Jiafen Hu
- Jake Gittlen Cancer Research Foundation, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Lynn R Budgeon
- Jake Gittlen Cancer Research Foundation, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Nancy M Cladel
- Jake Gittlen Cancer Research Foundation, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Timothy D Culp
- Jake Gittlen Cancer Research Foundation, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Karla K Balogh
- Jake Gittlen Cancer Research Foundation, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Neil D Christensen
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Jake Gittlen Cancer Research Foundation, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
10
|
Hu J, Cladel N, Peng X, Balogh K, Christensen ND. Protective immunity with an E1 multivalent epitope DNA vaccine against cottontail rabbit papillomavirus (CRPV) infection in an HLA-A2.1 transgenic rabbit model. Vaccine 2007; 26:809-16. [PMID: 18187239 DOI: 10.1016/j.vaccine.2007.11.081] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2007] [Revised: 11/23/2007] [Accepted: 11/29/2007] [Indexed: 10/22/2022]
Abstract
Cottontail rabbit papillomavirus (CRPV)/rabbit model is widely used to study pathogenesis of papillomavirus infections and malignant tumor progression. Recently, we established HLA-A2.1 transgenic rabbit lines and demonstrated efficacy for the testing of immunogenicity of a well-known A2-resticted epitope (HPV16E7/82-90) [Hu J, Peng X, Schell TD, Budgeon LR, Cladel NM, Christensen ND. An HLA-A2.1-transgenic rabbit model to study immunity to papillomavirus infection. J Immunol 2006;177(11):8037-45]. In the present study, we screened five HLA-A2.1 restricted epitopes from CRPVE1 (selected using online MHCI epitope prediction software) and constructed a multivalent epitope DNA vaccine (CRPVE1ep1-5). CRPVE1ep1-5 and a control DNA vaccine (Ub3) were then delivered intracutaneously onto normal and HLA-A2.1 transgenic rabbits, respectively, by a helium-driven gene-gun delivery system. One, two or three immunizations were given to different groups of animals from both New Zealand White outbred and EIII/JC inbred genetic background. Two and three immunizations with CRPVE1ep1-5 DNA vaccine provided complete protection against viral DNA infection of HLA-A2.1 transgenic rabbits from both genetic backgrounds but not in the control-vaccinated groups. One immunization, however, failed to protect HLA-A2.1 transgenic rabbits against viral DNA infection. This study further demonstrated that the HLA-A2.1 transgenic rabbits can be used to test the immunogenicity of HLA-A2.1 restricted epitopes identified by MHCI epitope predication software.
Collapse
Affiliation(s)
- Jiafen Hu
- Jake Gittlen Cancer Research Foundation, Department of Pathology, Pennsylvania State University college of medicine, Hershey, PA 17033, USA
| | | | | | | | | |
Collapse
|
11
|
Dillner J, Arbyn M, Dillner L. Translational mini-review series on vaccines: Monitoring of human papillomavirus vaccination. Clin Exp Immunol 2007; 148:199-207. [PMID: 17437418 PMCID: PMC1868871 DOI: 10.1111/j.1365-2249.2007.03384.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Persistent infection with oncogenic human papillomavirus (HPV) is a necessary cause of cervical cancer. Moreover, HPV type 16 (and to a lesser degree HPV type 18) is linked with more rare cancers, namely cancer of the vulva, vagina, penis, anus, oropharynx and larynx. Effective prophylactic vaccines have been developed. In this review, we briefly address immunological aspects of HPV infection and the results of HPV vaccination trials. Internationally standardized monitoring and evaluation of prophylactic HPV vaccination programmes will be essential for arriving at the most (cost-)effective strategies for cancer control.
Collapse
Affiliation(s)
- J Dillner
- WHO Global Reference Laboratory for HPV Diagnosis and Control, Department of Clinical Microbiology, University Hospital, SE-20502 Malmö, Sweden.
| | | | | |
Collapse
|
12
|
Abstract
Papilloma viruses (PV) have been known to cause benign and malignant tumors in animals for more than 100 years. It took over 20 years to win general acceptance for their causative role in anogenital carcinomas in humans in particular in cervial carcinoma. Extensive research has led to the development of a prophylactic vaccine which is now commercially available. It remains to be investigated if HPV-specific therapeutic vaccines can be developed.
Collapse
Affiliation(s)
- M Müller
- Deutsches Krebsforschungszentrum Heidelberg, Heidelberg, Deutschland
| | | |
Collapse
|
13
|
Hu J, Peng X, Budgeon LR, Cladel NM, Balogh KK, Christensen ND. Establishment of a cottontail rabbit papillomavirus/HLA-A2.1 transgenic rabbit model. J Virol 2007; 81:7171-7. [PMID: 17459918 PMCID: PMC1933279 DOI: 10.1128/jvi.00200-07] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Three transgenic rabbit lines that express a well-characterized human major histocompatibility complex class I (MHC-I) gene (HLA-A2.1) have been established. All three lines carry the HLA-A2.1 heavy chain and are able to pass the transgene to their offspring with both the outbred and the inbred EIII/JC genetic background. HLA-A2.1 colocalizes exclusively with rabbit MHC-I on the cell surfaces. These HLA-A2.1 transgenic rabbits demonstrated infection patterns similar to those found after cottontail rabbit papillomavirus (CRPV) challenge when compared with results in normal rabbits, although higher regression rates were found in HLA-A2.1 transgenic rabbits. Because the CRPV genome can accommodate significant modifications, the CRPV/HLA-A2.1 rabbit model has the potential to be used to screen HLA-A2.1-restricted immunogenic epitopes from human papillomaviruses in the context of in vivo papillomavirus infection.
Collapse
Affiliation(s)
- Jiafen Hu
- Jake Gittlen Cancer Research Foundation, 500 University Dr., Pennsylvania State University, Hershey, PA 17033, USA
| | | | | | | | | | | |
Collapse
|
14
|
Brandsma JL, Shylankevich M, Su Y, Roberts A, Rose JK, Zelterman D, Buonocore L. Vesicular stomatitis virus-based therapeutic vaccination targeted to the E1, E2, E6, and E7 proteins of cottontail rabbit papillomavirus. J Virol 2007; 81:5749-58. [PMID: 17392369 PMCID: PMC1900321 DOI: 10.1128/jvi.02835-06] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Persistent human papillomavirus (HPV)-associated benign and malignant lesions are a major cause of morbidity and mortality worldwide. Vaccination against HPV early proteins could provide an effective means of treating individuals with established infections. Recombinant vesicular stomatitis virus (VSV) vectors have been used previously to elicit strong humoral and cellular immune responses and develop prophylactic vaccines. We have shown that VSV vectors also can be used to elicit therapeutic immunity in the cottontail rabbit papillomavirus (CRPV)-rabbit model of high-risk HPV infection. In the present study, three new VSV vectors expressing the CRPV E1, E2, or E7 protein were produced and compared to the previously generated VSV-E6 vector for therapeutic efficacy. To determine whether vaccine efficacy could be augmented by simultaneous vaccination against two CRPV proteins, the four vaccines were delivered individually and in all possible pairings to rabbits 1 week after CRPV infection. Control rabbits received the recombinant wild-type VSV vector or medium only. Cumulative papilloma volumes were computed for analysis of the data. The analyses showed that VSV-based vaccination against the E1, E2, E6, or E7 protein significantly reduced papilloma volumes relative to those of the controls. Furthermore, VSV-based CRPV vaccination cured all of the papillomas in 5 of 30 rabbits. Of the individual vaccines, VSV-E7 was the most effective. The VSV-E7 vaccine alone was the most effective, as it reduced cumulative papilloma volumes by 96.9% overall, relative to those of the controls, and ultimately eliminated all of the disease in all of the vaccinees. Vaccine pairing was not, however, found to be beneficial, suggesting antigenic competition between the coexpressed CRPV proteins. These preclinical results, obtained in a physiologically relevant animal model of HPV infection, demonstrate that VSV vectors deserve serious consideration for further development as therapeutic antitumor vaccines.
Collapse
Affiliation(s)
- Janet L Brandsma
- Section of Comparative Medicine, Yale University School of Medicine, 375 Cedar Street, 2 FMB, New Haven, CT 06520-8016, USA.
| | | | | | | | | | | | | |
Collapse
|
15
|
Arbyn M, Dillner J. Review of current knowledge on HPV vaccination: An Appendix to the European Guidelines for Quality Assurance in Cervical Cancer Screening. J Clin Virol 2007; 38:189-97. [PMID: 17258503 DOI: 10.1016/j.jcv.2006.12.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Revised: 11/28/2006] [Accepted: 12/07/2006] [Indexed: 11/23/2022]
Abstract
The recognition of a strong etiological relationship between infection with high-risk human papillomavirusses and cervical cancer has prompted research to develop and evaluate prophylactic and therapeutic vaccines. One prophylactic quadrivalent vaccine using L1 virus-like particles (VLP) of HPV 6, 11, 16 and 18 is available on the European market since the end of 2006 and it is expected that a second bivalent vaccine containing VLPs of HPV16 and HPV18 will become available in 2007. Each year, HPV16 and HPV18 cause approximately 43,000 cases of cervical cancer in the European continent. Results from the phase-IIb and III trials published thus far indicate that the L1 VLP HPV vaccine is safe and well-tolerated. It offers HPV-naive women a very high level of protection against HPV persistent infection and cervical intra-epithelial lesions associated with the types included in the vaccine. HPV vaccination should be offered to girls before onset of sexual activity. While prophylactic vaccination is likely to provide important future health gains, cervical screening will need to be continued for the whole generation of women that is already infected with the HPV types included in the vaccine. Phase IV studies are needed to demonstrate protection against cervical cancer and to verify duration of protection, occurrence of replacement by non-vaccine types and to define future policies for screening of vaccinated cohorts. The European Guidelines on Quality Assurance for Cervical Cancer Screening provides guidance for secondary prevention by detection and management of precursors lesions of the cervix. The purpose of the appendix on vaccination is to present current knowledge. Developing guidelines for future use of HPV vaccines in Europe, is the object of a new grant offered by the European Commission.
Collapse
Affiliation(s)
- Marc Arbyn
- Unit of Cancer Epidemiology, Scientific Institute of Public Health, Brussels, Belgium.
| | | |
Collapse
|
16
|
Rodríguez-Cerdeira C, Alba Menéndez A, Vilata Corell JJ. Desarrollo de nuevas vacunas frente al virus del papiloma humano. ACTA ACUST UNITED AC 2007. [DOI: 10.1016/s0213-9251(07)73014-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
17
|
Ait Menguellet S, Collinet P, Houfflin Debarge V, Nayama M, Vinatier D, Leroy JL. Management of multicentric lesions of the lower genital tract. Eur J Obstet Gynecol Reprod Biol 2006; 132:116-20. [PMID: 16713062 DOI: 10.1016/j.ejogrb.2006.04.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2005] [Revised: 02/21/2006] [Accepted: 04/10/2006] [Indexed: 10/24/2022]
Abstract
OBJECTIVES To report management and outcome of multicentric lesions of the lower genital tract. To define risk factors of recurrence. STUDY DESIGN Retrospective review of multicentric dysplasias treated in our colposcopic clinic between 1996 and 2003. Multicentric dysplasias included CIN with VAIN and/or VIN. After primary treatment, follow-up was colposcopic, cytologic and virologic. RESULTS Forty-four patients presented multicentric lesions out of 998 patients referred for CIN (4.4%). The average age was 36.8 years. Immunologic disorders were present in 20.4%. Ninety-one percent had cervicovaginal or cervicovulvar lesions, only 9% had three sites of genital dysplasia. 53.3% of lesions were concomitant. 79.5% of CIN were high grade, 62.5% of VAIN low grade and 62.5% of VIN high grade. Therapeutic modalities were as follows: conization for CIN (70.4%), CO2 laser for VAIN (33.3%) and surgery for VIN (41.7%). Forty patients were followed and had at least one post-treatment cytologic control; 55% of them had residual disease. Out of the 23 patients with at least two negative controls after treatment, 43.5% presented recurrence. Risk of recurrence was not statistically bound to such parameters as tabagism, immunologic disorder, high grade lesions, non-surgical treatment, and persistence of HPV infection after treatment. CONCLUSION Multicentric dysplasias are associated with high rate of residual lesion and recurrence. Management of these lesions require long term follow-up.
Collapse
Affiliation(s)
- Sonia Ait Menguellet
- Hôpital Jeanne de Flandre, Clinique de Gynécologie, Centre Hospitalier Régional Universitaire de Lille, 2 Avenue Oscar Lambret, 59 037 Lille Cedex, France
| | | | | | | | | | | |
Collapse
|
18
|
Antonis AFG, Bruschke CJM, Rueda P, Maranga L, Casal JI, Vela C, Hilgers LAT, Belt PBGM, Weerdmeester K, Carrondo MJT, Langeveld JPM. A novel recombinant virus-like particle vaccine for prevention of porcine parvovirus-induced reproductive failure. Vaccine 2006; 24:5481-90. [PMID: 16730104 DOI: 10.1016/j.vaccine.2006.03.089] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2005] [Revised: 03/24/2006] [Accepted: 03/28/2006] [Indexed: 11/17/2022]
Abstract
A novel vaccine against porcine parvovirus (PPV), composed of recombinant virus-like particles (PPV-VLPs) produced with the baculovirus expression vector system (BEVS) at industrial scale, was tested for its immunogenicity and protective potency. A formulation of submicrogram amounts of PPV-VLPs in a water-in-mineral oil adjuvant evoked high serum antibody titres in both guinea pigs, used as reference model, and target species, pigs. A single immunisation with 0.7microg of this antigen yielded complete foetal protection against PPV infection after challenge with a virulent strain of this virus. Furthermore, also in the presence of mild adjuvants the protective action of these PPV-VLPs is excellent. This recombinant subunit vaccine overcomes some of the drawbacks of classical PPV vaccines.
Collapse
Affiliation(s)
- Adriaan F G Antonis
- Animal Sciences Group (ASG, Previously ID-Lelystad), P.O. Box 65, 8200 AB Lelystad, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Qian J, Dong Y, Pang YYS, Ibrahim R, Berzofsky JA, Schiller JT, Khleif SN. Combined prophylactic and therapeutic cancer vaccine: Enhancing CTL responses to HPV16 E2 using a chimeric VLP in HLA-A2 mice. Int J Cancer 2006; 118:3022-9. [PMID: 16425257 DOI: 10.1002/ijc.21781] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
We identified the strategies to induce a CTL response to human papillomavirus (HPV) 16 E2 in HLA-A2 transgenic mice (AAD). A chimeric HPV16 virus-like particle (VLP) that includes full length HPV16 E7 and E2 (VLP-E7E2) was generated. The combination of E2 and E7 has the advantage that E2 is expressed in early dysplasia and neoplasia lesions, where E7 is expressed in more advance lesions. Since T cell response to E2 is less defined, we first evaluated the strategies to enhancing CD8(+) T cell responses to HPV E7, using different combinations of immune-modulators with VLP-E7E2. Data showed that the CTL response to E7 could be significantly enhanced by coinjection of GM-CSF and anti-CD40 antibodies with chimeric VLP-E7E2 without adjuvant. However, using the same combination, a low level of CD8(+) T cell response to E2 was detected. To enhance the CD8+ T cell response to E2, we analyzed T cell epitopes from E2 sequence. A heterogenous prime-boost with chimeric VLP-E7E2 and E2 peptides was performed. The data showed that the priming with chimeric VLP-E7E2, followed by boosting with E2 peptides, gave a better CTL response than 2 immunizations with E2 peptides. The enhanced immunity is due to the increase of CD11c(+) and CD11c(+) CD40(+) double positive dendritic cells in mice that received immune-modulators, GM-CSF and anti-CD40. Furthermore, the level of anti-L1 antibodies remains similar in mice immunized with chimeric VLP with/without immune-modulators. Thus, the data suggested that the chimeric VLP-E7E2 has a therapeutic potential for the treatment of HPV-associated CINs and cancer without diminishing VLPs potential as a prophylactic vaccine by inducing anti-L1 antibodies against free virus.
Collapse
Affiliation(s)
- Jiahua Qian
- Vaccine Branch, NCI, National Naval Medical Center, Bldg 8, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
The ability to generate human papillomavirus virus (HPV)-like particles by the synthesis and self-assembly in vitro of the major virus capsid protein L1 has transformed our prospects for preventing cervical carcinoma in women. These particles provide vaccines that are immunogenic and safe, and data from proof-of-principle efficacy trials strongly suggest that they will protect against persistent HPV infection and cervical intraepithelial neoplasia. However, the duration of protection provided by these vaccines is not known, the antibody responses induced are HPV-type-specific and immunisation must occur pre-exposure to the virus. Second-generation vaccines could include an early antigen for protection post exposure and alternative delivery systems might be needed for the developing world. Therapeutic vaccines for low-grade intraepithelial disease are realistic but high-grade disease presents major hurdles for immunotherapies.
Collapse
Affiliation(s)
- Margaret Stanley
- Department of Pathology, Tennis Court Road, Cambridge CB2 1QP, UK.
| |
Collapse
|
21
|
Affiliation(s)
- Mario Preti
- Department of Obstetrics and Gynaecology, University of Torino, Torino, Italy.
| | | | | | | |
Collapse
|
22
|
Peng S, Trimble C, Ji H, He L, Tsai YC, Macaes B, Hung CF, Wu TC. Characterization of HPV-16 E6 DNA vaccines employing intracellular targeting and intercellular spreading strategies. J Biomed Sci 2005; 12:689-700. [PMID: 16200349 DOI: 10.1007/s11373-005-9012-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2005] [Accepted: 07/25/2005] [Indexed: 10/25/2022] Open
Abstract
Human papillomavirus (HPV) E6 and E7 are consistently expressed and are responsible for the malignant transformation of HPV-associated lesions. Thus, E6 and E7 represent ideal targets for therapeutic HPV vaccine development. We have previously used the gene gun approach to test several intracellular targeting and intercellular spreading strategies targeting HPV-16 E7. These strategies include the use of the sorting signal of lysosome-associated membrane protein (LAMP-1), Mycobacterium tuberculosis heat shock protein 70 (HSP70), calreticulin (CRT) and herpes simplex virus type 1 (HSV-1) VP22 proteins. All of these strategies have been shown to be capable of enhancing E7-DNA vaccine potency. In the current study, we have characterized DNA vaccines employing these intracellular targeting or intercellular spreading strategies targeting HPV-16 E6 for their ability to generate E6-specific CD8+ T cell immune responses and antitumor effects against an E6-expressing tumor cell line, TC-1, in C57BL/6 mice. We found that all the intracellular targeting strategies (CRT, LAMP-1, HSP70) as well as the intercellular spreading strategy (VP22) were able to enhance E6 DNA vaccine potency, although the orientation of HSP70 linked to E6 antigen in the E6 DNA vaccine appears to be important for the HSP70 strategy to work. The enhanced E6-specific CD8+ T cell immune response in vaccinated mice also translated into potent antitumor effects against TC-1 tumor cells. Our data indicate that all of the intracellular targeting and intercellular spreading strategies that have been shown to enhance E7 DNA vaccine potency were also able to enhance E6 DNA vaccine potency.
Collapse
Affiliation(s)
- Shiwen Peng
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Baculovirus as versatile vectors for protein expression in insect and mammalian cells. Nat Biotechnol 2005; 23:567-75. [PMID: 15877075 PMCID: PMC3610534 DOI: 10.1038/nbt1095] [Citation(s) in RCA: 694] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Today, many thousands of recombinant proteins, ranging from cytosolic enzymes to membrane-bound proteins, have been successfully produced in baculovirus-infected insect cells. Yet, in addition to its value in producing recombinant proteins in insect cells and larvae, this viral vector system continues to evolve in new and unexpected ways. This is exemplified by the development of engineered insect cell lines to mimic mammalian cell glycosylation of expressed proteins, baculovirus display strategies and the application of the virus as a mammalian-cell gene delivery vector. Novel vector design and cell engineering approaches will serve to further enhance the value of baculovirus technology.
Collapse
|
24
|
Johnston KB, Monteiro JM, Schultz LD, Chen L, Wang F, Ausensi VA, Dell EC, Santos EB, Moore RA, Palker TJ, Stanley MA, Jansen KU. Protection of beagle dogs from mucosal challenge with canine oral papillomavirus by immunization with recombinant adenoviruses expressing codon-optimized early genes. Virology 2005; 336:208-18. [PMID: 15892962 DOI: 10.1016/j.virol.2005.03.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2004] [Revised: 12/16/2004] [Accepted: 03/21/2005] [Indexed: 10/25/2022]
Abstract
Replication-deficient adenoviral (rAd5) vaccines containing codon-optimized E1, E2, E4, and E7 genes of canine oral papillomavirus (COPV) were tested singly or in combination to determine which vaccines could protect against mucosal challenge with COPV. In three studies, groups of 4-6 beagle dogs were immunized subcutaneously (s.c.) with 10(11) rAd5 at 8-10 weeks and 4-6 weeks prior to challenge with infectious COPV particles at multiple oral mucosal sites. Control dogs were immunized with equivalent doses of rAd5 expressing human papillomavirus (HPV) type 16 L1 (rAd5-HPV-16 L1). In the first study, complete protection from COPV-induced papillomas was achieved by immunization with rAd5 vaccine combinations expressing either E1 + E2 or E1 + E2 + E4 + E7; whereas two of six dogs immunized with rAd5-E4 + rAd5-E7 and six of six rAd5-HPV16-L1-immunized control dogs developed oral papillomas. In two subsequent studies, rAd5-E1 and rAd5-E2 vaccines were tested singly or in combination to assess levels of protective immunity to COPV challenge. Subcutaneous immunization with either one or two doses of rAd5 expressing the COPV E1 and E2 genes could protect > 90% of challenged dogs from wart formation. In contrast, all eight dogs immunized with rAd5-HPV-16 L1 developed papillomas at multiple sites. Protection was accompanied by significant IFN-gamma responses to COPV E1 and E2 peptides. Partial protection was conferred by two immunizations with either rAd5-E1 (6 of 9 protected) or rAd5-E2 (8 of 9 protected). These data indicate that rAd5 expressing papillomavirus E1 and E2 proteins can induce strong protective responses even in outbred populations under practical immunization conditions.
Collapse
Affiliation(s)
- Kimberly B Johnston
- Vaccines and Biologics Research, Merck Research Laboratories, West Point, PA 19486, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Accardi L, Donà MG, Di Bonito P, Giorgi C. Intracellular anti-E7 human antibodies in single-chain format inhibit proliferation of HPV16-positive cervical carcinoma cells. Int J Cancer 2005; 116:564-70. [PMID: 15818614 DOI: 10.1002/ijc.21052] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The E7 tumor antigen of human papillomavirus type 16 (HPV16) is a validated target for immunodiagnosis and immunotherapy of HPV-associated cervical cancer. Anti-HPV16 E7 antibodies in scFv format were isolated from a human antibody phage display library and characterized. With the aim of interfering with the oncogenic activity of E7 protein, the most reactive of the selected antibody fragments was expressed by eukaryotic vectors in different compartments of the HPV16-positive cervical carcinoma SiHa cell line. The intracellular antibodies (intrabodies) were tested for their ability of inhibiting cell proliferation. A significant inhibition was obtained targeting the intrabodies to the nuclear and secretory compartments whereas no significant effect was observed in case of cytoplasmic localization. Inhibition was highly specific as no antiproliferative effect was obtained either with the E7-specific intrabodies in HPV-negative cells nor with irrelevant intrabodies in SiHa cells.
Collapse
Affiliation(s)
- Luisa Accardi
- Department of Infectious, Parasitic and Immunomediated Diseases, Istituto Superiore di Sanità, Rome, Italy.
| | | | | | | |
Collapse
|
26
|
Insinga RP, Dasbach EJ, Elbasha EH. Assessing the annual economic burden of preventing and treating anogenital human papillomavirus-related disease in the US: analytic framework and review of the literature. PHARMACOECONOMICS 2005; 23:1107-22. [PMID: 16277547 DOI: 10.2165/00019053-200523110-00004] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The anogenital human papillomavirus (HPV) is estimated to be the most commonly occurring sexually transmitted infection in the US. Comprehensive estimates of the annual economic burden associated with the prevention and treatment of anogenital HPV-related disease in the US population are currently unavailable. The purpose of this paper is to (i) outline an analytic framework from which to estimate the annual economic burden of preventing and treating anogenital HPV-related disease in the US; (ii) review available US literature concerning the annual economic burden of HPV; and (iii) highlight gaps in current knowledge where further study is particularly warranted. Among eight US studies identified that describe the annual economic burden pertaining to one or more aspects of anogenital HPV-related disease, three met the review eligibility criteria (published between 1990 and 2004, examined multiple facets of annual anogenital HPV-related economic burden, and clearly articulated the data and methods used in the estimation process). All costs were adjusted to 2004 US dollars. Estimates of the annual direct medical costs associated with cervical cancer were comparable across studies (range 300-400 million US dollars). In contrast, there was a wide range across studies for estimates of the annual direct medical costs associated with cervical intraepithelial neoplasia (range 700 million US dollars-2.3 billion US dollars). Only one study reported direct medical costs for anogenital warts (200 million US dollars) and routine cervical cancer screening (2.3 billion US dollars). No studies examined direct medical costs attributable to HPV-related anal, penile, vaginal or vulvar cancers, or the work and productivity losses resulting from time spent receiving medical care, morbidity or mortality. Current economic burden estimates would suggest annual direct medical costs associated with the prevention and treatment of anogenital warts and cervical HPV-related disease of at least 4 billion US dollars. This figure would likely rise to at least 5 billion US dollars per year if direct medical costs associated with other disease entities caused by the sexual transmission of HPV were included, with further additions to the economic burden totalling in the billions of dollars if work and productivity losses were incorporated, a research priority for future studies.
Collapse
Affiliation(s)
- Ralph P Insinga
- Department of Health Economic Statistics, Merck Research Laboratories, Blue Bell, PA 19422, USA.
| | | | | |
Collapse
|
27
|
Abstract
Traditional successful antiviral vaccines have relied mostly on live-attenuated viruses. Live-attenuated HIV vaccine candidates are not ideal as they pose risks of reversion, recombination or mutations. Other current HIV vaccine candidates have difficulties generating broadly effective neutralising antibodies and cytotoxic T cell immune responses to primary HIV isolates. Virus-like-particles (VLPs) have been demonstrated to be safe to administer to animals and human patients as well as being potent and efficient stimulators of cellular and humoral immune responses. Therefore, VLPs are being considered as possible HIV vaccines. Chimeric HIV-1 VLPs constructed with either HIV or SIV capsid protein plus HIV immune epitopes and immuno-stimulatory molecules have further improved on early VLP designs, leading to enhanced immune stimulation. The administration of VLP vaccines via mucosal surfaces has also emerged as a promising strategy with which to elicit mucosal and systemic humoral and cellular immune responses. Additionally, new information on antigen processing and the presentation of particulate antigens by dendritic cells (DCs) has created new strategies for improved VLP vaccine candidates. This paper reviews the field of HIV-1 VLP vaccine development, focusing on recent studies that will likely uncover promising prospects for new HIV vaccines.
Collapse
Affiliation(s)
- Linh X Doan
- Molecular Surgeon Research Center, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | |
Collapse
|
28
|
Abstract
There is currently sufficient evidence to conclude that human papillomavirus (HPV) plays a role in the pathogenesis of a distinct subset of head and neck squamous cell cancers (HNSCC), particularly tonsillar cancers. There is a strong and consistent association between high-risk HPV types, specifically HPV16, a known human carcinogen, and these distinctive oropharyngeal cancers with molecular characteristics indicative of viral oncogene function. Risk for HPV-HNSCC is increased by certain sexual behaviors after consideration of alcohol and tobacco exposure, consistent with an extensive literature that has established HPV infection as a sexually transmitted disease. Furthermore, exposure to HPV16 has been associated with increased risk for subsequent development of oropharyngeal cancer. Prophylactic and therapeutic vaccines targeted against the viral capsid components and oncoproteins will provide the ultimate evidence for a role for HPV in HNSCC, if demonstrated to be effective in the prevention or therapy of this disease. It is time for clinician scientists to translate knowledge of this newly recognized disease entity into potential applications for the prevention, detection, and treatment of HPV-HNSCC.
Collapse
Affiliation(s)
- Maura L Gillison
- Department of Medical Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins School of Medicine, Bunting-Blaustein Cancer Research Building, 1650 Orleans Street, Baltimore, MD 21231, USA
| |
Collapse
|
29
|
Smyth LJC, Van Poelgeest MIE, Davidson EJ, Kwappenberg KMC, Burt D, Sehr P, Pawlita M, Man S, Hickling JK, Fiander AN, Tristram A, Kitchener HC, Offringa R, Stern PL, Van Der Burg SH. Immunological responses in women with human papillomavirus type 16 (HPV-16)-associated anogenital intraepithelial neoplasia induced by heterologous prime-boost HPV-16 oncogene vaccination. Clin Cancer Res 2004; 10:2954-61. [PMID: 15131030 DOI: 10.1158/1078-0432.ccr-03-0703] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The purpose is to study the immunogenicity of heterologous prime-boost human papillomavirus (HPV) oncogene vaccination in patients with anogenital intraepithelial neoplasia (AGIN). EXPERIMENTAL DESIGN Twenty-nine women with high-grade AGIN received three i.m. doses of TA-CIN (HPV-16 L2/E6/E7 protein) at four weekly intervals followed by a single dermal scarification of vaccinia HPV-16/18 E6/E7 and were followed up for 12 weeks. Immunity to HPV-16 was assessed by lymphoproliferation, IFN-gamma enzyme-linked immunospot (ELISPOT), and ELISA. RESULTS The patient group significantly responded to TA-CIN and not to the control antigen HPV-6 L2/E7 at all postvaccination time points when compared with baseline responses (P < or = 0.05). Ten of the patients showed at least a 3-fold increase in TA-CIN-specific proliferation at one or more time points after vaccination. Comparison of stimulation with HPV-16 E6- or E7-GST fusion proteins showed that proliferative responses were biased to HPV-16 E6. This bias was also seen by IFN-gamma ELISPOT using overlapping peptides, with HPV-16 E6- or E7-specific T cells being detected in 9 and 2 patients, respectively. In addition, vaccination resulted in the induction of antibodies against the HPV-16 oncoproteins. Of the 6 clinical responders, 2 patients showed both a proliferative TA-CIN-specific response and an E6-specific IFN-gamma response, whereas 3 other patients displayed E6-specific reactivity only. Stable disease was recorded in 19 patients, 8 of whom showed a concomitant TA-CIN-specific proliferative and/or E6-specific T-cell response. Of the 4 progressors, 2 failed to make a T-cell response and 2 responded by either proliferation or E6 ELISPOT alone. CONCLUSIONS The prime-boost regimen is immunogenic in AGIN patients (humoral and cellular immunity), but there is no simple relationship between induction of systemic HPV-16-specific immunity and clinical outcome. Other factors that may play a role in the eradication of long-term established AGIN lesions need to be determined to identify the patient group that would benefit from immunotherapy with the vaccines used in this study.
Collapse
Affiliation(s)
- Lucy J C Smyth
- Cancer Research UK Immunology Group, Paterson Institute for Cancer Research, Christie Hospital, Manchester, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
A subset of human papillomaviruses (HPVs) promote anogenital malignancy, including cervical cancer, and prevention and treatment strategies that reflect the causal role of HPV are being developed. Vaccines based on HPV virus-like particles induce genotype-specific virus-neutralizing antibody and prevent infection with HPV. Persistent papillomavirus infection is required for the development of papillomavirus-associated cancer and, therefore, therapeutic vaccines are being developed to eliminate established papillomavirus infection. Such vaccines test principles for the growing field of tumour-antigen-specific immunotherapy. This article reviews progress in the field and draws conclusions for the development of future prophylactic and therapeutic viral vaccines.
Collapse
Affiliation(s)
- Ian H Frazer
- Centre for Immunology and Cancer Research, The University of Queensland, Princess Alexandra Hospital, Ipswich Road, Woolloongabba, Queensland 4102, Australia.
| |
Collapse
|