1
|
Braun MR, Moore AC, Lindbloom JD, Hodgson KA, Dora EG, Tucker SN. Elimination of Human Papillomavirus 16-Positive Tumors by a Mucosal rAd5 Therapeutic Vaccination in a Pre-Clinical Murine Study. Vaccines (Basel) 2024; 12:955. [PMID: 39339987 PMCID: PMC11435741 DOI: 10.3390/vaccines12090955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/07/2024] [Accepted: 08/13/2024] [Indexed: 09/30/2024] Open
Abstract
Therapeutic vaccination can harness the body's cellular immune system to target and destroy cancerous cells. Several treatment options are available to eliminate pre-cancerous and cancerous lesions caused by human papillomaviruses (HPV), but may not result in a long-term cure. Therapeutic vaccination may offer an effective, durable, and minimally intrusive alternative. We developed mucosally delivered, recombinant, non-replicating human adenovirus type 5 (rAd5)-vectored vaccines that encode HPV16's oncogenic proteins E6 and E7 alongside a molecular dsRNA adjuvant. The induction of antigen-specific T cells and the therapeutic efficacy of rAd5 were evaluated in a mouse model of HPV tumorigenesis where E6E7-transformed cells, TC-1, were implanted subcutaneously in C57BL/6 mice. After tumor growth, mice were treated intranasally with rAd5 vaccines expressing the wildtype form of E6E7 (rAd5-16/E6E7Wt) in combination with an anti-PD-1 antibody or isotype control. Animals treated with rAd5-16/E6E7Wt with and without anti-PD-1 had significant reductions in tumor volume and increased survival compared to controls. Further, animals treated with rAd5-16/E6E7Wt had increased CD4+ and CD8+ tumor-infiltrating lymphocytes (TILs) and produced a cytotoxic tumor microenvironment. In a second study, the immunogenicity of a non-transformative form of E6E7 (rAd5-16/E6E7Mu) and a vaccine encoding predicted T cell epitopes of E6E7 (rAd5-16/E6E7epi) were evaluated. These vaccines elicited significant reductions in TC-1 tumor volume and increased survival of animals. Antigen-specific CD8+ T effector memory cells were observed in the animals treated with E6E7-encoding rAd5, but not in the rAd5-empty group. The work described here demonstrates that this mucosal vaccination can be used therapeutically to elicit specific cellular immunity and further identifies a clinical candidate with great potential for the treatment and prevention of human cervical cancer.
Collapse
Affiliation(s)
- Molly R Braun
- Vaxart Inc., 170 Harbor Way Suite 300, South San Francisco, CA 94080, USA
| | - Anne C Moore
- Vaxart Inc., 170 Harbor Way Suite 300, South San Francisco, CA 94080, USA
- School of Biochemistry and Cell Biology, University College Cork, T12 XF62 Cork, Ireland
- National Institute of Bioprocessing Research and Training, A94 X099 Dublin, Ireland
| | | | | | - Emery G Dora
- Vaxart Inc., 170 Harbor Way Suite 300, South San Francisco, CA 94080, USA
| | - Sean N Tucker
- Vaxart Inc., 170 Harbor Way Suite 300, South San Francisco, CA 94080, USA
| |
Collapse
|
2
|
Peng S, Ferrall L, Gaillard S, Wang C, Chi WY, Huang CH, Roden RBS, Wu TC, Chang YN, Hung CF. Development of DNA Vaccine Targeting E6 and E7 Proteins of Human Papillomavirus 16 (HPV16) and HPV18 for Immunotherapy in Combination with Recombinant Vaccinia Boost and PD-1 Antibody. mBio 2021; 12:e03224-20. [PMID: 33468698 PMCID: PMC7845631 DOI: 10.1128/mbio.03224-20] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy for cervical cancer should target high-risk human papillomavirus types 16 and 18, which cause 50% and 20% of cervical cancers, respectively. Here, we describe the construction and characterization of the pBI-11 DNA vaccine via the addition of codon-optimized human papillomavirus 18 (HPV18) E7 and HPV16 and 18 E6 genes to the HPV16 E7-targeted DNA vaccine pNGVL4a-SigE7(detox)HSP70 (DNA vaccine pBI-1). Codon optimization of the HPV16/18 E6/E7 genes in pBI-11 improved fusion protein expression compared to that in DNA vaccine pBI-10.1 that utilized the native viral sequences fused 3' to a signal sequence and 5' to the HSP70 gene of Mycobacterium tuberculosis Intramuscular vaccination of mice with pBI-11 DNA better induced HPV antigen-specific CD8+ T cell immune responses than pBI-10.1 DNA. Furthermore, intramuscular vaccination with pBI-11 DNA generated stronger therapeutic responses for C57BL/6 mice bearing HPV16 E6/E7-expressing TC-1 tumors. The HPV16/18 antigen-specific T cell-mediated immune responses generated by pBI-11 DNA vaccination were further enhanced by boosting with tissue-antigen HPV vaccine (TA-HPV). Combination of the pBI-11 DNA and TA-HPV boost vaccination with PD-1 antibody blockade significantly improved the control of TC-1 tumors and extended the survival of the mice. Finally, repeat vaccination with clinical-grade pBI-11 with or without clinical-grade TA-HPV was well tolerated in vaccinated mice. These preclinical studies suggest that the pBI-11 DNA vaccine may be used with TA-HPV in a heterologous prime-boost strategy to enhance HPV 16/18 E6/E7-specific CD8+ T cell responses, either alone or in combination with immune checkpoint blockade, to control HPV16/18-associated tumors. Our data serve as an important foundation for future clinical translation.IMPORTANCE Persistent expression of high-risk human papillomavirus (HPV) E6 and E7 is an obligate driver for several human malignancies, including cervical cancer, wherein HPV16 and HPV18 are the most common types. PD-1 antibody immunotherapy helps a subset of cervical cancer patients, and its efficacy might be improved by combination with active vaccination against E6 and/or E7. For patients with HPV16+ cervical intraepithelial neoplasia grade 2/3 (CIN2/3), the precursor of cervical cancer, intramuscular vaccination with a DNA vaccine targeting HPV16 E7 and then a recombinant vaccinia virus expressing HPV16/18 E6-E7 fusion proteins (TA-HPV) was safe, and half of the patients cleared their lesions in a small study (NCT00788164). Here, we sought to improve upon this therapeutic approach by developing a new DNA vaccine that targets E6 and E7 of HPV16 and HPV18 for administration prior to a TA-HPV booster vaccination and for application against cervical cancer in combination with a PD-1-blocking antibody.
Collapse
Affiliation(s)
- Shiwen Peng
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Louise Ferrall
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Stephanie Gaillard
- Department of Oncology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Obstetrics and Gynecology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Chenguang Wang
- Department of Oncology Biostatistics, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Wei-Yu Chi
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Chuan-Hsiang Huang
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Richard B S Roden
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - T-C Wu
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Obstetrics and Gynecology, The Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Chien-Fu Hung
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Obstetrics and Gynecology, The Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Panahi HA, Bolhassani A, Javadi G, Noormohammadi Z, Agi E. Development of multiepitope therapeutic vaccines against the most prevalent high-risk human papillomaviruses. Immunotherapy 2020; 12:459-479. [DOI: 10.2217/imt-2019-0196] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aim: Our goal was the development of DNA- or peptide-based multiepitope vaccines targeting HPV E7, E6 and E5 oncoproteins in tumor mouse model. Materials & methods: After designing the multiepitope E7, E6 and E5 constructs from four types of high risk HPVs (16, 18, 31 & 45) using bioinformatics tools, mice vaccination was performed by different homologous and heterologous modalities in a prophylactic setting. Then, anti-tumor effects of the best prophylactic strategies were studied in a therapeutic setting. Results: In both prophylactic and therapeutic experiments, groups receiving homologous E7+E6+E5 polypeptide, and heterologous E7+E6+E5 DNA prime/polypeptide boost were successful in complete rejection of tumors. Conclusion: The designed multiepitope constructs can be considered as promising candidates to develop effective therapeutic HPV vaccines.
Collapse
Affiliation(s)
- Heidar Ali Panahi
- Department of Hepatitis & AIDS, Pasteur Institute of Iran, Tehran, Iran
- Department of Biology, School of Basic Sciences, Science & Research Branch, Islamic Azad University, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis & AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Gholamreza Javadi
- Department of Biology, School of Basic Sciences, Science & Research Branch, Islamic Azad University, Tehran, Iran
| | - Zahra Noormohammadi
- Department of Biology, School of Basic Sciences, Science & Research Branch, Islamic Azad University, Tehran, Iran
| | - Elnaz Agi
- Iranian Comprehensive Hemophilia Care Center, Tehran, Iran
| |
Collapse
|
4
|
Ma Y, Yang A, Peng S, Qiu J, Farmer E, Hung CF, Wu TC. Characterization of HPV18 E6-specific T cell responses and establishment of HPV18 E6-expressing tumor model. Vaccine 2017; 35:3850-3858. [PMID: 28599791 DOI: 10.1016/j.vaccine.2017.05.081] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 05/02/2017] [Accepted: 05/26/2017] [Indexed: 02/02/2023]
Abstract
Human papillomavirus (HPV) has been identified as the primary etiologic factor of cervical cancer, and subsets of anogenital and oropharyngeal cancers. HPV18 is the second most prevalent high-risk HPV type after HPV16. Furthermore, HPV18 is responsible for approximately 12% of cervical squamous cell carcinoma and 37% of cervical adenocarcinoma cases worldwide. In this study, we aimed to characterize the HPV18-E6-specific epitope and establish an HPV18 animal tumor model to evaluate the E6-specific immune response induced by our DNA vaccine. We vaccinated naïve C57BL/6 mice with a prototype DNA vaccine, pcDNA3-HPV18-E6, via intramuscular injection followed by electroporation, and analyzed the E6-specific CD8+ T cell responses by flow cytometry using a reported T cell epitope. We then characterized the MHC restriction element for the characterized HPV18-E6 epitope. Additionally, we generated an HPV18-E6-expressing tumor cell line to study the antitumor effect mediated by E6-specific immunity. We observed a robust HPV18-E6aa67-75 peptide-specific CD8+ T cell response after vaccination with pcDNA3-HPV18-E6. Further characterization demonstrated that this epitope was mainly restricted by H-2Kb, but was also weakly presented by HLA-A∗0201, as previously reported. We observed that vaccination with pcDNA3-HPV18-E6 significantly inhibited the growth of HPV18-E6-expressing tumor cells, TC-1/HPV18-E6, in mice. An antibody depletion study demonstrated that both CD4+ and CD8+ T cells are necessary for the observed antitumor immunity. The characterization of HPV18-E6-specific T cell responses and the establishment of HPV18-E6-expressing tumor cell line provide infrastructures for further development of HPV18-E6 targeted immunotherapy.
Collapse
Affiliation(s)
- Ying Ma
- Department of Gynecology and Obstetrics, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong Province, China; Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, United States.
| | - Andrew Yang
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, United States.
| | - Shiwen Peng
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, United States.
| | - Jin Qiu
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China; Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, United States.
| | - Emily Farmer
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, United States.
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, United States; Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD, United States.
| | - T-C Wu
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, United States; Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD, United States; Department of Obstetrics and Gynecology, Johns Hopkins Medical Institutions, Baltimore, MD, United States; Department of Molecular Microbiology and Immunology, Johns Hopkins Medical Institutions, Baltimore, MD, United States.
| |
Collapse
|
5
|
DEL RÍO-OSPINA L, SOTO-DE LEÓN SC, CAMARGO M, SÁNCHEZ R, MORENO-PÉREZ DA, PÉREZ-PRADOS A, PATARROYO ME, PATARROYO MA. Multiple high-risk HPV genotypes are grouped by type and are associated with viral load and risk factors. Epidemiol Infect 2017; 145:1479-1490. [PMID: 28185605 PMCID: PMC9203302 DOI: 10.1017/s0950268817000188] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 12/20/2016] [Accepted: 01/10/2017] [Indexed: 11/07/2022] Open
Abstract
Investigating whether high-risk human papillomavirus (HR-HPV) types tend to become grouped in a particular way and whether factors are associated with such grouping is important for measuring the real impact of vaccination. In total, 219 women proving positive for HPV as detected by real-time PCR were included in the study. Each sample was analysed for detecting and quantifying six viral types and the hydroxymethylbilane synthase gene. Multiple correspondence analysis led to determining grouping patterns for six HR-HPV types and simultaneous association with multiple variables and whether viral load was related to the coexistence of other viral types. Two grouping profiles were identified: the first included HPV-16 and HPV-45 and the second profile was represented by HPV-31, HPV-33 and HPV-58. Variables such as origin, contraceptive method, births and pregnancies, educational level, healthcare affiliation regime, atypical squamous cells of undetermined significance and viral load were associated with these grouping profiles. Different socio-demographic characteristics were found when coinfection occurred by phylogenetically related HPV types and when coinfection was due to non-related types. Biological characteristics, the number of viral copies, temporality regarding acquiring infection and competition between viral types could influence the configuration of grouping patterns. Characteristics related to women and HPV, influence such interactions between coexisting HPV types reflecting the importance of their evaluation.
Collapse
Affiliation(s)
- L. DEL RÍO-OSPINA
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá D.C., Colombia
- School of Medicine, Universidad Nacional de Colombia, Bogotá D.C., Colombia
| | - S. C. SOTO-DE LEÓN
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá D.C., Colombia
- Universidad de Ciencias Aplicadas y Ambientales (UDCA), Bogotá D.C., Colombia
| | - M. CAMARGO
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá D.C., Colombia
- PhD Programme in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá D.C., Colombia
| | - R. SÁNCHEZ
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá D.C., Colombia
- School of Medicine, Universidad Nacional de Colombia, Bogotá D.C., Colombia
| | - D. A. MORENO-PÉREZ
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá D.C., Colombia
- PhD Programme in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá D.C., Colombia
| | - A. PÉREZ-PRADOS
- Mathematics Department, Universidad Pública de Navarra, Pamplona, Spain
| | - M. E. PATARROYO
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá D.C., Colombia
- School of Medicine, Universidad Nacional de Colombia, Bogotá D.C., Colombia
| | - M. A. PATARROYO
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá D.C., Colombia
- School of Medicine and Health Sciences, Universidad del Rosario, Bogotá D.C., Colombia
| |
Collapse
|
6
|
Nunes CT, Miners KL, Dolton G, Pepper C, Fegan C, Mason MD, Man S. A novel tumor antigen derived from enhanced degradation of bax protein in human cancers. Cancer Res 2011; 71:5435-44. [PMID: 21697278 DOI: 10.1158/0008-5472.can-11-0393] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancer cells frequently exhibit defects in apoptosis, which contribute to increased survival and chemotherapeutic resistance. For example, genetic mutations or abnormal proteasomal degradation can reduce expression of Bax which limits apoptosis. In cancers where abnormal proteasomal degradation of Bax occurs, we hypothesized that Bax peptides that bind to human leukocyte antigen (HLA) class I molecules would be generated for presentation to CD8(+) T cells. To test this hypothesis, we generated T cells against pooled Bax peptides, using the blood of healthy human donors. Although T-cell responses were of low frequency (0.15%), a CD8(+) T-cell clone (KSIVB17) was isolated that optimally recognized Bax(136-144) peptide (IMGWTLDFL) presented by HLA-A*0201. KSIVB17 was able to recognize and kill a variety of HLA-matched cancer cells including primary tumor cells from chronic lymphocytic leukemia (CLL). No reactivity was seen against HLA-matched, nontransformed cells such as PHA blasts and skin fibroblasts. Furthermore, KSIVB17 reactivity corresponded with the proteasomal degradation patterns of Bax protein observed in cancer cells. Taken together, our findings suggest a new concept for tumor antigens based on regulatory proteins that are ubiquitously expressed in normal cells, but that have abnormally enhanced degradation in cancer cells. Bax degradation products offer candidate immune antigens in cancers such as CLL in which increased Bax degradation correlates with poor clinical prognosis.
Collapse
Affiliation(s)
- Cláudia Trindade Nunes
- Departments of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Cardiff, Wales, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
7
|
Zhao L, Liu B, Ren J, Feng J, Pang Z, Gao J, Zhang H, Tan W, Tian H, Ruan L. Immunogenicity in mice and rhesus monkeys vaccinated with recombinant vaccinia virus expressing bivalent E7E6 fusion proteins from human papillomavirus types 16 and 18. Virol J 2011; 8:302. [PMID: 21672263 PMCID: PMC3135557 DOI: 10.1186/1743-422x-8-302] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Accepted: 06/15/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Persistent infection with high-risk human papillomavirus (HPV) is a predominant cause of cervical cancer, and HPV16 and HPV18 occur in 50% and 20% of cervical cancer cases, respectively. The viral oncogenes E6 and E7 are constitutively expressed by HPV-associated tumour cells and can therefore be used as target antigens for immunotherapy. In this study, we constructed a recombinant vaccinia virus co-expressing the HPV16/18 E7E6 fusion proteins (rVVJ16/18E7E6) for use as a therapeutic vaccine for the treatment of HPV16⁺ and HPV18⁺ cancers. METHODS We constructed a bivalent recombinant vaccinia virus expressing modified E7E6 fusion proteins of HPV type 16 and 18 (rVVJ16/18E7E6) based on the vaccinia virus Tiantan strain. We then defined the cellular immune responses to the virus in mice and rhesus monkeys and assessed antitumour efficacy of these responses in mice using the TC-1 tumour challenge model. RESULTS Our data demonstrated that rVVJ16/18E7E6 was able to elicit varying levels of CD8⁺ T cell immune responses and lysis of target cells in mice in response to peptides HPV16E7₄₉₋₅₇ and HPV18E6₆₇₋₇₅. Furthermore, the virus was also able to induce anti-tumour responses in the HPV16⁺ TC-1 tumour challenge model, including partial protection (30-40%) and delayed tumour appearance. In addition, the virus was able to induce immune responses in rhesus monkeys. CONCLUSIONS The recombinant vaccinia virus rVVJ16/18E7E6 can generate clear and significant cellular immunity in both mice and rhesus monkeys. These data provide a basis for the use of this recombinant virus as a potential vaccine candidate for further study.
Collapse
Affiliation(s)
- Li Zhao
- State Key Laboratory for Molecular Virology and Genetic Engineering, Biotech Center for Viral Disease Emergency, National Institute for Viral Disease Control and Prevention, China CDC, No.155 Changbailu, Changpingqu, Beijing, 102206, China
| | - Binlei Liu
- Department of Immunology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Jiao Ren
- State Key Laboratory for Molecular Virology and Genetic Engineering, Biotech Center for Viral Disease Emergency, National Institute for Viral Disease Control and Prevention, China CDC, No.155 Changbailu, Changpingqu, Beijing, 102206, China
| | - Jing Feng
- State Key Laboratory for Molecular Virology and Genetic Engineering, Biotech Center for Viral Disease Emergency, National Institute for Viral Disease Control and Prevention, China CDC, No.155 Changbailu, Changpingqu, Beijing, 102206, China
| | - Zheng Pang
- State Key Laboratory for Molecular Virology and Genetic Engineering, Biotech Center for Viral Disease Emergency, National Institute for Viral Disease Control and Prevention, China CDC, No.155 Changbailu, Changpingqu, Beijing, 102206, China
| | - Jian Gao
- State Key Laboratory for Molecular Virology and Genetic Engineering, Biotech Center for Viral Disease Emergency, National Institute for Viral Disease Control and Prevention, China CDC, No.155 Changbailu, Changpingqu, Beijing, 102206, China
| | - Hui Zhang
- State Key Laboratory for Molecular Virology and Genetic Engineering, Biotech Center for Viral Disease Emergency, National Institute for Viral Disease Control and Prevention, China CDC, No.155 Changbailu, Changpingqu, Beijing, 102206, China
| | - Wenjie Tan
- State Key Laboratory for Molecular Virology and Genetic Engineering, Biotech Center for Viral Disease Emergency, National Institute for Viral Disease Control and Prevention, China CDC, No.155 Changbailu, Changpingqu, Beijing, 102206, China
| | - Houwen Tian
- State Key Laboratory for Molecular Virology and Genetic Engineering, Biotech Center for Viral Disease Emergency, National Institute for Viral Disease Control and Prevention, China CDC, No.155 Changbailu, Changpingqu, Beijing, 102206, China
| | - Li Ruan
- State Key Laboratory for Molecular Virology and Genetic Engineering, Biotech Center for Viral Disease Emergency, National Institute for Viral Disease Control and Prevention, China CDC, No.155 Changbailu, Changpingqu, Beijing, 102206, China
| |
Collapse
|
8
|
Matijevic M, Hedley ML, Urban RG, Chicz RM, Lajoie C, Luby TM. Immunization with a poly (lactide co-glycolide) encapsulated plasmid DNA expressing antigenic regions of HPV 16 and 18 results in an increase in the precursor frequency of T cells that respond to epitopes from HPV 16, 18, 6 and 11. Cell Immunol 2011; 270:62-9. [PMID: 21550027 PMCID: PMC7094646 DOI: 10.1016/j.cellimm.2011.04.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Revised: 03/24/2011] [Accepted: 04/14/2011] [Indexed: 01/08/2023]
Abstract
A phase II trial was conducted in subjects with human papillomavirus (HPV) associated high-grade cervical dysplasia testing the safety and efficacy of a microparticle encapsulated pDNA vaccine. Amolimogene expresses T cell epitopes from E6 and E7 proteins of HPV types 16 and 18. An analysis was performed on a subset of HLA-A2+ subjects to test whether CD8+ T cells specific to HPV 16, 18, 6 and 11 were increased in response to amolimogene immunization. Of the 21 subjects receiving amolimogene, 11 had elevated CD8+ T cell responses to HPV 16 and/or 18 peptides and seven of these also had increases to corresponding HPV 6 and/or 11 peptides. In addition, T cells primed and expanded in vitro with an HPV 18 peptide demonstrated cross-reactivity to the corresponding HPV 11 peptide. These data demonstrate that treatment with amolimogene elicits T cell responses to HPV 16, 18, 6 and 11.
Collapse
|
9
|
Xi LF, Hughes JP, Edelstein ZR, Kiviat NB, Koutsky LA, Mao C, Ho J, Schiffman M. Human Papillomavirus (HPV) type 16 and type 18 DNA Loads at Baseline and Persistence of Type-Specific Infection during a 2-year follow-up. J Infect Dis 2009; 200:1789-97. [PMID: 19848609 DOI: 10.1086/647993] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Studies of viral load-associated persistence of human papillomavirus (HPV) infection are rare, with inconsistent results reported. METHODS The study subjects were 741 and 289 women who were positive for HPV type 16 (HPV-16) and HPV type 18 (HPV-18), respectively, at the time of enrollment into in the ASCUS-LSIL (Atypical Squamous Cells of Undetermined Significance-Low-Grade Squamous Intraepithelial Lesion) Triage Study and who returned 1 or more times for HPV testing during a biannual 2-year follow-up. The numbers of HPV-16 and HPV-18 copies per nanogram of cellular DNA at baseline were measured by use of real-time polymerase chain reaction. RESULTS Women with, compared with women without, persistent infection at month 6 of follow-up had a higher viral load at enrollment (P< .001, for HPV-16; P=.01, for HPV-18). The association of each 1-log(10) increase in viral load with persistence of HPV-16 or HPV-18 during the first 6 months of the study was statistically significant among women with multiple HPV types at enrollment (for HPV-16: odds ratio [OR], 1.53 [95% confidence interval {CI}, 1.29-1.82]; for HPV-18: OR, 1.35 [95% CI, 1.09-1.68]) but not among women with monotype infections (in tests assessing the interaction between viral load and coinfection, P=.002 for HPV-16 and P=.34 for HPV-18). Among women who continued to have positive results at month 6, 12, or 18, persistence of infection for another 6 months was unassociated with the viral load at baseline. CONCLUSION Prevalent infection with a higher viral load of HPV-16 or HPV-18 was associated with short- but not long-term persistence.
Collapse
Affiliation(s)
- Long Fu Xi
- Department of Pathology, School of Medicine, University of Washington, Seattle, USA.
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Xi LF, Edelstein ZR, Meyers C, Ho J, Cherne SL, Schiffman M. Human papillomavirus types 16 and 18 DNA load in relation to coexistence of other types, particularly those in the same species. Cancer Epidemiol Biomarkers Prev 2009; 18:2507-12. [PMID: 19690188 PMCID: PMC2745080 DOI: 10.1158/1055-9965.epi-09-0482] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Infection with multiple human papillomavirus (HPV) types is common. However, it is unknown whether viral DNA load is related to the coexistence of other types. METHODS Study subjects were 802 and 303 women who were positive for HPV16 and HPV18, respectively, at enrollment into the Atypical Squamous Cells of Undetermined Significance and Low-Grade Squamous Intraepithelial Lesion Triage Study. HPV16 and HPV18 E7 copies per nanogram of cellular DNA in cervical swab samples were measured by real-time PCR in triplicate. RESULTS Concurrent coinfection was common in this population of women with minor cervical lesions; multiple HPV types were detected in 573 (71.4%) of 802 HPV16-positive women and 227 (74.9%) of 303 HPV18-positive women. The adjusted odds ratio associating coinfection with per 1 log unit increase in HPV16 DNA load was 0.78 (95% confidence interval, 0.68-0.89); it was 0.64 (95% confidence interval, 0.52-0.79) for a similar analysis of HPV18 DNA load. Women with, compared with without, coinfection of A9 species types possessed a significantly lower HPV16 DNA load (P < 0.001), whereas women with, compared with without, coinfection of A7 species types possessed a significantly lower HPV18 DNA load (P = 0.001). A trend of decrease in HPV16 DNA load with increasing number of the coexisting non-HPV16 A9 species types was statistically significant (P(trend) = 0.001). CONCLUSION Coinfection with other types was associated with lower HPV16 and HPV18 DNA load. The extent of reduction was correlated to phylogenetic distance of the coexisting types to HPV16 and HPV18, respectively.
Collapse
Affiliation(s)
- Long Fu Xi
- Department of Pathology, School of Medicine, University of Washington, Seattle, Washington, USA.
| | | | | | | | | | | |
Collapse
|
11
|
Suneetha PV, Schlaphoff V, Wang C, Stegmann KA, Fytili P, Sarin SK, Manns MP, Cornberg M, Wedemeyer H. Effect of peptide pools on effector functions of antigen-specific CD8+ T cells. J Immunol Methods 2009; 342:33-48. [PMID: 19135447 DOI: 10.1016/j.jim.2008.11.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2008] [Revised: 10/24/2008] [Accepted: 11/24/2008] [Indexed: 01/07/2023]
Abstract
Peptide pools are routinely used to study antigen specific T cell responses, both in epitope discovery as well as immune monitoring. However, optimal assay conditions such as concentration of peptides or the best possible number of peptides per pool have not been defined. Thus, we examined whether different peptide concentrations or varying number of peptides per pool influence effector functions of antigen-specific human T-cells. PBMC isolated from HLA-A2-positive individuals with known responses to frequently recognised dominant CD8+ T cell epitopes derived from four different viruses (influenza virus, CMV, EBV, or HCV) were studied. PBMC were cultured with one of these HLA-A2 restricted peptides and varying concentrations of overlapping peptide pools derived from unrelated viruses specific for the hepatitis D and E viruses, the subjects have not been exposed to. Importantly, unrelated peptide pools inhibited the proliferation of IV-M1(58), CMVpp65(495-503), EBV-BMLF(1259-267) and HCV NS3(1073)-specific CD8 T-cells in a dose dependent manner. Similarly, an increase in the number of peptides per pool also impaired antigen specific CD8+ T cell proliferation. In contrast, secretion of cytokines such as IL-2, IL-10, IFN-gamma, TNF-alpha or IP-10 as well as cytotoxicity was not affected by these unrelated peptide pools. The inhibition of proliferation could be restored by blocking PD-1/PDL-1 interaction and was not dependent on DMSO when DMSO concentration was <or=0.5%. Thus, peptide-specific CD8 T-cell proliferation but not cytokine production may be largely underestimated when using a peptide pool which warrants caution in immunomonitoring during clinical trials and in epitope discovery studies.
Collapse
|
12
|
Cellular immunity induced by a novel HPV18 DNA vaccine encoding an E6/E7 fusion consensus protein in mice and rhesus macaques. Vaccine 2008; 26:5210-5. [PMID: 18455277 DOI: 10.1016/j.vaccine.2008.03.069] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Human papilloma-virus (HPV) infection is the major cause of cervical cancer. HPV18 is the most prevalent high-risk HPV after type 16 that accounts for the largest number of cervical cancer cases worldwide. Currently, although prophylactic vaccines have been developed, there is still an urgent need to develop therapeutic HPV vaccines for targeting tumors post-infection. In this study, we utilize a novel multi-phase strategy for HPV18 antigen development with the goal of increasing anti-HPV18 cellular immunity. Our data show that this construct can induce strong cellular immune responses against HPV18 E6 and E7 antigens in a murine model. Moreover, when applied to rhesus monkeys, this construct is also able to elicit cellular immunity. These data suggest such DNA immunogens are candidates for further study in the eventual context of immunotherapy for HPV-associated cancers.
Collapse
|