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

Shiwen Peng, Louise Ferrall, Stephanie Gaillard, Chenguang Wang, Wei-Yu Chi, Chuan-Hsiang Huang, Richard B S Roden, T-C Wu, Yung-Nien Chang, Chien-Fu Hung, Shiwen Peng, Louise Ferrall, Stephanie Gaillard, Chenguang Wang, Wei-Yu Chi, Chuan-Hsiang Huang, Richard B S Roden, T-C Wu, Yung-Nien Chang, Chien-Fu Hung

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.

Keywords: DNA vaccine; E6; E7; HPV-associated cancer; HPV16; HPV18; PD-1; TA-HPV; human papillomavirus.

Copyright © 2021 Peng et al.

Figures

FIG 1
FIG 1
Schematic of the DNA constructs and map of pBI-11. (A) Schematic of the DNA constructs that are cloned into the pNGVL4a plasmid. The genes in the plasmids pBI-1, pBI-10.1, pBI-11, and pBI-12 are depicted. pBI-1 is in use for clinical trials under the name pNGVL4a-Sig/E7(detox)/HSP70. Blue indicates the native sequence of the target antigen. HSP70 is derived from Mycobacterium tuberculosis. Red indicates the gene was codon optimized prior to cloning it into the vector. (B) DNA construct map of pBI-11.
FIG 2
FIG 2
Western blot analysis to characterize the expression of the encoded HPV16/18 E6/E7 fusion protein in transfected cells. 293 expi cells were transfected with the designated plasmids and analyzed by Western blotting to assess the levels of target antigens. HPV16 E7-specific (A) and HPV18 and HPV16 E6-specific (B) MAbs were used as probes for the Western blot analysis of the antigen expression of the DNA vaccine constructs. 293 cells were transfected with the vaccines. (C) GAPDH was used as the loading control.
FIG 3
FIG 3
Characterization of HPV16 E7 and HPV18 E6 antigen presentation by cells transfected with the various DNA vaccines. 293-Db or 293 Kb cells were transfected with either pBI-10.1, pBI-11, or pBI-12 DNA vaccine or mock transfected using Lipofectamine 2000. Twenty-four hours later, these cells were harvested and cocultured with either murine H-2Db-restricted HPV16 E7 (aa 49 to 57) peptide-specific CD8+ T cells or murine H-2Kb-restricted HPV 18 E6 (aa 67 to 75) peptide-specific CD8+ T cells in the presence of GolgiPlug. The cells were then harvested, and IFN-γ intracellular staining was performed to determine the activation of HPV16 E7 or HPV18 E6 antigen-specific CD8+ T cells. (A) Bar graph to summarize flow cytometry data for activation of HPV16 E7-specific CD8+ T cells. (B) Bar graph to summarize the flow cytometry data for the activation of HPV18 E6-specific CD8+ T cells.
FIG 4
FIG 4
Comparison of HPV16/18 E6/E7-specific CD8+ T cell responses generated by the various HPV DNA vaccines. (A) Schematic illustration of the experimental design. Briefly, 5- to 8-week-old female C57BL/6 mice (5 mice/group) were vaccinated with either 25 μg/mouse of pBI-10.1, pBI-11, or pBI-12 DNA vaccine through intramuscular injection. The mice were boosted twice with the same dose and regimen with 1-week intervals. Seven days after the last vaccination, splenocytes were prepared from the vaccinated mice and stimulated with HPV16 E7 (aa 49 to 57) peptide, HPV16 E6 (aa 50 to 57) peptide, or HPV18 E6 (aa 67 to 75) peptide in the presence of GolgiPlug overnight. The splenocytes were stained with PE-conjugated anti-mouse CD8a. After permeabilization and fixation, the cells were further stained with FITC-conjugated anti-mouse IFN-γ. The cells were acquired with a FACSCalibur flow cytometer, and data were analyzed with CellQuest Pro software. (B) Bar graphs summarizing the data from flow cytometry analysis of HPV16 E6 (a), HPV16 E7 (b), and. HPV18 E6 (c) peptide-specific CD8+ T cell responses analyzed by IFN-γ intracellular staining. (d) Splenocytes pulsed without peptide as negative control. N.S., not significant.
FIG 5
FIG 5
Characterization of HPV antigen-specific CD8+ T cell-mediated immune responses and therapeutic antitumor effects of the various DNA vaccines in mice injected with HPV16 E6/E7-expressing tumors, model TC-1. Six- to eight-week-old female C57BL/6 mice (5 mice/group) were injected with 2 × 105 of TC-1 cells subcutaneously on day 0. On day 3, the mice were vaccinated with either pBI-10.1, pBI-11, or pBI-12 DNA (25 μg/50 μl/mouse) through i.m. injection and boosted twice as indicated or left untreated as control. (A) Schema of the experiment. (B) Detection of HPV16 E7-specific CD8+ T cells in peripheral blood using HPV16 E7 (aa 49 to 57) peptide-loaded tetramer staining. (C) Detection of HPV18 E6-specific CD8+ T cells in peripheral blood using HPV18 E6 (aa 67 to 75) peptide-loaded tetramer staining. (D) Summary of the TC-1 tumor volumes of the mice. (E) Kaplan-Meier analysis of the survival of TC-1 tumor-bearing mice.
FIG 6
FIG 6
Characterization of HPV antigen-specific CD8+ T cell-mediated immune responses in mice vaccinated with pBI-11 DNA alone or followed by TA-HPV. Six- to eight-week-old female C57BL/6 mice (5 mice/group) were vaccinated with pBI-11 DNA (25 μg/50 μl/mouse) through i.m. injection. The mice were boosted with the same regimen 7 days later. One week after the second vaccination, one group of the mice was vaccinated with pBI-11 DNA (25 μg/50 μl/mouse) through i.m. injection (DDD). Another group of mice was vaccinated with TA-HPV (1 × 106 PFU/50 μl/mouse) through i.m. injection (DDV). (A) Schema of the experiment. Six days after the last vaccination, peripheral blood was collected from the vaccinated or naive mice for HPV16 E7 tetramer staining. Fourteen days after the last vaccination, splenocytes were prepared from the vaccinated mice and stimulated with either HPV16 E6 (aa 50 to 57), HPV16 E7 (aa 49 to 57), or HPV18 E6 (aa 67 to 75) peptide in the presence of GolgiPlug. Intracellular IFN-γ cytokine staining assay was performed to detect antigen-specific CD8+ T cells. The cells were acquired with a FACSCalibur flow cytometer, and data were analyzed with CellQuest Pro software. (B) Summary of the HPV16 E7 tetramer flow cytometry analysis. (C) Intracellular IFN-γ cytokine staining assay after stimulation with either HPV16 E6 (aa 50 to 57) peptide (5 μg/ml), HPV16 E7 (aa 49 to 57) peptide (1 μg/ml), or HPV18 E6 (aa 67 to 75) peptide (1 μg/ml).
FIG 7
FIG 7
Characterization of the HPV antigen-specific immune response and antitumor effects in a TC-1 tumor-bearing mouse treated with DDV with or without anti-PD-1. Six- to eight-week-old female C57BL/6 mice (5 to 8 mice/group) were injected with 2 × 105 of TC-1 cells subcutaneously on day 0. On day 3, the mice were divided into 4 groups. The first group was used as the untreated control. The second group was injected with purified anti-mouse PD-1 monoclonal antibody (MAb; clone 29F.1A12, 200 μg/mouse) via intraperitoneal injection. The treatment was repeated every other day. The third group was vaccinated with pBI-11 DNA (25 μg/50 μl/mouse) through i.m. injection and boosted once 3 days later. The mice were further boosted with TA-HPV vaccinia virus 3 days later through skin scarification. The fourth group was treated with both anti-mouse PD-1 MAb and pBI-11 DNA vaccine prime followed by TA-HPV vaccinia virus boost as described in Materials and Methods. (A) Schema of the experiment. (B) Detection of HPV16 E7-specific CD8+ T cells in peripheral blood using HPV16 E7 tetramer staining. (C) Summary of the TC-1 tumor volumes in the mice. (D) Kaplan-Meier analysis of the survival of TC-1 tumor-bearing mice.

References

    1. Serrano B, Brotons M, Bosch FX, Bruni L. 2018. Epidemiology and burden of HPV-related disease. Best Pract Res Clin Obstet Gynaecol 47:14–26. doi:10.1016/j.bpobgyn.2017.08.006.
    1. Parkin DM 2006. The global health burden of infection-associated cancers in the year 2002. Int J Cancer 118:3030–3044. doi:10.1002/ijc.21731.
    1. Small W, Jr, Bacon MA, Bajaj A, Chuang LT, Fisher BJ, Harkenrider MM, Jhingran A, Kitchener HC, Mileshkin LR, Viswanathan AN, Gaffney DK. 2017. Cervical cancer: a global health crisis. Cancer 123:2404–2412. doi:10.1002/cncr.30667.
    1. Brianti P, De Flammineis E, Mercuri SR. 2017. Review of HPV-related diseases and cancers. New Microbiol 40:80–85.
    1. Walboomers JMM, Jacobs MV, Manos MM, Bosch FX, Kummer JA, Shah KV, Snijders PJF, Peto J, Meijer CJLM, Muñoz N. 1999. Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol 189:12–19. doi:10.1002/(SICI)1096-9896(199909)189:1<12::AID-PATH431>;2-F.
    1. Gillison ML, Chaturvedi AK, Anderson WF, Fakhry C. 2015. Epidemiology of human papillomavirus-positive head and neck squamous cell carcinoma. J Clin Oncol 33:3235–3242. doi:10.1200/JCO.2015.61.6995.
    1. D'Souza G, Dempsey A. 2011. The role of HPV in head and neck cancer and review of the HPV vaccine. Prev Med 53 Suppl 1:S5–S11. doi:10.1016/j.ypmed.2011.08.001.
    1. Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Pineros M, Znaor A, Bray F. 2019. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer 144:1941–1953. doi:10.1002/ijc.31937.
    1. Lewis A, Kang R, Levine A, Maghami E. 2015. The new face of head and neck cancer: the HPV epidemic. Oncology (Williston Park) 29:616–626.
    1. Spence T, Bruce J, Yip KW, Liu FF. 2016. HPV associated head and neck cancer. Cancers (Basel) 8:75. doi:10.3390/cancers8080075.
    1. Hildesheim A, Herrero R, Wacholder S, Rodriguez AC, Solomon D, Bratti MC, Schiller JT, Gonzalez P, Dubin G, Porras C, Jimenez SE, Lowy DR, Costa Rican H, Costa Rican HPV Vaccine Trial Group. 2007. Effect of human papillomavirus 16/18 L1 viruslike particle vaccine among young women with preexisting infection: a randomized trial. JAMA 298:743–753. doi:10.1001/jama.298.7.743.
    1. Yang A, Farmer E, Lin J, Wu TC, Hung CF. 2017. The current state of therapeutic and T cell-based vaccines against human papillomaviruses. Virus Res 231:148–165. doi:10.1016/j.virusres.2016.12.002.
    1. Doorbar J, Egawa N, Griffin H, Kranjec C, Murakami I. 2015. Human papillomavirus molecular biology and disease association. Rev Med Virol 25 Suppl 1:2–23. doi:10.1002/rmv.1822.
    1. Sanclemente G, Gill DK. 2002. Human papillomavirus molecular biology and pathogenesis. J Eur Acad Dermatol Venereol 16:231–240. doi:10.1046/j.1473-2165.2002.00419.x.
    1. Yugawa T, Kiyono T. 2009. Molecular mechanisms of cervical carcinogenesis by high-risk human papillomaviruses: novel functions of E6 and E7 oncoproteins. Rev Med Virol 19:97–113. doi:10.1002/rmv.605.
    1. Boursnell ME, Rutherford E, Hickling JK, Rollinson EA, Munro AJ, Rolley N, McLean CS, Borysiewicz LK, Vousden K, Inglis SC. 1996. Construction and characterisation of a recombinant vaccinia virus expressing human papillomavirus proteins for immunotherapy of cervical cancer. Vaccine 14:1485–1494. doi:10.1016/s0264-410x(96)00117-x.
    1. Kaufmann AM, Stern PL, Rankin EM, Sommer H, Nuessler V, Schneider A, Adams M, Onon TS, Bauknecht T, Wagner U, Kroon K, Hickling J, Boswell CM, Stacey SN, Kitchener HC, Gillard J, Wanders J, Roberts JS, Zwierzina H. 2002. Safety and immunogenicity of TA-HPV, a recombinant vaccinia virus expressing modified human papillomavirus (HPV)-16 and HPV-18 E6 and E7 genes, in women with progressive cervical cancer. Clin Cancer Res 8:3676–3685.
    1. Borysiewicz LK, Fiander A, Nimako M, Man S, Wilkinson GW, Westmoreland D, Evans AS, Adams M, Stacey SN, Boursnell ME, Rutherford E, Hickling JK, Inglis SC. 1996. A recombinant vaccinia virus encoding human papillomavirus types 16 and 18, E6 and E7 proteins as immunotherapy for cervical cancer. Lancet 347:1523–1527. doi:10.1016/s0140-6736(96)90674-1.
    1. Maldonado L, Teague JE, Morrow MP, Jotova I, Wu TC, Wang C, Desmarais C, Boyer JD, Tycko B, Robins HS, Clark RA, Trimble CL. 2014. Intramuscular therapeutic vaccination targeting HPV16 induces T cell responses that localize in mucosal lesions. Sci Transl Med 6:221ra13. doi:10.1126/scitranslmed.3007323.
    1. Davidson EJ, Boswell CM, Sehr P, Pawlita M, Tomlinson AE, McVey RJ, Dobson J, Roberts JS, Hickling J, Kitchener HC, Stern PL. 2003. Immunological and clinical responses in women with vulval intraepithelial neoplasia vaccinated with a vaccinia virus encoding human papillomavirus 16/18 oncoproteins. Cancer Res 63:6032–6041.
    1. Baldwin PJ, van der Burg SH, Boswell CM, Offringa R, Hickling JK, Dobson J, Roberts JS, Latimer JA, Moseley RP, Coleman N, Stanley MA, Sterling JC. 2003. Vaccinia-expressed human papillomavirus 16 and 18 e6 and e7 as a therapeutic vaccination for vulval and vaginal intraepithelial neoplasia. Clin Cancer Res 9:5205–5213.
    1. Qiu J, Peng S, Ma Y, Yang A, Farmer E, Cheng MA, Roden RBS, Wu TC, Chang YN, Hung CF. 2018. Epithelial boost enhances antigen expression by vaccinia virus for the generation of potent CD8+ T cell-mediated antitumor immunity following DNA priming vaccination. Virology 525:205–215. doi:10.1016/j.virol.2018.09.019.
    1. Sharma P, Allison JP. 2015. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 161:205–214. doi:10.1016/j.cell.2015.03.030.
    1. Chen CH, Wang TL, Hung CF, Yang Y, Young RA, Pardoll DM, Wu TC. 2000. Enhancement of DNA vaccine potency by linkage of antigen gene to an HSP70 gene. Cancer Res 60:1035–1042.
    1. Arnold-Schild D, Hanau D, Spehner D, Schmid C, Rammensee HG, de la Salle H, Schild H. 1999. Cutting edge: receptor-mediated endocytosis of heat shock proteins by professional antigen-presenting cells. J Immunol 162:3757–3760.
    1. Hsu KF, Hung CF, Cheng WF, He L, Slater LA, Ling M, Wu TC. 2001. Enhancement of suicidal DNA vaccine potency by linking Mycobacterium tuberculosis heat shock protein 70 to an antigen. Gene Ther 8:376–383. doi:10.1038/sj.gt.3301408.
    1. Cheng WF, Hung CF, Lin KY, Ling M, Juang J, He L, Lin CT, Wu TC. 2003. CD8+ T cells, NK cells and IFN-gamma are important for control of tumor with downregulated MHC class I expression by DNA vaccination. Gene Ther 10:1311–1320. doi:10.1038/sj.gt.3301982.
    1. Trimble C, Lin CT, Hung CF, Pai S, Juang J, He L, Gillison M, Pardoll D, Wu L, Wu TC. 2003. Comparison of the CD8+ T cell responses and antitumor effects generated by DNA vaccine administered through gene gun, biojector, and syringe. Vaccine 21:4036–4042. doi:10.1016/s0264-410x(03)00275-5.
    1. Kim JW, Hung CF, Juang J, He L, Kim TW, Armstrong DK, Pai SI, Chen PJ, Lin CT, Boyd DA, Wu TC. 2004. Comparison of HPV DNA vaccines employing intracellular targeting strategies. Gene Ther 11:1011–1018. doi:10.1038/sj.gt.3302252.
    1. Trimble CL, Peng S, Kos F, Gravitt P, Viscidi R, Sugar E, Pardoll D, Wu TC. 2009. A phase I trial of a human papillomavirus DNA vaccine for HPV16+ cervical intraepithelial neoplasia 2/3. Clin Cancer Res 15:361–367. doi:10.1158/1078-0432.CCR-08-1725.
    1. Sun YY, Peng S, Han L, Qiu J, Song L, Tsai Y, Yang B, Roden RB, Trimble CL, Hung CF, Wu TC. 2016. Local HPV recombinant vaccinia boost following priming with an HPV DNA vaccine enhances local HPV-specific CD8+ T-cell-mediated tumor control in the genital tract. Clin Cancer Res 22:657–669. doi:10.1158/1078-0432.CCR-15-0234.
    1. Peng S, Qiu J, Yang A, Yang B, Jeang J, Wang JW, Chang YN, Brayton C, Roden RBS, Hung CF, Wu TC. 2016. Optimization of heterologous DNA-prime, protein boost regimens and site of vaccination to enhance therapeutic immunity against human papillomavirus-associated disease. Cell Biosci 6:16. doi:10.1186/s13578-016-0080-z.
    1. Kutzler MA, Weiner DB. 2008. DNA vaccines: ready for prime time? Nat Rev Genet 9:776–788. doi:10.1038/nrg2432.
    1. Liu Y, Wu L, Tong R, Yang F, Yin L, Li M, You L, Xue J, Lu Y. 2019. PD-1/PD-L1 inhibitors in cervical cancer. Front Pharmacol 10:65. doi:10.3389/fphar.2019.00065.
    1. McCusker MG, Orkoulas-Razis D, Mehra R. 2020. Potential of pembrolizumab in metastatic or recurrent head and neck cancer: evidence to date. Onco Targets Ther 13:3047–3059. doi:10.2147/OTT.S196252.
    1. Steele KE, Tan TH, Korn R, Dacosta K, Brown C, Kuziora M, Zimmermann J, Laffin B, Widmaier M, Rognoni L, Cardenes R, Schneider K, Boutrin A, Martin P, Zha J, Wiestler T. 2018. Measuring multiple parameters of CD8+ tumor-infiltrating lymphocytes in human cancers by image analysis. J Immunother Cancer 6:20. doi:10.1186/s40425-018-0326-x.
    1. Tang H, Wang Y, Chlewicki LK, Zhang Y, Guo J, Liang W, Wang J, Wang X, Fu YX. 2016. Facilitating T cell infiltration in tumor microenvironment overcomes resistance to PD-L1 blockade. Cancer Cell 29:285–296. doi:10.1016/j.ccell.2016.02.004.
    1. Ma Y, Yang A, Peng S, Qiu J, Farmer E, Hung CF, Wu TC. 2017. Characterization of HPV18 E6-specific T cell responses and establishment of HPV18 E6-expressing tumor model. Vaccine 35:3850–3858. doi:10.1016/j.vaccine.2017.05.081.
    1. Peng S, Ji H, Trimble C, He L, Tsai YC, Yeatermeyer J, Boyd DA, Hung CF, Wu TC. 2004. Development of a DNA vaccine targeting human papillomavirus type 16 oncoprotein E6. J Virol 78:8468–8476. doi:10.1128/JVI.78.16.8468-8476.2004.
    1. Cheng MA, Farmer E, Huang C, Lin J, Hung CF, Wu TC. 2018. Therapeutic DNA vaccines for human papillomavirus and associated diseases. Hum Gene Ther 29:971–996. doi:10.1089/hum.2017.197.
    1. Chandra J, Dutton JL, Li B, Woo W-P, Xu Y, Tolley LK, Yong M, Wells JW, R Leggatt G, Finlayson N, Frazer IH. 2017. DNA vaccine encoding HPV16 oncogenes E6 and E7 induces potent cell-mediated and humoral immunity which protects in tumor challenge and drives E7-expressing skin graft rejection. J Immunother 40:62–70. doi:10.1097/CJI.0000000000000156.
    1. Lv J, Liu C, Huang H, Meng L, Jiang B, Cao Y, Zhou Z, She T, Qu L, Wei Song S, Shou C. 2013. Suppression of breast tumor growth by DNA vaccination against phosphatase of regenerating liver 3. Gene Ther 20:834–845. doi:10.1038/gt.2013.5.
    1. Peng S, Trimble C, He L, Tsai YC, Lin CT, Boyd DA, Pardoll D, Hung CF, Wu TC. 2006. Characterization of HLA-A2-restricted HPV-16 E7-specific CD8+ T-cell immune responses induced by DNA vaccines in HLA-A2 transgenic mice. Gene Ther 13:67–77. doi:10.1038/sj.gt.3302607.
    1. Fiander AN, Tristram AJ, Davidson EJ, Tomlinson AE, Man S, Baldwin PJ, Sterling JC, Kitchener HC. 2006. Prime-boost vaccination strategy in women with high-grade, noncervical anogenital intraepithelial neoplasia: clinical results from a multicenter phase II trial. Int J Gynecol Cancer 16:1075–1081. doi:10.1111/j.1525-1438.2006.00598.x.
    1. Adams M, Borysiewicz L, Fiander A, Man S, Jasani B, Navabi H, Lipetz C, Evans AS, Mason M. 2001. Clinical studies of human papilloma vaccines in pre-invasive and invasive cancer. Vaccine 19:2549–2556. doi:10.1016/S0264-410X(00)00488-6.
    1. Chen Z, Pang N, Du R, Zhu Y, Fan L, Cai D, Ding Y, Ding J. 2016. Elevated expression of programmed death-1 and programmed death ligand-1 negatively regulates immune response against cervical cancer cells. Mediators Inflamm 2016:6891482. doi:10.1155/2016/6891482.
    1. Peng S, Tan M, Li YD, Cheng MA, Farmer E, Ferrall L, Gaillard S, Roden RBS, Hung CF, Wu TC. 2020. PD-1 blockade synergizes with intratumoral vaccination of a therapeutic HPV protein vaccine and elicits regression of tumor in a preclinical model. Cancer Immunol Immunother doi:10.1007/s00262-020–02754-x.
    1. Mezache L, Paniccia B, Nyinawabera A, Nuovo GJ. 2015. Enhanced expression of PD L1 in cervical intraepithelial neoplasia and cervical cancers. Mod Pathol 28:1594–1602. doi:10.1038/modpathol.2015.108.
    1. Liu C, Lu J, Tian H, Du W, Zhao L, Feng J, Yuan D, Li Z. 2017. Increased expression of PD-L1 by the human papillomavirus 16 E7 oncoprotein inhibits anticancer immunity. Mol Med Rep 15:1063–1070. doi:10.3892/mmr.2017.6102.
    1. Allouch S, Malki A, Allouch A, Gupta I, Vranic S, Al Moustafa AE. 2020. High-risk HPV oncoproteins and PD-1/PD-L1 interplay in human cervical cancer: recent evidence and future directions. Front Oncol 10:914. doi:10.3389/fonc.2020.00914.
    1. Fu J, Malm IJ, Kadayakkara DK, Levitsky H, Pardoll D, Kim YJ. 2014. Preclinical evidence that PD1 blockade cooperates with cancer vaccine TEGVAX to elicit regression of established tumors. Cancer Res 74:4042–4052. doi:10.1158/0008-5472.CAN-13-2685.
    1. Rekoske BT, Smith HA, Olson BM, Maricque BB, McNeel DG. 2015. PD-1 or PD-L1 blockade restores antitumor efficacy following SSX2 epitope-modified DNA vaccine immunization. Cancer Immunol Res 3:946–955. doi:10.1158/2326-6066.CIR-14-0206.
    1. McCarthy C, Youde SJ, Man S. 2006. Definition of an HPV18/45 cross-reactive human T-cell epitope after DNA immunisation of HLA-A2/KB transgenic mice. Int J Cancer 118:2514–2521. doi:10.1002/ijc.21643.
    1. Alvarez RD, Huh WK, Bae S, Lamb LS, Jr, Conner MG, Boyer J, Wang C, Hung C-F, Sauter E, Paradis M, Adams EA, Hester S, Jackson BE, Wu TC, Trimble CL. 2016. A pilot study of pNGVL4a-CRT/E7(detox) for the treatment of patients with HPV16+ cervical intraepithelial neoplasia 2/3 (CIN2/3). Gynecol Oncol 140:245–252. doi:10.1016/j.ygyno.2015.11.026.
    1. Demers GW, Espling E, Harry JB, Etscheid BG, Galloway DA. 1996. Abrogation of growth arrest signals by human papillomavirus type 16 E7 is mediated by sequences required for transformation. J Virol 70:6862–6869. doi:10.1128/JVI.70.10.6862-6869.1996.
    1. Edmonds C, Vousden KH. 1989. A point mutational analysis of human papillomavirus type 16 E7 protein. J Virol 63:2650–2656. doi:10.1128/JVI.63.6.2650-2656.1989.
    1. Cassetti MC, McElhiney SP, Shahabi V, Pullen JK, Le Poole IC, Eiben GL, Smith LR, Kast WM. 2004. Antitumor efficacy of Venezuelan equine encephalitis virus replicon particles encoding mutated HPV16 E6 and E7 genes. Vaccine 22:520–527. doi:10.1016/j.vaccine.2003.07.003.
    1. Jewers RJ, Hildebrandt P, Ludlow JW, Kell B, McCance DJ. 1992. Regions of human papillomavirus type 16 E7 oncoprotein required for immortalization of human keratinocytes. J Virol 66:1329–1335. doi:10.1128/JVI.66.3.1329-1335.1992.
    1. Brehm A, Nielsen SJ, Miska EA, McCance DJ, Reid JL, Bannister AJ, Kouzarides T. 1999. The E7 oncoprotein associates with Mi2 and histone deacetylase activity to promote cell growth. EMBO J 18:2449–2458. doi:10.1093/emboj/18.9.2449.
    1. Antinore MJ, Birrer MJ, Patel D, Nader L, McCance DJ. 1996. The human papillomavirus type 16 E7 gene product interacts with and trans-activates the AP1 family of transcription factors. EMBO J 15:1950–1960. doi:10.1002/j.1460-2075.1996.tb00546.x.
    1. Zhang Y, Fan S, Meng Q, Ma Y, Katiyar P, Schlegel R, Rosen EM. 2005. BRCA1 interaction with human papillomavirus oncoproteins. J Biol Chem 280:33165–33177. doi:10.1074/jbc.M505124200.
    1. McIntyre MC, Ruesch MN, Laimins LA. 1996. Human papillomavirus E7 oncoproteins bind a single form of cyclin E in a complex with cdk2 and p107. Virology 215:73–82. doi:10.1006/viro.1996.0008.
    1. Zhou ZX, Zhao C, Li QQ, Zeng Y. 2014. A novel mutant of human papillomavirus type 18 E6E7 fusion gene and its transforming activity. Asian Pac J Cancer Prev 15:7395–7399. doi:10.7314/apjcp.2014.15.17.7395.
    1. Dalal S, Gao Q, Androphy EJ, Band V. 1996. Mutational analysis of human papillomavirus type 16 E6 demonstrates that p53 degradation is necessary for immortalization of mammary epithelial cells. J Virol 70:683–688. doi:10.1128/JVI.70.2.683-688.1996.
    1. Gao Q, Singh L, Kumar A, Srinivasan S, Wazer DE, Band V. 2001. Human papillomavirus type 16 E6-induced degradation of E6TP1 correlates with its ability to immortalize human mammary epithelial cells. J Virol 75:4459–4466. doi:10.1128/JVI.75.9.4459-4466.2001.
    1. Gardiol D, Kuhne C, Glaunsinger B, Lee SS, Javier R, Banks L. 1999. Oncogenic human papillomavirus E6 proteins target the discs large tumour suppressor for proteasome-mediated degradation. Oncogene 18:5487–5496. doi:10.1038/sj.onc.1202920.
    1. Simonson SJ, Difilippantonio MJ, Lambert PF. 2005. Two distinct activities contribute to human papillomavirus 16 E6's oncogenic potential. Cancer Res 65:8266–8273. doi:10.1158/0008-5472.CAN-05-1651.
    1. Shai A, Brake T, Somoza C, Lambert PF. 2007. The human papillomavirus E6 oncogene dysregulates the cell cycle and contributes to cervical carcinogenesis through two independent activities. Cancer Res 67:1626–1635. doi:10.1158/0008-5472.CAN-06-3344.
    1. Spanos WC, Geiger J, Anderson ME, Harris GF, Bossler AD, Smith RB, Klingelhutz AJ, Lee JH. 2008. Deletion of the PDZ motif of HPV16 E6 preventing immortalization and anchorage-independent growth in human tonsil epithelial cells. Head Neck 30:139–147. doi:10.1002/hed.20673.
    1. Pim D, Storey A, Thomas M, Massimi P, Banks L. 1994. Mutational analysis of HPV-18 E6 identifies domains required for p53 degradation in vitro, abolition of p53 transactivation in vivo and immortalisation of primary BMK cells. Oncogene 9:1869–1876.
    1. Gu Z, Pim D, Labrecque S, Banks L, Matlashewski G. 1994. DNA damage induced p53 mediated transcription is inhibited by human papillomavirus type 18 E6. Oncogene 9:629–633.
    1. Thomas M, Matlashewski G, Pim D, Banks L. 1996. Induction of apoptosis by p53 is independent of its oligomeric state and can be abolished by HPV-18 E6 through ubiquitin mediated degradation. Oncogene 13:265–273.
    1. Vande Pol SB, Klingelhutz AJ. 2013. Papillomavirus E6 oncoproteins. Virology 445:115–137. doi:10.1016/j.virol.2013.04.026.
    1. Stern LJ, Wiley DC. 1994. Antigenic peptide binding by class I and class II histocompatibility proteins. Behring Inst Mitt 1994:1–10.
    1. Chen C, Li Z, Huang H, Suzek BE, Wu CH, UniProt Consortium. 2013. A fast peptide match service for UniProt Knowledgebase. Bioinformatics 29:2808–2809. doi:10.1093/bioinformatics/btt484.
    1. Vita R, Mahajan S, Overton JA, Dhanda SK, Martini S, Cantrell JR, Wheeler DK, Sette A, Peters B. 2019. The Immune Epitope Database (IEDB): 2018 update. Nucleic Acids Res 47:D339–D343. doi:10.1093/nar/gky1006.
    1. Lin KY, Guarnieri FG, Staveley-O'Carroll KF, Levitsky HI, August JT, Pardoll DM, Wu TC. 1996. Treatment of established tumors with a novel vaccine that enhances major histocompatibility class II presentation of tumor antigen. Cancer Res 56:21–26.
    1. Peng S, Mattox A, Best SR, Barbu AM, Burns JA, Akpeng B, Jeang J, Yang B, Ishida E, Hung CF, Wu TC, Pai SI. 2016. Identification of the murine H-2D(b) and human HLA-A*0201 MHC class I-restricted HPV6 E7-specific cytotoxic T lymphocyte epitopes. Cancer Immunol Immunother 65:261–271. doi:10.1007/s00262-016-1793-x.
    1. Wang TL, Ling M, Shih IM, Pham T, Pai SI, Lu Z, Kurman RJ, Pardoll DM, Wu TC. 2000. Intramuscular administration of E7-transfected dendritic cells generates the most potent E7-specific anti-tumor immunity. Gene Ther 7:726–733. doi:10.1038/sj.gt.3301160.
    1. Adams M, Borysiewicz L, Fiander A, Man S, Jasani B, Navabi H, Evans AS, Mason M. 2001. Clinical studies of human papilloma vaccines in cervical cancer. Adv Exp Med Biol 495:419–427. doi:10.1007/978-1-4615-0685-0_61.

Source: PubMed

3
Subskrybuj