Haoru Dong, Xinhua Shu, Qiang Xu, Chen Zhu, Andreas M. Kaufmann, Zhi-Ming Zheng, Andreas E. Albers and Xu Qian. Current Status of Human Papillomavirus-Related Head and Neck Cancer: From Viral Genome to Patient Care[J]. Virologica Sinica, 2021, 36(6): 1284-1302. doi: 10.1007/s12250-021-00413-8
Citation: Haoru Dong, Xinhua Shu, Qiang Xu, Chen Zhu, Andreas M. Kaufmann, Zhi-Ming Zheng, Andreas E. Albers, Xu Qian. Current Status of Human Papillomavirus-Related Head and Neck Cancer: From Viral Genome to Patient Care .VIROLOGICA SINICA, 2021, 36(6) : 1284-1302.  http://dx.doi.org/10.1007/s12250-021-00413-8

人乳头瘤病毒相关头颈部肿瘤研究进展:从病毒基因组到临床实践

  • 通讯作者: 钱旭, Qian_michelle2014@163.com, ORCID: http://orcid.org/0000-0003-0629-4460
  • 收稿日期: 2021-01-21
    录用日期: 2021-05-18
    出版日期: 2021-06-21
  • 近年来,对高危型人乳头瘤病毒(HPV)感染引起头颈部鳞癌发生和发展的研究不断地深入。据报道,约38%-80%的口咽部鳞癌以及大部分的宫颈癌都与HPV感染相关。病毒癌基因E6和E7的表达是致癌的关键。HPV阳性口咽部鳞癌表现出与阴性肿瘤不同的生物学行为和良好的预后。因此,为减少治疗带来的毒副作用和提高患者生存质量,一些临床研究开始探索降低HPV阳性口咽部鳞癌患者的治疗强度。但是,如何有效并且准确地筛选患者以接受不同强度的治疗仍然存在较大的挑战。与此同时,随着免疫治疗的不断临床应用,进一步理解HPV阳性肿瘤的免疫特性,深入研究患者对免疫治疗的不同反应,将有助于提高免疫治疗的疗效。本文就HPV相关头颈部鳞癌的生物学特性,以及目前对该类患者的临床研究进行综述。随着检测技术的不断进步,发现HPV相关头颈部鳞癌的特异性生物学标志物或治疗靶点将有助于临床对该类患者进行针对性的治疗。

Current Status of Human Papillomavirus-Related Head and Neck Cancer: From Viral Genome to Patient Care

  • Corresponding author: Xu Qian, Qian_michelle2014@163.com
  • ORCID: http://orcid.org/0000-0003-0629-4460
  • Received Date: 21 January 2021
    Accepted Date: 18 May 2021
    Published Date: 21 June 2021
  • Human papillomavirus (HPV) infection identified as a definitive human carcinogen is increasingly being recognized for its role in carcinogenesis of human cancers. Up to 38%–80% of head and neck squamous cell carcinoma (HNSCC) in oropharyngeal location (OPSCC) and nearly all cervical cancers contain the HPV genome which is implicated in causing cancer through its oncoproteins E6 and E7. Given by the biologically distinct HPV-related OPSCC and a more favorable prognosis compared to HPV-negative tumors, clinical trials on de-escalation treatment strategies for these patients have been studied. It is therefore raised the questions for the patient stratification if treatment de-escalation is feasible. Moreover, understanding the crosstalk of HPV-mediated malignancy and immunity with clinical insights from the proportional response rate to immune checkpoint blockade treatments in patients with HNSCC is of importance to substantially improve the treatment efficacy. This review discusses the biology of HPV-related HNSCC as well as successful clinically findings with promising candidates in the pipeline for future directions. With the advent of various sequencing technologies, further biomolecules associated with HPV-related HNSCC progression are currently being identified to be used as potential biomarkers or targets for clinical decisions throughout the continuum of cancer care.


  • 加载中
    1. Aggarwal C, Cohen RB, Morrow MP, Kraynyak KA, Sylvester AJ, Knoblock DM, Bauml JM, Weinstein GS, Lin A, Boyer J, Sakata L, Tan S, Anton A, Dickerson K, Mangrolia D, Vang R, Dallas M, Oyola S, Duff S, Esser M, Kumar R, Weiner D, Csiki I, Bagarazzi ML (2019) Immunotherapy targeting HPV16/18 generates potent immune responses in HPV-associated head and neck cancer. Clin Cancer Res 25: 110–124
        doi: 10.1158/1078-0432.CCR-18-1763

    2. Agrawal N, Frederick MJ, Pickering CR, Bettegowda C, Chang K, Li RJ, Fakhry C, Xie TX, Zhang J, Wang J, Zhang N, El-Naggar AK, Jasser SA, Weinstein JN, Treviño L, Drummond JA, Muzny DM, Wu Y, Wood LD, Hruban RH, Westra WH, Koch WM, Califano JA, Gibbs RA, Sidransky D, Vogelstein B, Velculescu VE, Papadopoulos N, Wheeler DA, Kinzler KW, Myers JN (2011) Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1. Science 333: 1154–1157
        doi: 10.1126/science.1206923

    3. Ajiro M, Jia R, Zhang L, Liu X, Zheng ZM (2012) Intron definition and a branch site adenosine at nt 385 control RNA splicing of HPV16 E6*I and E7 expression. PLoS ONE 7: e46412
        doi: 10.1371/journal.pone.0046412

    4. Ajiro M, Tang S, Doorbar J, Zheng ZM (2016) Serine/Arginine-rich splicing factor 3 and heterogeneous nuclear ribonucleoprotein A1 regulate alternative RNA splicing and gene expression of human papillomavirus 18 through two functionally distinguishable cis elements. J Virol 90: 9138–9152
        doi: 10.1128/JVI.00965-16

    5. Akagi K, Li J, Broutian TR, Padilla-Nash H, Xiao W, Jiang B, Rocco JW, Teknos TN, Kumar B, Wangsa D, He D, Ried T, Symer DE, Gillison ML (2014) Genome-wide analysis of HPV integration in human cancers reveals recurrent, focal genomic instability. Genome Res 24: 185–199
        doi: 10.1101/gr.164806.113

    6. Albers A, Abe K, Hunt J, Wang J, Lopez-Albaitero A, Schaefer C, Gooding W, Whiteside TL, Ferrone S, DeLeo A, Ferris RL (2005) Antitumor activity of human papillomavirus type 16 E7-specific T cells against virally infected squamous cell carcinoma of the head and neck. Cancer Res 65: 11146–11155
        doi: 10.1158/0008-5472.CAN-05-0772

    7. Albers AE, Qian X, Kaufmann AM, Coordes A (2017) Meta analysis: HPV and p16 pattern determines survival in patients with HNSCC and identifies potential new biologic subtype. Sci Rep 7: 16715
        doi: 10.1038/s41598-017-16918-w

    8. Albers AE, Strauss L, Liao T, Hoffmann TK, Kaufmann AM (2010) T cell-tumor interaction directs the development of immunotherapies in head and neck cancer. Clin Dev Immunol 2010: 236378
        doi: 10.1155/2010/236378

    9. Altenhofen B, DeWees TA, Ahn JW, Yeat NC, Goddu S, Chen I, Lewis JS Jr, Thorstad WL, Chole RA (2017) Human papilloma virus load and PD-1/PD-L1, CD8(+) and FOXP3 in anal cancer patients treated with chemoradiotherapy: Rationale for immunotherapy. Oncoimmunology 6: e1288331
        doi: 10.1080/2162402X.2017.1288331

    10. Balermpas P, Martin D, Wieland U, Rave-Frank M, Strebhardt K, Rödel C, Fokas E, Rödel F (2017) Human papilloma virus load and PD-1/PD-L1, CD8(+) and FOXP3 in anal cancer patients treated with chemoradiotherapy: rationale for immunotherapy. Oncoimmunology 6: e1288331
        doi: 10.1080/2162402X.2017.1288331

    11. Bauml J, Seiwert TY, Pfister DG, Worden F, Liu SV, Gilbert J, Saba NF, Weiss J, Wirth L, Sukari A, Kang H, Gibson MK, Massarelli E, Powell S, Meister A, Shu X, Cheng JD, Haddad R (2017) Pembrolizumab for platinum- and cetuximab-refractory head and neck cancer: results from a single-arm, phase Ⅱ study. J Clin Oncol 35: 1542–1549
        doi: 10.1200/JCO.2016.70.1524

    12. Beachler DC, Abraham AG, Silverberg MJ, Jing Y, Fakhry C, Gill MJ, Dubrow R, Kitahata MM, Klein MB, Burchell AN, Korthuis PT, Moore RD, D'Souza G (2014) Incidence and risk factors of HPV-related and HPV-unrelated Head and Neck Squamous Cell Carcinoma in HIV-infected individuals. Oral Oncol 50: 1169–1176
        doi: 10.1016/j.oraloncology.2014.09.011

    13. Bhatt KH, Neller MA, Srihari S, Crooks P, Lekieffre L, Aftab BT, Liu H, Smith C, Kenny L, Porceddu S, Khanna R (2020) Profiling HPV-16-specific T cell responses reveals broad antigen reactivities in oropharyngeal cancer patients. J Exp Med 217: e20200389
        doi: 10.1084/jem.20200389

    14. Bigelow EO, Seiwert TY, Fakhry C (2020) Deintensification of treatment for human papillomavirus-related oropharyngeal cancer: Current state and future directions. Oral Oncol 105: 104652
        doi: 10.1016/j.oraloncology.2020.104652

    15. Boscolo-Rizzo P, Del Mistro A, Bussu F, Lupato V, Baboci L, Almadori G (2013) New insights into human papillomavirus-associated head and neck squamous cell carcinoma. Acta Otorhinolaryngol Ital 33: 77–87

    16. Boscolo-Rizzo P, Furlan C, Lupato V, Polesel J, Fratta E (2017) Novel insights into epigenetic drivers of oropharyngeal squamous cell carcinoma: role of HPV and lifestyle factors. Clin Epigenetics 9: 124
        doi: 10.1186/s13148-017-0424-5

    17. Böttinger P, Schreiber K, Hyjek E, Krausz T, Spiotto MT, Steiner M, Idel C, Booras H, Beck-Engeser G, Riederer J, Willimsky G, Wolf SP, Karrison T, Jensen E, Weichselbaum RR, Nakamura Y, Yew PY, Lambert PF, Kurita T, Kiyotani K, Leisegang M, Schreiber H (2020) Cooperation of genes in HPV16 E6/E7-dependent cervicovaginal carcinogenesis trackable by endoscopy and independent of exogenous estrogens or carcinogens. Carcinogenesis 41: 1605–1615
        doi: 10.1093/carcin/bgaa027

    18. Bouvard V, Baan R, Straif K, Grosse Y, Secretan B, El Ghissassi F, Benbrahim-Tallaa L, Guha N, Freeman C, Galichet L, Cogliano V (2009) A review of human carcinogens–Part B: biological agents. Lancet Oncol 10: 321–322
        doi: 10.1016/S1470-2045(09)70096-8

    19. Božinović K, Sabol I, Rakušić Z, Jakovčević A, Šekerija M, Lukinović J, Prgomet D, Grce M (2019) HPV-driven oropharyngeal squamous cell cancer in Croatia - Demography and survival. PLoS ONE 14: e0211577
        doi: 10.1371/journal.pone.0211577

    20. Brant AC, Majerciak V, Moreira MAM, Zheng ZM (2019) HPV18 utilizes two alternative branch sites for E6*I splicing to produce E7 protein. Virol Sin 34: 211–221
        doi: 10.1007/s12250-019-00098-0

    21. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A (2018) Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68: 394–424
        doi: 10.3322/caac.21492

    22. Broutian TR, Jiang B, Li J, Akagi K, Gui S, Zhou Z, Xiao W, Symer DE, Gillison ML (2020) Human papillomavirus insertions identify the PIM family of serine/threonine kinases as targetable driver genes in head and neck squamous cell carcinoma. Cancer Lett 476: 23–33
        doi: 10.1016/j.canlet.2020.01.012

    23. Buexm LA, Soares-Lima SC, Brennan P, Fernandes PV (2020) Hpv impact on oropharyngeal cancer patients treated at the largest cancer center from Brazil. Cancer Lett 477: 70–75
        doi: 10.1016/j.canlet.2020.02.023

    24. Burley M, Roberts S, Parish JL (2020) Epigenetic regulation of human papillomavirus transcription in the productive virus life cycle. Semin Immunopathol 42: 159–171
        doi: 10.1007/s00281-019-00773-0

    25. Burns MB, Temiz NA, Harris RS (2013) Evidence for APOBEC3B mutagenesis in multiple human cancers. Nat Genet 45: 977–983
        doi: 10.1038/ng.2701

    26. Burtness B, Harrington KJ, Greil R, Soulières D, Tahara M, De Castro G, Psyrri Jr A, Basté N, Neupane P, Bratland Å, Fuereder T, Hughes BGM, Mesía R, Ngamphaiboon N, Rordorf T, Wan Ishak WZ, Hong RL, González Mendoza R, Roy A, Zhang Y, Gumuscu B, Cheng JD, Jin F, Rischin D (2019) Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): a randomised, open-label, phase 3 study. Lancet 394: 1915–1928
        doi: 10.1016/S0140-6736(19)32591-7

    27. Castellsagué X, Alemany L, Quer M, Halec G, Quirós B, Tous S, Clavero O, Alòs L, Biegner T, Szafarowski T, Alejo M, Holzinger D, Cadena E, Claros E, Hall G, Laco J, Poljak M, Benevolo M, Kasamatsu E, Mehanna H, Ndiaye C, Guimerà N, Lloveras B, León X, Ruiz-Cabezas JC, Alvarado-Cabrero I, Kang CS, Oh JK, Garcia-Rojo M, Iljazovic E, Ajayi OF, Duarte F, Nessa A, Tinoco L, Duran-Padilla MA, Pirog EC, Viarheichyk H, Morales H, Costes V, Félix A, Germar MJ, Mena M, Ruacan A, Jain A, Mehrotra R, Goodman MT, Lombardi LE, Ferrera A, Malami S, Albanesi EI, Dabed P, Molina C, López-Revilla R, Mandys V, González ME, Velasco J, Bravo IG, Quint W, Pawlita M, Muñoz N, de Sanjosé S, Xavier Bosch F (2016) HPV Involvement in Head and Neck Cancers: Comprehensive Assessment of Biomarkers in 3680 Patients. J Natl Cancer Inst 108: 403
        doi: 10.1093/jnci/djv403

    28. Chargi N, Bril SI, Swartz JE, Wegner I, Willems SM, de Bree R (2020) Skeletal muscle mass is an imaging biomarker for decreased survival in patients with oropharyngeal squamous cell carcinoma. Oral Oncol 101: 104519
        doi: 10.1016/j.oraloncology.2019.104519

    29. Chaturvedi AK, Anderson WF, Lortet-Tieulent J, Curado MP, Ferlay J, Franceschi S, Rosenberg PS, Bray F, Gillison ML (2013) Worldwide trends in incidence rates for oral cavity and oropharyngeal cancers. J Clin Oncol 31: 4550–4559
        doi: 10.1200/JCO.2013.50.3870

    30. Chen TC, Wu CT, Ko JY, Yang TL, Lou PJ, Wang CP, Chang YL (2020) Clinical characteristics and treatment outcome of oropharyngeal squamous cell carcinoma in an endemic betel quid region. Sci Rep 10: 526
        doi: 10.1038/s41598-019-57177-1

    31. Chor JS, Vlantis AC, Chow TL, Fung SC, Ng FY, Lau CH, Chan AB, Ho LC, Kwong WH, Fung MN, Lam EW, Mak KL, Lam HC, Kok AS, Ho WC, Yeung AC, Chan PK (2016) The role of human papillomavirus in head and neck squamous cell carcinoma: a case control study on a southern Chinese population. J Med Virol 88: 877–887
        doi: 10.1002/jmv.24405

    32. Christensen JT, Grønhøj C, Zamani M, Brask J, Kjær EKR, Lajer H, von Buchwald C (2019) Association between oropharyngeal cancers with known HPV and p16 status and cervical intraepithelial neoplasia: a Danish population-based study. Acta Oncol 58: 267–272
        doi: 10.1080/0284186X.2018.1546059

    33. Chuang SC, Scelo G, Tonita JM, Tamaro S, Jonasson JG, Kliewer EV, Hemminki K, Weiderpass E, Pukkala E, Tracey E, Friis S, Pompe-Kirn V, Brewster DH, Martos C, Chia KS, Boffetta P, Brennan P, Hashibe M (2008) Risk of second primary cancer among patients with head and neck cancers: a pooled analysis of 13 cancer registries. Int J Cancer 123: 2390–2396
        doi: 10.1002/ijc.23798

    34. Cillo AR, Kürten CHL, Tabib T, Qi Z, Onkar S, Wang T, Liu A, Duvvuri U, Kim S, Soose RJ, Oesterreich S, Chen W, Lafyatis R, Bruno TC, Ferris RL, Vignali DAA (2020) Immune Landscape of Viral- and Carcinogen-Driven Head and Neck Cancer. Immunity 52: 183-199. e189
        doi: 10.1016/j.immuni.2019.11.014

    35. Colevas AD, Bahleda R, Braiteh F, Balmanoukian A, Brana I, Chau NG, Sarkar I, Molinero L, Grossman W, Kabbinavar F, Fassò M, O'Hear C, Powderly J (2018) Safety and clinical activity of atezolizumab in head and neck cancer: results from a phase I trial. Ann Oncol 29: 2247–2253
        doi: 10.1093/annonc/mdy411

    36. Conner KL, Shaik AN, Ekinci E, Kim S, Ruterbusch JJ, Cote ML, Patrick SM (2020) HPV induction of APOBEC3 enzymes mediate overall survival and response to cisplatin in head and neck cancer. DNA Repair (amst) 87: 102802
        doi: 10.1016/j.dnarep.2020.102802

    37. Contreras-Paredes A, De la Cruz-Hernández E, Martínez-Ramírez I, Dueñas-González A, Lizano M (2009) E6 variants of human papillomavirus 18 differentially modulate the protein kinase B/phosphatidylinositol 3-kinase (akt/PI3K) signaling pathway. Virology 383: 78–85
        doi: 10.1016/j.virol.2008.09.040

    38. Cramer JD, Burtness B, Ferris RL (2019) Immunotherapy for head and neck cancer: Recent advances and future directions. Oral Oncol 99: 104460
        doi: 10.1016/j.oraloncology.2019.104460

    39. Cui T, Enroth S, Ameur A, Gustavsson I, Lindquist D, Gyllensten U (2019) Invasive cervical tumors with high and low HPV titer represent molecular subgroups with different disease etiology. Carcinogenesis 40: 269–278
        doi: 10.1093/carcin/bgy183

    40. De Cicco R, de Melo MR, Nicolau UR, Pinto CAL, Villa LL, Kowalski LP (2020) Impact of human papillomavirus status on survival and recurrence in a geographic region with a low prevalence of HPV-related cancer: a retrospective cohort study. Head Neck 42: 93–102
        doi: 10.1002/hed.25985

    41. De Felice F, Bird T, Michaelidou A, Thavaraj S, Odell E, Sandison A, Hall G, Morgan P, Lyons A, Cascarini L, Fry A, Oakley R, Simo R, Jeannon JP, Lei M, Guerrero Urbano T (2020) Radical (chemo) radiotherapy in oropharyngeal squamous cell carcinoma: comparison of TNM 7th and 8th staging systems. Radiother Oncol 145: 146–153
        doi: 10.1016/j.radonc.2019.12.021

    42. de Martel C, Georges D, Bray F, Ferlay J, Clifford GM (2020) Global burden of cancer attributable to infections in 2018: a worldwide incidence analysis. Lancet Glob Health 8: e180–e190
        doi: 10.1016/S2214-109X(19)30488-7

    43. Dickstein DR, Egerman MA, Bui AH, Doucette JT, Sharma S, Liu J, Gupta V, Miles BA, Genden E, Westra WH, Misiukiewicz K, Posner MR, Bakst RL (2020) A new face of the HPV epidemic: oropharyngeal cancer in the elderly. Oral Oncol 109: 104687
        doi: 10.1016/j.oraloncology.2020.104687

    44. Dsouza G, Carey TE, William WN Jr, Nguyen ML, Ko EC, Jt R, Pai SI, Gupta V, Walline HM, Lee JJ, Wolf GT, Shin DM, Grandis JR, Ferris RL (2014) Epidemiology of head and neck squamous cell cancer among HIV-infected patients. J Acquir Immune Defic Syndr 65: 603–610
        doi: 10.1097/QAI.0000000000000083

    45. Eckhardt M, Zhang W, Gross AM, Von Dollen J, Johnson JR, Franks-Skiba KE, Swaney DL, Johnson TL, Jang GM, Shah PS, Brand TM, Archambault J, Kreisberg JF, Grandis JR, Ideker T, Krogan NJ (2018) Multiple routes to oncogenesis are promoted by the human papillomavirus-host protein network. Cancer Discov 8: 1474–1489
        doi: 10.1158/2159-8290.CD-17-1018

    46. Elhalawani H, Mohamed ASR, Elgohari B, Lin TA, Sikora AG, Lai SY, Abusaif A, Phan J, Morrison WH, Gunn GB, Rosenthal DI, Garden AS, Fuller CD, Sandulache VC (2020) Tobacco exposure as a major modifier of oncologic outcomes in human papillomavirus (HPV) associated oropharyngeal squamous cell carcinoma. BMC Cancer 20: 912
        doi: 10.1186/s12885-020-07427-7

    47. Facompre ND, Rajagopalan P, Sahu V, Pearson AT, Montone KT, James CD, Gleber-Netto FO, Weinstein GS, Jalaly J, Lin A, Rustgi AK, Nakagawa H, Califano JA, Pickering CR, White EA, Windle BE, Morgan IM, Cohen RB, Gimotty PA, Basu D (2020) Identifying predictors of HPV-related head and neck squamous cell carcinoma progression and survival through patient-derived models. Int J Cancer 147: 3236–3249
        doi: 10.1002/ijc.33125

    48. Fakhry C, Lacchetti C, Rooper LM, Jordan RC, Rischin D, Sturgis EM, Bell D, Lingen MW, Harichand-Herdt S, Thibo J, Zevallos J, Perez-Ordonez B (2018) Human papillomavirus testing in head and neck carcinomas: ASCO clinical practice guideline endorsement of the college of american pathologists guideline. J Clin Oncol 36: 3152–3161
        doi: 10.1200/JCO.18.00684

    49. Fakhry C, Waterboer T, Westra WH, Rooper LM, Windon M, Troy T, Koch W, Gourin CG, Bender N, Yavvari S, Kiess AP, Miles BA, Ryan WR, Ha PK, Eisele DW, D'Souza G (2020) Distinct biomarker and behavioral profiles of human papillomavirus-related oropharynx cancer patients by age. Oral Oncol 101: 104522
        doi: 10.1016/j.oraloncology.2019.104522

    50. Ferris RL, Blumenschein G Jr, Fayette J, Guigay J, Colevas AD, Licitra L, Harrington KJ, Kasper S, Vokes EE, Even C, Worden F, Saba NF, Docampo LCI, Haddad R, Rordorf T, Kiyota N, Tahara M, Lynch M, Jayaprakash V, Li L, Gillison ML (2018) Nivolumab vs investigator's choice in recurrent or metastatic squamous cell carcinoma of the head and neck: 2-year long-term survival update of CheckMate 141 with analyses by tumor PD-L1 expression. Oral Oncol 81: 45–51
        doi: 10.1016/j.oraloncology.2018.04.008

    51. Freitag J, Wald T, Kuhnt T, Gradistanac T, Kolb M, Dietz A, Wiegand S, Wichmann G (2020) Extracapsular extension of neck nodes and absence of human papillomavirus 16-DNA are predictors of impaired survival in p16-positive oropharyngeal squamous cell carcinoma. Cancer 126: 1856–1872
        doi: 10.1002/cncr.32667

    52. Galvis MM, Borges GA, Oliveira TB, Toledo IP, Castilho RM, Guerra ENS, Kowalski LP, Squarize CH (2020) Immunotherapy improves efficacy and safety of patients with HPV positive and negative head and neck cancer: a systematic review and meta-analysis. Crit Rev Oncol Hematol 150: 102966
        doi: 10.1016/j.critrevonc.2020.102966

    53. Gameiro SF, Ghasemi F, Barrett JW, Koropatnick J, Nichols AC, Mymryk JS, Maleki Vareki S (2018) Treatment-naïve HPV+ head and neck cancers display a T-cell-inflamed phenotype distinct from their HPV- counterparts that has implications for immunotherapy. Oncoimmunology 7: e1498439
        doi: 10.1080/2162402X.2018.1498439

    54. Gameiro SF, Ghasemi F, Barrett JW, Nichols AC, Mymryk JS (2019) High level expression of MHC-Ⅱ in HPV+ head and neck cancers suggests that tumor epithelial cells serve an important role as accessory antigen presenting cells. Cancers (basel) 11: 1129
        doi: 10.3390/cancers11081129

    55. Gandhapudi SK, Ward M, Bush JPC, Bedu-Addo F, Conn G, Woodward JG (2019) Antigen priming with enantiospecific cationic lipid nanoparticles induces potent antitumor CTL responses through novel induction of a type Ⅰ IFN response. J Immunol 202: 3524–3536
        doi: 10.4049/jimmunol.1801634

    56. García-Escudero R, Segrelles C, Dueñas M, Pombo M, Ballestín C, Alonso-Riaño M, Nenclares P, Álvarez-Rodríguez R, Sánchez-Aniceto G, Ruíz-Alonso A, López-Cedrún JL, Paramio JM, Lorz C (2018) Overexpression of PIK3CA in head and neck squamous cell carcinoma is associated with poor outcome and activation of the YAP pathway. Oral Oncol 79: 55–63
        doi: 10.1016/j.oraloncology.2018.02.014

    57. Gaździcka J, Gołąbek K, Strzelczyk JK, Ostrowska Z (2020) Epigenetic modifications in head and neck cancer. Biochem Genet 58: 213–244
        doi: 10.1007/s10528-019-09941-1

    58. Gazzaz MJ, Jeffery C, O'Connell D, Harris J, Seikaly H, Biron V (2019) Association of human papillomavirus related squamous cell carcinomas of the oropharynx and cervix. Papillomavirus Res 8: 100188
        doi: 10.1016/j.pvr.2019.100188

    59. Gillison ML, Akagi K, Xiao W, Jiang B, Pickard RKL, Li J, Swanson BJ, Agrawal AD, Zucker M, Stache-Crain B, Emde AK, Geiger HM, Robine N, Coombes KR, Symer DE (2019a) Human papillomavirus and the landscape of secondary genetic alterations in oral cancers. Genome Res 29: 1–17
        doi: 10.1101/gr.241141.118

    60. 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

    61. Gillison ML, Trotti AM, Harris J, Eisbruch A, Harari PM, Adelstein DJ, Jordan RCK, Zhao W, Sturgis EM, Burtness B, Ridge JA, Ringash J, Galvin J, Yao M, Koyfman SA, Blakaj DM, Razaq MA, Colevas AD, Beitler JJ, Jones CU, Dunlap NE, Seaward SA, Spencer S, Galloway TJ, Phan J, Dignam JJ, Le QT (2019b) Radiotherapy plus cetuximab or cisplatin in human papillomavirus-positive oropharyngeal cancer (NRG Oncology RTOG 1016): a randomised, multicentre, non-inferiority trial. Lancet 393: 40–50
        doi: 10.1016/S0140-6736(18)32779-X

    62. Girardi FM, Wagner VP, Martins MD, Abentroth AL, Hauth LA (2020) Prevalence of p16 expression in oropharyngeal squamous cell carcinoma in southern Brazil. Oral Surg Oral Med Oral Pathol Oral Radiol. https://doi.org/10.1016/j.oooo.2020.08.021
        doi: 10.1016/j.oooo.2020.08.021

    63. Grønhøj C, Jensen JS, Wagner S, Dehlendorff C, Friborg J, Andersen E, Wittekindt C, Würdemann N, Sharma SJ, Gattenlöhner S, Klussmann JP, von Buchwald C (2019a) Impact on survival of tobacco smoking for cases with oropharyngeal squamous cell carcinoma and known human papillomavirus and p16-status: a multicenter retrospective study. Oncotarget 10: 4655–4663
        doi: 10.18632/oncotarget.27079

    64. Grønhøj C, Kronberg Jakobsen K, Kjær E, Friborg J, von Buchwald C (2019b) Comorbidity in HPV+ and HPV- oropharyngeal cancer patients: a population-based, case-control study. Oral Oncol 96: 1–6
        doi: 10.1016/j.oraloncology.2019.06.035

    65. Guo T, Zambo KDA, Zamuner FT, Ou T, Hopkins C, Kelley DZ, Wulf HA, Winkler E, Erbe R, Danilova L, Considine M, Sidransky D, Favorov A, Florea L, Fertig EJ, Gaykalova DA (2020) Chromatin structure regulates cancer-specific alternative splicing events in primary HPV-related oropharyngeal squamous cell carcinoma. Epigenetics 15: 959–971
        doi: 10.1080/15592294.2020.1741757

    66. Hall SF, Griffiths RJ, O'Sullivan B, Liu FF (2019) The addition of chemotherapy to radiotherapy did not reduce the rate of distant metastases in low-risk HPV-related oropharyngeal cancer in a real-world setting. Head Neck 41: 2271–2276
        doi: 10.1002/hed.25679

    67. Hammarstedt L, Holzhauser S, Zupancic M, Kapoulitsa F, Ursu RG, Ramqvist T, Haeggblom L, Näsman A, Dalianis T, Marklund L (2020) The value of p16 and HPV DNA in non-tonsillar, non-base of tongue oropharyngeal cancer. Acta Otolaryngol 141: 89–94

    68. Haque SU, Niu L, Kuhnell D, Hendershot J, Biesiada J, Niu W, Hagan MC, Kelsey KT, Casper KA, Wise-Draper TM, Medvedovic M, Langevin SM (2018) Differential expression and prognostic value of long non-coding RNA in HPV-negative head and neck squamous cell carcinoma. Head Neck 40: 1555–1564
        doi: 10.1002/hed.25136

    69. Hatano T, Sano D, Takahashi H, Hyakusoku H, Isono Y, Shimada S, Sawakuma K, Takada K, Oikawa R, Watanabe Y, Yamamoto H, Itoh F, Myers JN, Oridate N (2017) Identification of human papillomavirus (HPV) 16 DNA integration and the ensuing patterns of methylation in HPV-associated head and neck squamous cell carcinoma cell lines. Int J Cancer 140: 1571–1580
        doi: 10.1002/ijc.30589

    70. Henderson S, Chakravarthy A, Su X, Boshoff C, Fenton TR (2014) APOBEC-mediated cytosine deamination links PIK3CA helical domain mutations to human papillomavirus-driven tumor development. Cell Rep 7: 1833–1841
        doi: 10.1016/j.celrep.2014.05.012

    71. Huang SH, O'Sullivan B, Su J, Ringash J, Bratman SV, Kim J, Hosni A, Bayley A, Cho J, Giuliani M, Hope A, Spreafico A, Hansen AR, Siu LL, Gilbert R, Irish JC, Goldstein D, de Almeida J, Tong L, Xu W, Waldron J (2020) Hypofractionated radiotherapy alone with 2.4 Gy per fraction for head and neck cancer during the COVID-19 pandemic: the Princess Margaret experience and proposal. Cancer 126: 3426–3437

    72. Huebbers CU, Verhees F, Poluschkin L, Olthof NC, Kolligs J, Siefer OG, Henfling M, Ramaekers FCS, Preuss SF, Beutner D, Seehawer J, Drebber U, Korkmaz Y, Lam WL, Vucic EA, Kremer B, Klussmann JP, Speel EM (2019) Upregulation of AKR1C1 and AKR1C3 expression in OPSCC with integrated HPV16 and HPV-negative tumors is an indicator of poor prognosis. Int J Cancer 144: 2465–2477
        doi: 10.1002/ijc.31954

    73. Jamieson LM, Antonsson A, Garvey G, Ju X, Smith M, Logan RM, Johnson NW, Hedges J, Sethi S, Dunbar T, Leane C, Hill I, Brown A, Roder D, De Souza M, Canfell K (2020) Prevalence of oral human papillomavirus infection among australian indigenous adults. JAMA Netw Open 3: e204951
        doi: 10.1001/jamanetworkopen.2020.4951

    74. Joseph AW, Ogawa T, Bishop JA, Lyford-Pike S, Chang X, Phelps TH, Westra WH, Pai SI (2013) Molecular etiology of second primary tumors in contralateral tonsils of human papillomavirus-associated index tonsillar carcinomas. Oral Oncol 49: 244–248
        doi: 10.1016/j.oraloncology.2012.09.009

    75. Keck MK, Zuo Z, Khattri A, Stricker TP, Brown CD, Imanguli M, Rieke D, Endhardt K, Fang P, Brägelmann J, DeBoer R, El-Dinali M, Aktolga S, Lei Z, Tan P, Rozen SG, Salgia R, Weichselbaum RR, Lingen MW, Story MD, Ang KK, Cohen EE, White KP, Vokes EE, Seiwert TY (2015) Integrative analysis of head and neck cancer identifies two biologically distinct HPV and three non-HPV subtypes. Clin Cancer Res 21: 870–881
        doi: 10.1158/1078-0432.CCR-14-2481

    76. Kerr DA, Arora KS, Mahadevan KK, Hornick JL, Krane JF, Rivera MN, Ting DT, Deshpande V, Faquin WC (2015) Performance of a branch chain RNA In Situ hybridization assay for the detection of high-risk human papillomavirus in head and neck squamous cell carcinoma. Am J Surg Pathol 39: 1643–1652
        doi: 10.1097/PAS.0000000000000516

    77. Kirnbauer R, Booy F, Cheng N, Lowy DR, Schiller JT (1992) Papillomavirus L1 major capsid protein self-assembles into virus-like particles that are highly immunogenic. Proc Natl Acad Sci U S A 89: 12180–12184
        doi: 10.1073/pnas.89.24.12180

    78. Kofler B, Borena W, Dudas J, Innerhofer V, Dejaco D, Steinbichler TB, Widmann G, von Laer D, Riechelmann H (2020) Post-treatment HPV surface brushings and risk of relapse in oropharyngeal carcinoma. Cancers (basel) 12: 1069
        doi: 10.3390/cancers12051069

    79. Kondo S, Wakae K, Wakisaka N, Nakanishi Y, Ishikawa K, Komori T, Moriyama-Kita M, Endo K, Murono S, Wang Z, Kitamura K, Nishiyama T, Yamaguchi K, Shigenobu S, Muramatsu M, Yoshizaki T (2017) APOBEC3A associates with human papillomavirus genome integration in oropharyngeal cancers. Oncogene 36: 1687–1697
        doi: 10.1038/onc.2016.335

    80. Koneva LA, Zhang Y, Virani S, Hall PB, McHugh JB, Chepeha DB, Wolf GT, Carey TE, Rozek LS, Sartor MA (2018) HPV integration in HNSCC correlates with survival outcomes, immune response signatures, and candidate drivers. Mol Cancer Res 16: 90–102
        doi: 10.1158/1541-7786.MCR-17-0153

    81. Kopczyńska M, Kolenda T, Guglas K, Sobocińska J, Teresiak A, Bliźniak R, Mackiewicz A, Mackiewicz J, Lamperska K (2020) PRINS lncRNA Is a new biomarker candidate for HPV infection and prognosis of head and neck squamous cell carcinomas. Diagnostics (basel) 10: 762
        doi: 10.3390/diagnostics10100762

    82. Kostareli E, Hielscher T, Zucknick M, Baboci L, Wichmann G, Holzinger D, Mücke O, Pawlita M, Del Mistro A, Boscolo-Rizzo P, Da Mosto MC, Tirelli G, Plinkert P, Dietz A, Plass C, Weichenhan D, Hess J (2016) Gene promoter methylation signature predicts survival of head and neck squamous cell carcinoma patients. Epigenetics 11: 61–73
        doi: 10.1080/15592294.2015.1137414

    83. Kostareli E, Holzinger D, Bogatyrova O, Hielscher T, Wichmann G, Keck M, Lahrmann B, Grabe N, Flechtenmacher C, Schmidt CR, Seiwert T, Dyckhoff G, Dietz A, Höfler D, Pawlita M, Benner A, Bosch FX, Plinkert P, Plass C, Weichenhan D, Hess J (2013) HPV-related methylation signature predicts survival in oropharyngeal squamous cell carcinomas. J Clin Invest 123: 2488–2501
        doi: 10.1172/JCI67010

    84. Krishna S, Ulrich P, Wilson E, Parikh F, Narang P, Yang S, Read AK, Kim-Schulze S, Park JG, Posner M, Wilson Sayres MA, Sikora A, Anderson KS (2018) Human papilloma virus specific immunogenicity and dysfunction of CD8(+) T cells in head and neck cancer. Cancer Res 78: 6159–6170
        doi: 10.1158/0008-5472.CAN-18-0163

    85. Kwon S, Ahn SH, Jeong WJ, Jung YH, Bae YJ, Paik JH, Chung JH, Kim H (2020) Estrogen receptor α as a predictive biomarker for survival in human papillomavirus-positive oropharyngeal squamous cell carcinoma. J Transl Med 18: 240
        doi: 10.1186/s12967-020-02396-8

    86. Lang Kuhs KA, Kreimer AR, Trivedi S, Holzinger D, Pawlita M, Pfeiffer RM, Gibson SP, Schmitt NC, Hildesheim A, Waterboer T, Ferris RL (2017) Human papillomavirus 16 E6 antibodies are sensitive for human papillomavirus-driven oropharyngeal cancer and are associated with recurrence. Cancer 123: 4382–4390
        doi: 10.1002/cncr.30966

    87. Law EK, Levin-Klein R, Jarvis MC, Kim H, Argyris PP, Carpenter MA, Starrett GJ, Temiz NA, Larson LK, Durfee C, Burns MB, Vogel RI, Stavrou S, Aguilera AN, Wagner S, Largaespada DA, Starr TK, Ross SR, Harris RS (2020) APOBEC3A catalyzes mutation and drives carcinogenesis in vivo. J Exp Med 217: e20200261
        doi: 10.1084/jem.20200261

    88. Leemans CR, Snijders PJF, Brakenhoff RH (2018) The molecular landscape of head and neck cancer. Nat Rev Cancer 18: 269–282
        doi: 10.1038/nrc.2018.11

    89. Lesseur C, Diergaarde B, Olshan AF, Wünsch-Filho V, Ness AR, Liu G, Lacko M, Eluf-Neto J, Franceschi S, Lagiou P, Macfarlane GJ, Richiardi L, Boccia S, Polesel J, Kjaerheim K, Zaridze D, Johansson M, Menezes AM, Curado MP, Robinson M, Ahrens W, Canova C, Znaor A, Castellsagué X, Conway DI, Holcátová I, Mates D, Vilensky M, Healy CM, Szeszenia-Dąbrowska N, Fabiánová E, Lissowska J, Grandis JR, Weissler MC, Tajara EH, Nunes FD, de Carvalho MB, Thomas S, Hung RJ, Peters WH, Herrero R, Cadoni G, Bueno-de-Mesquita HB, Steffen A, Agudo A, Shangina O, Xiao X, Gaborieau V, Chabrier A, Anantharaman D, Boffetta P, Amos CI, McKay JD, Brennan P (2016) Genome-wide association analyses identify new susceptibility loci for oral cavity and pharyngeal cancer. Nat Genet 48: 1544–1550
        doi: 10.1038/ng.3685

    90. Lewis JS Jr (2020) Human papillomavirus testing in head and neck squamous cell carcinoma in 2020: where are we now and where are we going? Head Neck Pathol 14: 321–329
        doi: 10.1007/s12105-019-01117-y

    91. Lewis JS Jr, Beadle B, Bishop JA, Chernock RD, Colasacco C, Lacchetti C, Moncur JT, Rocco JW, Schwartz MR, Seethala RR, Thomas NE, Westra WH, Faquin WC (2018) Human papillomavirus testing in head and neck carcinomas: guideline from the college of American pathologists. Arch Pathol Lab Med 142: 559–597
        doi: 10.5858/arpa.2017-0286-CP

    92. Lindsay CD, Kostiuk MA, Harris J, O'Connell DA, Seikaly H, Biron VL (2017) Efficacy of EZH2 inhibitory drugs in human papillomavirus-positive and human papillomavirus-negative oropharyngeal squamous cell carcinomas. Clin Epigenetics 9: 95
        doi: 10.1186/s13148-017-0390-y

    93. Liu H, Xu J, Yang Y, Wang X, Wu E, Majerciak V, Zhang T, Steenbergen RDM, Wang HK, Banerjee NS, Li Y, Lu W, Meyers C, Zhu J, Xie X, Chow LT, Zheng ZM (2021) Oncogenic HPV promotes the expression of the long noncoding RNA lnc-FANCI-2 through E7 and YY1. Proc Natl Acad Sci U S A 118: e2014195118
        doi: 10.1073/pnas.2014195118

    94. Liu J, Yang XL, Zhang SW, Zhu LP, Chen WQ (2018) Incidence, mortality, and temporal patterns of oropharyngeal cancer in China: a population-based study. Cancer Commun (lond) 38: 75
        doi: 10.1186/s40880-018-0345-5

    95. Liu X, Liu P, Chernock RD, Kuhs KAL, Lewis JS Jr, Luo J, Gay HA, Thorstad WL, Wang X (2020) A prognostic gene expression signature for oropharyngeal squamous cell carcinoma. EBioMedicine 61: 102805
        doi: 10.1016/j.ebiom.2020.102805

    96. Lui VW, Hedberg ML, Li H, Vangara BS, Pendleton K, Zeng Y, Lu Y, Zhang Q, Du Y, Gilbert BR, Freilino M, Sauerwein S, Peyser ND, Xiao D, Diergaarde B, Wang L, Chiosea S, Seethala R, Johnson JT, Kim S, Duvvuri U, Ferris RL, Romkes M, Nukui T, Kwok-Shing Ng P, Garraway LA, Hammerman PS, Mills GB, Grandis JR (2013) Frequent mutation of the PI3K pathway in head and neck cancer defines predictive biomarkers. Cancer Discov 3: 761–769
        doi: 10.1158/2159-8290.CD-13-0103

    97. Luo M, Wang H, Wang Z, Cai H, Lu Z, Li Y, Du M, Huang G, Wang C, Chen X, Porembka MR, Lea J, Frankel AE, Fu YX, Chen ZJ, Gao J (2017) A STING-activating nanovaccine for cancer immunotherapy. Nat Nanotechnol 12: 648–654
        doi: 10.1038/nnano.2017.52

    98. Luo X, Donnelly CR, Gong W, Heath BR, Hao Y, Donnelly LA, Moghbeli T, Tan YS, Lin X, Bellile E, Kansy BA, Carey TE, Brenner JC, Cheng L, Polverini PJ, Morgan MA, Wen H, Prince ME, Ferris RL, Xie Y, Young S, Wolf GT, Chen Q, Lei YL (2020) HPV16 drives cancer immune escape via NLRX1-mediated degradation of STING. J Clin Invest 130: 1635–1652
        doi: 10.1172/JCI129497

    99. Lyford-Pike S, Peng S, Young GD, Taube JM, Westra WH, Akpeng B, Bruno TC, Richmon JD, Wang H, Bishop JA, Chen L, Drake CG, Topalian SL, Pardoll DM, Pai SI (2013) Evidence for a role of the PD-1: PD-L1 pathway in immune resistance of HPV-associated head and neck squamous cell carcinoma. Cancer Res 73: 1733–1741
        doi: 10.1158/0008-5472.CAN-12-2384

    100. Lyu X, Zhang M, Li G, Jiang Y, Qiao Q (2019) PD-1 and PD-L1 expression predicts radiosensitivity and clinical outcomes in head and neck cancer and is associated with HPV infection. J Cancer 10: 937–948
        doi: 10.7150/jca.27199

    101. Mac M, Moody CA (2020) Epigenetic regulation of the human papillomavirus life cycle. Pathogens 9: 483
        doi: 10.3390/pathogens9060483

    102. Madathil S, Rousseau MC, Joseph L, Coutlée F, Schlecht NF, Franco E, Nicolau B (2020) Latency of tobacco smoking for head and neck cancer among HPV-positive and HPV-negative individuals. Int J Cancer 147: 56–64
        doi: 10.1002/ijc.32708

    103. Marquard FE, Jücker M (2020) PI3K/AKT/mTOR signaling as a molecular target in head and neck cancer. Biochem Pharmacol 172: 113729
        doi: 10.1016/j.bcp.2019.113729

    104. Massarelli E, William W, Johnson F, Kies M, Ferrarotto R, Guo M, Feng L, Lee JJ, Tran H, Kim YU, Haymaker C, Bernatchez C, Curran M, Zecchini Barrese T, Rodriguez Canales J, Wistuba I, Li L, Wang J, van der Burg SH, Melief CJ, Glisson B (2019) Combining immune checkpoint blockade and tumor-specific vaccine for patients with incurable human papillomavirus 16-related cancer: a phase 2 clinical trial. JAMA Oncol 5: 67–73
        doi: 10.1001/jamaoncol.2018.4051

    105. McLaughlin-Drubin ME, Park D, Munger K (2013) Tumor suppressor p16INK4A is necessary for survival of cervical carcinoma cell lines. Proc Natl Acad Sci U S A 110: 16175–16180
        doi: 10.1073/pnas.1310432110

    106. Mehanna H, Robinson M, Hartley A, Kong A, Foran B, Fulton-Lieuw T, Dalby M, Mistry P, Sen M, O'Toole L, Al Booz H, Dyker K, Moleron R, Whitaker S, Brennan S, Cook A, Griffin M, Aynsley E, Rolles M, De Winton E, Chan A, Srinivasan D, Nixon I, Grumett J, Leemans CR, Buter J, Henderson J, Harrington K, McConkey C, Gray A, Dunn J (2019) Radiotherapy plus cisplatin or cetuximab in low-risk human papillomavirus-positive oropharyngeal cancer (De-ESCALaTE HPV): an open-label randomised controlled phase 3 trial. Lancet 393: 51–60
        doi: 10.1016/S0140-6736(18)32752-1

    107. Mesri EA, Feitelson MA, Munger K (2014) Human viral oncogenesis: a cancer hallmarks analysis. Cell Host Microbe 15: 266–282
        doi: 10.1016/j.chom.2014.02.011

    108. 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

    109. Mifsud M, Eskander A, Irish J, Gullane P, Gilbert R, Brown D, de Almeida JR, Urbach DR, Goldstein DP (2017) Evolving trends in head and neck cancer epidemiology: Ontario, Canada 1993–2010. Head Neck 39: 1770–1778
        doi: 10.1002/hed.24829

    110. Miyauchi S, Sanders PD, Guram K, Kim SS, Paolini F, Venuti A, Cohen EEW, Gutkind JS, Califano JA, Sharabi AB (2020) HPV16 E5 mediates resistance to PD-L1 blockade and can be targeted with rimantadine in head and neck cancer. Cancer Res 80: 732–746
        doi: 10.1158/0008-5472.CAN-19-1771

    111. Molony P, Werner R, Martin C, Callanan D, Nauta I, Heideman D, Sheahan P, Heffron C, Feeley L (2020) The role of tumour morphology in assigning HPV status in oropharyngeal squamous cell carcinoma. Oral Oncol 105: 104670
        doi: 10.1016/j.oraloncology.2020.104670

    112. Mourad M, Jetmore T, Jategaonkar AA, Moubayed S, Moshier E, Urken ML (2017) Epidemiological Trends Of Head And Neck Cancer in the United States: A SEER population study. J Oral Maxillofac Surg 75: 2562–2572
        doi: 10.1016/j.joms.2017.05.008

    113. Mroz EA, Tward AD, Pickering CR, Myers JN, Ferris RL, Rocco JW (2013) High intratumor genetic heterogeneity is related to worse outcome in patients with head and neck squamous cell carcinoma. Cancer 119: 3034–3042
        doi: 10.1002/cncr.28150

    114. Ni G, Huang K, Luan Y, Cao Z, Chen S, Ma B, Yuan J, Wu X, Chen G, Wang T, Li H, Walton S, Liu F, Chen B, Wang Y, Pan X, Liu X, Frazer IH (2019) Human papillomavirus infection among head and neck squamous cell carcinomas in southern China. PLoS One 14: e0221045
        doi: 10.1371/journal.pone.022104506|2008|||

    115. Nohata N, Abba MC, Gutkind JS (2016) Unraveling the oral cancer lncRNAome: Identification of novel lncRNAs associated with malignant progression and HPV infection. Oral Oncol 59: 58–66
        doi: 10.1016/j.oraloncology.2016.05.014

    116. Nopmaneepaisarn T, Tangjaturonrasme N, Rawangban W, Vinayanuwattikun C, Keelawat S, Bychkov A (2019) Low prevalence of p16-positive HPV-related head-neck cancers in Thailand: tertiary referral center experience. BMC Cancer 19: 1050
        doi: 10.1186/s12885-019-6266-0

    117. Olthof NC, Huebbers CU, Kolligs J, Henfling M, Ramaekers FC, Cornet I, van Lent-Albrechts JA, Stegmann AP, Silling S, Wieland U, Carey TE, Walline HM, Gollin SM, Hoffmann TK, de Winter J, Kremer B, Klussmann JP, Speel EJ (2015) Viral load, gene expression and mapping of viral integration sites in HPV16-associated HNSCC cell lines. Int J Cancer 136: E207-218
        doi: 10.1002/ijc.29112

    118. Olthof NC, Speel EJ, Kolligs J, Haesevoets A, Henfling M, Ramaekers FC, Preuss SF, Drebber U, Wieland U, Silling S, Lam WL, Vucic EA, Kremer B, Klussmann JP, Huebbers CU (2014) Comprehensive analysis of HPV16 integration in OSCC reveals no significant impact of physical status on viral oncogene and virally disrupted human gene expression. PLoS ONE 9: e88718
        doi: 10.1371/journal.pone.0088718

    119. Outh-Gauer S, Morini A, Tartour E, Lépine C, Jung AC, Badoual C (2020) The microenvironment of head and neck cancers: papillomavirus involvement and potential impact of immunomodulatory treatments. Head Neck Pathol 14: 330–340
        doi: 10.1007/s12105-020-01147-x

    120. Pal A, Kundu R (2019) Human papillomavirus E6 and E7: the cervical cancer hallmarks and targets for therapy. Front Microbiol 10: 3116

    121. Papillon-Cavanagh S, Lu C, Gayden T, Mikael LG, Bechet D, Karamboulas C, Ailles L, Karamchandani J, Marchione DM, Garcia BA, Weinreb I, Goldstein D, Lewis PW, Dancu OM, Dhaliwal S, Stecho W, Howlett CJ, Mymryk JS, Barrett JW, Nichols AC, Allis CD, Majewski J, Jabado N (2017) Impaired H3K36 methylation defines a subset of head and neck squamous cell carcinomas. Nat Genet 49: 180–185
        doi: 10.1038/ng.3757

    122. Parfenov M, Pedamallu CS, Gehlenborg N, Freeman SS, Danilova L, Bristow CA, Lee S, Hadjipanayis AG, Ivanova EV, Wilkerson MD, Protopopov A, Yang L, Seth S, Song X, Tang J, Ren X, Zhang J, Pantazi A, Santoso N, Xu AW, Mahadeshwar H, Wheeler DA, Haddad RI, Jung J, Ojesina AI, Issaeva N, Yarbrough WG, Hayes DN, Grandis JR, El-Naggar AK, Meyerson M, Park PJ, Chin L, Seidman JG, Hammerman PS, Kucherlapati R (2014) Characterization of HPV and host genome interactions in primary head and neck cancers. Proc Natl Acad Sci U S A 111: 15544–15549
        doi: 10.1073/pnas.1416074111

    123. Parikh F, Duluc D, Imai N, Clark A, Misiukiewicz K, Bonomi M, Gupta V, Patsias A, Parides M, Demicco EG, Zhang DY, Kim-Schulze S, Kao J, Gnjatic S, Oh S, Posner MR, Sikora AG (2014) Chemoradiotherapy-induced upregulation of PD-1 antagonizes immunity to HPV-related oropharyngeal cancer. Cancer Res 74: 7205–7216
        doi: 10.1158/0008-5472.CAN-14-1913

    124. Pickering CR, Zhang J, Yoo SY, Bengtsson L, Moorthy S, Neskey DM, Zhao M, Ortega Alves MV, Chang K, Drummond J, Cortez E, Xie TX, Zhang D, Chung W, Issa JP, Zweidler-McKay PA, Wu X, El-Naggar AK, Weinstein JN, Wang J, Muzny DM, Gibbs RA, Wheeler DA, Myers JN, Frederick MJ (2013) Integrative genomic characterization of oral squamous cell carcinoma identifies frequent somatic drivers. Cancer Discov 3: 770–781
        doi: 10.1158/2159-8290.CD-12-0537

    125. Pim D, Massimi P, Dilworth SM, Banks L (2005) Activation of the protein kinase B pathway by the HPV-16 E7 oncoprotein occurs through a mechanism involving interaction with PP2A. Oncogene 24: 7830–7838
        doi: 10.1038/sj.onc.1208935

    126. Puram SV, Tirosh I, Parikh AS, Patel AP, Yizhak K, Gillespie S, Rodman C, Luo CL, Mroz EA, Emerick KS, Deschler DG, Varvares MA, Mylvaganam R, Rozenblatt-Rosen O, Rocco JW, Faquin WC, Lin DT, Regev A, Bernstein BE (2017) Single-cell transcriptomic analysis of primary and metastatic tumor ecosystems in head and neck cancer. Cell 171: 1611-1624. e1624
        doi: 10.1016/j.cell.2017.10.044

    127. Qi Z, Barrett T, Parikh AS, Tirosh I, Puram SV (2019) Single-cell sequencing and its applications in head and neck cancer. Oral Oncol 99: 104441
        doi: 10.1016/j.oraloncology.2019.104441

    128. Qian X, Kaufmann AM, Chen C, Tzamalis G, Hofmann VM, Keilholz U, Hummel M, Albers AE (2016) Prevalence and associated survival of high-risk HPV-related adenoid cystic carcinoma of the salivary glands. Int J Oncol 49: 803–811
        doi: 10.3892/ijo.2016.3563

    129. Qian X, Leonard F, Wenhao Y, Sudhoff H, Hoffmann TK, Ferrone S, Kaufmann AM, Albers AE (2020) Immunotherapeutics for head and neck squamous cell carcinoma stem cells. HNO 68: 94–99
        doi: 10.1007/s00106-020-00819-y

    130. Qian X, Nguyen DT, Dong Y, Sinikovic B, Kaufmann AM, Myers JN, Albers AE, Graviss EA (2019) Prognostic score predicts survival in HPV-negative head and neck squamous cell cancer patients. Int J Biol Sci 15: 1336–1344
        doi: 10.7150/ijbs.33329

    131. Qin Y, Ekmekcioglu S, Forget MA, Szekvolgyi L, Hwu P, Grimm EA, Jazaeri AA, Roszik J (2017) Cervical cancer neoantigen landscape and immune activity is associated with human papillomavirus master regulators. Front Immunol 8: 689
        doi: 10.3389/fimmu.2017.00689

    132. Rasmussen JH, Grønhøj C, Håkansson K, Friborg J, Andersen E, Lelkaitis G, Klussmann JP, Wittekindt C, Wagner S, Vogelius IR, von Buchwald C (2019) Risk profiling based on p16 and HPV DNA more accurately predicts location of disease relapse in patients with oropharyngeal squamous cell carcinoma. Ann Oncol 30: 629–636
        doi: 10.1093/annonc/mdz010

    133. Ravanelli M, Grammatica A, Maddalo M, Ramanzin M, Agazzi GM, Tononcelli E, Battocchio S, Bossi P, Vezzoli M, Maroldi R, Farina D (2020) Pretreatment DWI with histogram analysis of the ADC in predicting the outcome of advanced oropharyngeal cancer with known human papillomavirus status treated with chemoradiation. AJNR Am J Neuroradiol 41: 1473–1479
        doi: 10.3174/ajnr.A6695

    134. Reid P, Marcu LG, Olver I, Moghaddasi L, Staudacher AH, Bezak E (2019) Diversity of cancer stem cells in head and neck carcinomas: the role of HPV in cancer stem cell heterogeneity, plasticity and treatment response. Radiother Oncol 135: 1–12
        doi: 10.1016/j.radonc.2019.02.016

    135. Ren J, Yang W, Su J, Ren X, Fazelzad R, Albert T, Habbous S, Goldstein DP, de Almeida JR, Hansen A, Jang R, Bratman SV, Hope A, Chen R, Wang J, Xu Y, Cheng D, Zhao Y, Xu W, Liu G (2019) Human papillomavirus and p16 immunostaining, prevalence and prognosis of squamous carcinoma of unknown primary in the head and neck region. Int J Cancer 145: 1465–1474
        doi: 10.1002/ijc.32164

    136. Ren S, Gaykalova D, Wang J, Guo T, Danilova L, Favorov A, Fertig E, Bishop J, Khan Z, Flam E, Wysocki PT, DeJong P, Ando M, Liu C, Sakai A, Fukusumi T, Haft S, Sadat S, Califano JA (2018) Discovery and development of differentially methylated regions in human papillomavirus-related oropharyngeal squamous cell carcinoma. Int J Cancer 143: 2425–2436
        doi: 10.1002/ijc.31778

    137. Rettig EM, Gooi Z, Bardin R, Bogale M, Rooper L, Acha E, Koch WM (2019) Oral Human Papillomavirus Infection and Head and Neck Squamous Cell Carcinoma in Rural Northwest Cameroon. OTO Open 3: 2473974x18818415

    138. Roman A, Munger K (2013) The papillomavirus E7 proteins. Virology 445: 138–168
        doi: 10.1016/j.virol.2013.04.013

    139. Rotsides JM, Oliver JR, Moses LE, Tam M, Li Z, Schreiber D, Jacobson AS, Hu KS, Givi B (2020) Socioeconomic and racial disparities and survival of human papillomavirus-associated oropharyngeal squamous cell carcinoma. Otolaryngol Head Neck Surg 164: 131–138

    140. Ruicci KM, Meens J, Plantinga P, Stecho W, Pinto N, Yoo J, Fung K, MacNeil D, Mymryk JS, Barrett JW, Howlett CJ, Boutros PC, Ailles L, Nichols AC (2020) TAM family receptors in conjunction with MAPK signalling are involved in acquired resistance to PI3Kα inhibition in head and neck squamous cell carcinoma. J Exp Clin Cancer Res 39: 217
        doi: 10.1186/s13046-020-01713-9

    141. Ruicci KM, Meens J, Sun RX, Rizzo G, Pinto N, Yoo J, Fung K, MacNeil D, Mymryk JS, Barrett JW, Boutros PC, Ailles L, Nichols AC (2019a) A controlled trial of HNSCC patient-derived xenografts reveals broad efficacy of PI3Kα inhibition in controlling tumor growth. Int J Cancer 145: 2100–2106
        doi: 10.1002/ijc.32009

    142. Ruicci KM, Plantinga P, Pinto N, Khan MI, Stecho W, Dhaliwal SS, Yoo J, Fung K, MacNeil D, Mymryk JS, Barrett JW, Howlett CJ, Nichols AC (2019b) Disruption of the RICTOR/mTORC2 complex enhances the response of head and neck squamous cell carcinoma cells to PI3K inhibition. Mol Oncol 13: 2160–2177
        doi: 10.1002/1878-0261.12558

    143. Schiller JT, Castellsagué X, Garland SM (2012) A review of clinical trials of human papillomavirus prophylactic vaccines. Vaccine 30(Suppl 5): F123-138

    144. Schroeder L, Pring M, Ingarfield K, Pawlita M, Leary SD, Thomas SJ, Waylen A, Waterboer T, Ness AR (2020) HPV driven squamous cell head and neck cancer of unknown primary is likely to be HPV driven squamous cell oropharyngeal cancer. Oral Oncol 107: 104721
        doi: 10.1016/j.oraloncology.2020.104721

    145. Seiwert TY, Burtness B, Mehra R, Weiss J, Berger R, Eder JP, Heath K, McClanahan T, Lunceford J, Gause C, Cheng JD, Chow LQ (2016) Safety and clinical activity of pembrolizumab for treatment of recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-012): an open-label, multicentre, phase 1b trial. Lancet Oncol 17: 956–965
        doi: 10.1016/S1470-2045(16)30066-3

    146. Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A (2017) Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 168: 707–723
        doi: 10.1016/j.cell.2017.01.017

    147. Shen S, Saito Y, Ren S, Liu C, Guo T, Qualliotine J, Khan Z, Sadat S, Califano JA (2020) Targeting viral DNA and promoter hypermethylation in salivary rinses for recurrent HPV-positive oropharyngeal cancer. Otolaryngol Head Neck Surg 162: 512–519
        doi: 10.1177/0194599820903031

    148. Sievert M, Goncalves M, Mueller SK, Hecht M, Pflaum A, Grundtner P, Iro H, Gostian AO (2020) Impact of delayed adjuvant therapy after surgery in p16 positive oropharyngeal cancer: a retrospective analysis. Eur Rev Med Pharmacol Sci 24: 1211–1218

    149. Smith EM, Pawlita M, Rubenstein LM, Haugen TH, Hamsikova E, Turek LP (2010) Risk factors and survival by HPV-16 E6 and E7 antibody status in human papillomavirus positive head and neck cancer. Int J Cancer 127: 111–117
        doi: 10.1002/ijc.25015

    150. Smith EM, Ritchie JM, Summersgill KF, Klussmann JP, Lee JH, Wang D, Haugen TH, Turek LP (2004) Age, sexual behavior and human papillomavirus infection in oral cavity and oropharyngeal cancers. Int J Cancer 108: 766–772
        doi: 10.1002/ijc.11633

    151. Song L, Xie H, Tong F, Yan B, Zhang S, Fu E, Jing Q, Wei L (2019) Association of lnc-IL17RA-11 with increased radiation sensitivity and improved prognosis of HPV-positive HNSCC. J Cell Biochem 120: 17438–17448
        doi: 10.1002/jcb.29008

    152. Stevanović S, Pasetto A, Helman SR, Gartner JJ, Prickett TD, Howie B, Robins HS, Robbins PF, Klebanoff CA, Rosenberg SA, Hinrichs CS (2017) Landscape of immunogenic tumor antigens in successful immunotherapy of virally induced epithelial cancer. Science 356: 200–205
        doi: 10.1126/science.aak9510

    153. Stransky N, Egloff AM, Tward AD, Kostic AD, Cibulskis K, Sivachenko A, Kryukov GV, Lawrence MS, Sougnez C, McKenna A, Shefler E, Ramos AH, Stojanov P, Carter SL, Voet D, Cortés ML, Auclair D, Berger MF, Saksena G, Guiducci C, Onofrio RC, Parkin M, Romkes M, Weissfeld JL, Seethala RR, Wang L, Rangel-Escareño C, Fernandez-Lopez JC, Hidalgo-Miranda A, Melendez-Zajgla J, Winckler W, Ardlie K, Gabriel SB, Meyerson M, Lander ES, Getz G, Golub TR, Garraway LA, Grandis JR (2011) The mutational landscape of head and neck squamous cell carcinoma. Science 333: 1157–1160
        doi: 10.1126/science.1208130

    154. Strober W, Shishido S, Wood B, Lewis JS Jr, Kuhs K, Ferris RL, Faden DL (2020) Two for the price of one: prevalence, demographics and treatment implications of multiple HPV mediated Head and Neck Cancers. Oral Oncol 100: 104475
        doi: 10.1016/j.oraloncology.2019.104475

    155. Suh CH, Lee KH, Choi YJ, Chung SR, Baek JH, Lee JH, Yun J, Ham S, Kim N (2020) Oropharyngeal squamous cell carcinoma: radiomic machine-learning classifiers from multiparametric MR images for determination of HPV infection status. Sci Rep 10: 17525
        doi: 10.1038/s41598-020-74479-x

    156. Surviladze Z, Sterk RT, DeHaro SA, Ozbun MA (2013) Cellular entry of human papillomavirus type 16 involves activation of the phosphatidylinositol 3-kinase/Akt/mTOR pathway and inhibition of autophagy. J Virol 87: 2508–2517
        doi: 10.1128/JVI.02319-12

    157. Tang KD, Vasani S, Menezes L, Taheri T, Walsh LJ, Hughes BGM, Frazer IH, Kenny L, Scheper GC, Punyadeera C (2020) Oral HPV16 DNA as a screening tool to detect early oropharyngeal squamous cell carcinoma. Cancer Sci 111: 3854–3861
        doi: 10.1111/cas.14585

    158. Tang S, Tao M, McCoy JP Jr, Zheng ZM (2006) The E7 oncoprotein is translated from spliced E6*I transcripts in high-risk human papillomavirus type 16- or type 18-positive cervical cancer cell lines via translation reinitiation. J Virol 80: 4249–4263
        doi: 10.1128/JVI.80.9.4249-4263.2006

    159. The Cancer Genome Atlas Network (2015) Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature 517: 576–582
        doi: 10.1038/nature14129

    160. Tinhofer I, Jöhrens K, Keilholz U, Kaufmann A, Lehmann A, Weichert W, Stenzinger A, Stromberger C, Klinghammer K, Becker ET, Dommerich S, Stölzel K, Hofmann VM, Hildebrandt B, Moser L, Ervens J, Böttcher A, Albers A, Stabenow R, Reinecke A, Budach V, Hoffmeister B, Raguse JD (2015) Contribution of human papilloma virus to the incidence of squamous cell carcinoma of the head and neck in a European population with high smoking prevalence. Eur J Cancer 51: 514–521
        doi: 10.1016/j.ejca.2014.12.018

    161. Tumban E (2019) A current update on human papillomavirus-associated head and neck cancers. Viruses 11: 922
        doi: 10.3390/v11100922

    162. Vande Pol SB, Klingelhutz AJ (2013) Papillomavirus E6 oncoproteins. Virology 445: 115–137
        doi: 10.1016/j.virol.2013.04.026

    163. Ventz S, Trippa L, Schoenfeld JD (2019) Lessons learned from deescalation trials in favorable risk HPV-associated squamous cell head and neck cancer-a perspective on future trial designs. Clin Cancer Res 25: 7281–7286
        doi: 10.1158/1078-0432.CCR-19-0945

    164. Viros Porcuna D, Pollan Guisasola C, Viña Soria C, Cirauqui Cirauqui B, Pardo Muñoz L, Collurá F, Mesia Nin R (2020) Transoral robotic surgery for squamous cell carcinoma of the oropharynx in a primarily human papillomavirus-negative patient population. Clin Transl Oncol 22: 1303–1311
        doi: 10.1007/s12094-019-02256-y

    165. Wagner S, Prigge ES, Wuerdemann N, Reder H, Bushnak A, Sharma SJ, Obermueller T, von Knebel DM, Dreyer T, Gattenlöhner S, Wolf G, Pons-Kühnemann J, Wittekindt C, Klussmann JP (2020) Evaluation of p16(INK4a) expression as a single marker to select patients with HPV-driven oropharyngeal cancers for treatment de-escalation. Br J Cancer 123: 1114–1122
        doi: 10.1038/s41416-020-0964-x

    166. Wakabayashi R, Nakahama Y, Nguyen V, Espinoza JL (2019) The Host-Microbe Interplay in Human Papillomavirus-Induced Carcinogenesis. Microorganisms 7: 199
        doi: 10.3390/microorganisms7070199

    167. Wallace NA, Münger K (2018) The curious case of APOBEC3 activation by cancer-associated human papillomaviruses. PLoS Pathog 14: e1006717
        doi: 10.1371/journal.ppat.1006717

    168. Walline HM, Komarck CM, McHugh JB, Bellile EL, Brenner JC, Prince ME, McKean EL, Chepeha DB, Wolf GT, Worden FP, Bradford CR, Carey TE (2016) Genomic integration of high-risk HPV alters gene expression in oropharyngeal squamous cell carcinoma. Mol Cancer Res 14: 941–952
        doi: 10.1158/1541-7786.MCR-16-0105

    169. Wang X, Liu H, Ge H, Ajiro M, Sharma NR, Meyers C, Morozov P, Tuschl T, Klar A, Court D, Zheng ZM (2017) Viral DNA replication orientation and hnRNPs regulate transcription of the human papillomavirus 18 late promoter. Mbio 8: e00713–e00717
        doi: 10.1128/mBio.00713-17

    170. Wang X, Meyers C, Wang HK, Chow LT, Zheng ZM (2011) Construction of a full transcription map of human papillomavirus type 18 during productive viral infection. J Virol 85: 8080–8092
        doi: 10.1128/JVI.00670-11

    171. Wang X, Wang HK, Li Y, Hafner M, Banerjee NS, Tang S, Briskin D, Meyers C, Chow LT, Xie X, Tuschl T, Zheng ZM (2014) microRNAs are biomarkers of oncogenic human papillomavirus infections. Proc Natl Acad Sci U S A 111: 4262–4267
        doi: 10.1073/pnas.1401430111

    172. Ward MJ, Thirdborough SM, Mellows T, Riley C, Harris S, Suchak K, Webb A, Hampton C, Patel NN, Randall CJ, Cox HJ, Jogai S, Primrose J, Piper K, Ottensmeier CH, King EV, Thomas GJ (2014) Tumour-infiltrating lymphocytes predict for outcome in HPV-positive oropharyngeal cancer. Br J Cancer 110: 489–500
        doi: 10.1038/bjc.2013.639

    173. Weiss BG, Anczykowski MZ, Küffer S, Spiegel JL, Bertlich M, Canis M, Ihler F, Kitz J, Jakob M (2020) Prognostic impact of additional HPV diagnostics in 102 patients with p16-stratified advanced oropharyngeal squamous cell carcinoma. Eur Arch Otorhinolaryngol Undefined: https://doi.org/10.1007/s00405-020-06262-7
        doi: 10.1007/s00405-020-06262-7

    174. Welters MJP, Ma W, Santegoets S, Goedemans R, Ehsan I, Jordanova ES, van Ham VJ, van Unen V, Koning F, van Egmond SI, Charoentong P, Trajanoski Z, van der Velden LA, van der Burg SH (2018) Intratumoral HPV16-specific T cells constitute a type Ⅰ-oriented tumor microenvironment to improve survival in HPV16-driven oropharyngeal cancer. Clin Cancer Res 24: 634–647
        doi: 10.1158/1078-0432.CCR-17-2140

    175. White R, Abel S, Hasan S, Verma V, Greenberg L, Colonias A, Wegner RE (2020) Practice patterns and outcomes following radiation dose de-escalation for oropharyngeal cancer. Laryngoscope 130: E171-e176
        doi: 10.1002/lary.27897

    176. Wichmann G, Rosolowski M, Krohn K, Kreuz M, Boehm A, Reiche A, Scharrer U, Halama D, Bertolini J, Bauer U, Holzinger D, Pawlita M, Hess J, Engel C, Hasenclever D, Scholz M, Ahnert P, Kirsten H, Hemprich A, Wittekind C, Herbarth O, Horn F, Dietz A, Loeffler M (2015) The role of HPV RNA transcription, immune response-related gene expression and disruptive TP53 mutations in diagnostic and prognostic profiling of head and neck cancer. Int J Cancer 137: 2846–2857
        doi: 10.1002/ijc.29649

    177. Wiegand S, Wichmann G, Golusinski W, Leemans CR, Klussmann JP, Dietz A (2018) Highlights from the Second International Symposium on HPV infection in head and neck cancer. Eur Arch Otorhinolaryngol 275: 1365–1373
        doi: 10.1007/s00405-018-4954-z

    178. Wieland A, Patel MR, Cardenas MA, Eberhardt CS, Hudson WH, Obeng RC, Griffith CC, Wang X, Chen ZG, Kissick HT, Saba NF, Ahmed R (2020) Defining HPV-specific B cell responses in patients with head and neck cancer. Nature Undefined: https://doi.org/10.1038/s41586-020-2931-3
        doi: 10.1038/s41586-020-2931-3

    179. Wittekindt C, Wagner S, Bushnak A, Prigge ES, von Knebel DM, Würdemann N, Bernhardt K, Pons-Kühnemann J, Maulbecker-Armstrong C, Klussmann JP (2019) Increasing incidence rates of oropharyngeal squamous cell carcinoma in germany and significance of disease burden attributed to human papillomavirus. Cancer Prev Res (phila) 12: 375–382
        doi: 10.1158/1940-6207.CAPR-19-0098

    180. Wittekindt C, Wagner S, Sharma SJ, Würdemann N, Knuth J, Reder H, Klußmann JP (2018) HPV - a different view on Head and Neck Cancer. Laryngorhinootologie 97: S48-s113
        doi: 10.1055/s-0043-121596

    181. Xu Q, Fang M, Zhu J, Dong H, Cao J, Yan L, Leonard F, Oppel F, Sudhoff H, Kaufmann AM, Albers AE, Qian X (2020a) Insights into Nanomedicine for Immunotherapeutics in Squamous Cell Carcinoma of the head and neck. Int J Biol Sci 16: 2506–2517
        doi: 10.7150/ijbs.47068

    182. Xu S, Sun B, Zhou R, Shi C, Han Y, Li J, Sun J, Tian Z, Zhang Z, Li J (2020b) Evaluation of p16 as a surrogate marker for transcriptionally active human papillomavirus status of oropharyngeal squamous cell carcinoma in an eastern Chinese population. Oral Surg Oral Med Oral Pathol Oral Radiol 129: 236-245. e232
        doi: 10.1016/j.oooo.2019.11.008

    183. Xu T, Shen C, Wei Y, Hu C, Wang Y, Xiang J, Sun GH, Su F, Wang Q, Lu X (2020c) Human papillomavirus (HPV) in Chinese oropharyngeal squamous cell carcinoma (OPSCC): a strong predilection for the tonsil. Cancer Med 9: 6556–6564
        doi: 10.1002/cam4.3339

    184. Xue XY, Majerciak V, Uberoi A, Kim BH, Gotte D, Chen X, Cam M, Lambert PF, Zheng ZM (2017) The full transcription map of mouse papillomavirus type 1 (MmuPV1) in mouse wart tissues. PLoS Pathog 13: e1006715
        doi: 10.1371/journal.ppat.1006715

    185. Yang JQ, Wu M, Han FY, Sun YM, Zhang L, Liu HX (2020) High risk HPV detection by RNAscope in situ hybridization combined with Cdc2 protein expression by immunohistochemistry for prognosis of oropharyngeal squamous cell carcinoma. Int J Clin Exp Pathol 13: 2192–2200

    186. Yang W, Lu YP, Yang YZ, Kang JR, Jin YD, Wang HW (2017) Expressions of programmed death (PD)-1 and PD-1 ligand (PD-L1) in cervical intraepithelial neoplasia and cervical squamous cell carcinomas are of prognostic value and associated with human papillomavirus status. J Obstet Gynaecol Res 43: 1602–1612
        doi: 10.1111/jog.13411

    187. Yang W, Song Y, Lu YL, Sun JZ, Wang HW (2013) Increased expression of programmed death (PD)-1 and its ligand PD-L1 correlates with impaired cell-mediated immunity in high-risk human papillomavirus-related cervical intraepithelial neoplasia. Immunology 139: 513–522
        doi: 10.1111/imm.12101

    188. Young RJ, Bressel M, Porceddu S, Cernelc J, Savas P, Liu H, Urban D, Thai AA, Cooper C, Fua T, Neeson P, Rischin D, Solomon B (2020a) Validation and characterisation of prognostically significant PD-L1(+) immune cells in HPV+ oropharyngeal squamous cell carcinoma. Oral Oncol 101: 104516
        doi: 10.1016/j.oraloncology.2019.104516

    189. Young RJ, Solomon B, Corry J, Angel C, Kenny L, Porceddu S, Wratten C, Macann A, Jackson JE, Herschtal A, Rischin D (2020b) Validation of local p16 testing for determination of human papilloma virus status eligibility on a low risk oropharyngeal cancer trial - A Trans-Tasman Radiation Oncology Group study. Oral Oncol 110: 104988
        doi: 10.1016/j.oraloncology.2020.104988

    190. Zamani M, Grønhøj C, Jensen DH, Carlander AF, Agander T, Kiss K, Olsen C, Baandrup L, Nielsen FC, Andersen E, Friborg J, von Buchwald C (2020) The current epidemic of HPV-associated oropharyngeal cancer: an 18-year Danish population-based study with 2, 169 patients. Eur J Cancer 134: 52–59
        doi: 10.1016/j.ejca.2020.04.027

    191. Zapatka M, Borozan I, Brewer DS, Iskar M, Grundhoff A, Alawi M, Desai N, Sültmann H, Moch H, Cooper CS, Eils R, Ferretti V, Lichter P (2020) The landscape of viral associations in human cancers. Nat Genet 52: 320–330
        doi: 10.1038/s41588-019-0558-9

    192. Zhang J, Chen T, Yang X, Cheng H, Späth SS, Clavijo PE, Chen J, Silvin C, Issaeva N, Su X, Yarbrough WG, Annunziata CM, Chen Z, Van Waes C (2018) Attenuated TRAF3 fosters activation of alternative NF-κB and reduced expression of antiviral interferon, TP53, and RB to promote HPV-positive head and neck cancers. Cancer Res 78: 4613–4626
        doi: 10.1158/0008-5472.CAN-17-0642

    193. Zheng ZM, Baker CC (2006) Papillomavirus genome structure, expression, and post-transcriptional regulation. Front Biosci 11: 2286–2302
        doi: 10.2741/1971

    194. Zheng ZM, Tao M, Yamanegi K, Bodaghi S, Xiao W (2004) Splicing of a cap-proximal human Papillomavirus 16 E6E7 intron promotes E7 expression, but can be restrained by distance of the intron from its RNA 5' cap. J Mol Biol 337: 1091–1108
        doi: 10.1016/j.jmb.2004.02.023

    195. Zheng ZM, Wang X (2011) Regulation of cellular miRNA expression by human papillomaviruses. Biochim Biophys Acta 1809: 668–677
        doi: 10.1016/j.bbagrm.2011.05.005

    196. Zhou C, Tuong ZK, Frazer IH (2019) Papillomavirus immune evasion strategies target the infected cell and the local immune system. Front Oncol 9: 682
        doi: 10.3389/fonc.2019.00682

    197. Zhou G, Liu Z, Myers JN (2016) TP53 mutations in head and neck squamous cell carcinoma and their impact on disease progression and treatment response. J Cell Biochem 117: 2682–2692
        doi: 10.1002/jcb.25592

    198. Zhou J, Sun XY, Stenzel DJ, Frazer IH (1991) Expression of vaccinia recombinant HPV 16 L1 and L2 ORF proteins in epithelial cells is sufficient for assembly of HPV virion-like particles. Virology 185: 251–257
        doi: 10.1016/0042-6822(91)90772-4

    199. Zubair F, McMahon J, Afzali P, Cuschieri K, Swee Yan Y, Schipani S, Brands M, Ansell M (2020) Staging and treatment outcomes in oropharyngeal squamous cell carcinoma: a single-centre UK cohort. Br J Oral Maxillofac Surg 59: 28–34
        doi: 10.1016/j.bjoms.2020.07.007

  • 加载中

Figures(2) / Tables(1)

Article Metrics

Article views(11537) PDF downloads(35) Cited by(0)

Related
Proportional views
    通讯作者: 陈斌, bchen63@163.com
    • 1. 

      沈阳化工大学材料科学与工程学院 沈阳 110142

    1. 本站搜索
    2. 百度学术搜索
    3. 万方数据库搜索
    4. CNKI搜索

    Current Status of Human Papillomavirus-Related Head and Neck Cancer: From Viral Genome to Patient Care

      Corresponding author: Xu Qian, Qian_michelle2014@163.com
    • 1. Department of Clinical Laboratory, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
    • 2. Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
    • 3. Department of Cancer Prevention, Cancer Hospital University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
    • 4. Clinic for Gynecology, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin, 12203, Germany
    • 5. Tumor Virus RNA Biology Section, HIV Dynamics and Replication Program, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
    • 6. Department of Otolaryngology, Head and Neck Surgery, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin, 13353, Germany

    Abstract: 

    Human papillomavirus (HPV) infection identified as a definitive human carcinogen is increasingly being recognized for its role in carcinogenesis of human cancers. Up to 38%–80% of head and neck squamous cell carcinoma (HNSCC) in oropharyngeal location (OPSCC) and nearly all cervical cancers contain the HPV genome which is implicated in causing cancer through its oncoproteins E6 and E7. Given by the biologically distinct HPV-related OPSCC and a more favorable prognosis compared to HPV-negative tumors, clinical trials on de-escalation treatment strategies for these patients have been studied. It is therefore raised the questions for the patient stratification if treatment de-escalation is feasible. Moreover, understanding the crosstalk of HPV-mediated malignancy and immunity with clinical insights from the proportional response rate to immune checkpoint blockade treatments in patients with HNSCC is of importance to substantially improve the treatment efficacy. This review discusses the biology of HPV-related HNSCC as well as successful clinically findings with promising candidates in the pipeline for future directions. With the advent of various sequencing technologies, further biomolecules associated with HPV-related HNSCC progression are currently being identified to be used as potential biomarkers or targets for clinical decisions throughout the continuum of cancer care.

    • High-risk human papillomavirus (HR-HPV) infection leads to the development of human cancers in a variety of anatomical squamous tissue sites, including the head and neck regions. The incidence of HPV-related head and neck squamous cell carcinoma (HNSCC), especially oropharyngeal squamous cell carcinoma (OPSCC), has increased in the United States and Europe (Chaturvedi et al. 2013; Tinhofer et al. 2015; Wittekindt et al. 2019; Zamani et al. 2020). In some regions, HPV-related OPSCC accounts for 38%–80% of OPSCCs and 30% of HNSCCs (Boscolo-Rizzo et al. 2013; Chaturvedi et al. 2013; de Martel et al. 2020; Leemans et al. 2018) (Table 1). In particular, patients with HPV-related OPSCC have a better prognosis than those with HPV-negative OPSCC (Leemans et al. 2018). The proportion of comparatively younger patients is high among patients with HPV-related OPSCC (Leemans et al. 2018). Moreover, HPV-related OPSCCs are more sensitive to chemoradiotherapy and immune checkpoint blockade (ICB) treatments than HPV-unrelated tumors (Leemans et al. 2018). With the accumulated understanding of the new entity of HPV-related OPSCC, a new staging system in the 8th edition of the American Joint Committee on Cancer (AJCC) has been established toward a deintensified treatment protocol to reduce long-term associated morbidity for patients with HPV-related OPSCC. However, recent phase Ⅲ clinical trials (RTOG 1016 and De-ESCALaTE) have shown that a number of patients with HPV-related OPSCC who received deintensified treatment had an inferior outcome compared to those who received standard care (Gillison et al. 2019). Moreover, patients with HPV-related HNSCC at multiple sites, defined as one HPV-positive primary OPSCC and a second primary of any head and neck site, demonstrated distinct characteristics (i.e., a lower T and N stage) compared to patients with one primary tumor (Joseph et al. 2013; Strober et al. 2020). These observations lead to a further discussion on the feasibility of the current design of the de-escalation treatment protocol in clinical trials and highlight the necessity to refine the stratification strategy for patients with HPV-related HNSCC, in particular for OPSCC (Ventz et al. 2019).

      Time interval of collected data Samples Men/women Median Age HPV genotypes Detection method HPV + rate References
      China
      2012–2018 49 29/20 53 16, 18, 52 HPV 16/18 RNA ISH 28.6% Yang et al. (2020)
      2007–2019 152 127/25 NA 11, 16, 18, 33, 53, 58 HPV genotyping 65.1% Xu et al. (2020c)
      2014–2019 257 221/36 60 NA HPV Genotyping 18.3% Xu et al. (2020b)
      1999–2013 300 273/27 54 16, 33, 35, 56, 58, 68 HPV Genotyping and/or p16 IHC 25.0% Chen et al. (2020)
      USA
      2010–2014 1168 926/242 61 NA HPV 16/18 DNA ISH and/or p16 IHC 52.8% White et al. (2020)
      2010–2016 45, 940 38, 0381/7902 60 16, 18, 31, 33, 35, 36, 45, 51, 52, 56, 58, 59, 68, 26, 53, 66, 67, 69, 70, 73, 82, 85 HPV 16/18 DNA ISH and/or p16 IHC 67.5% Rotsides et al. (2020)
      1996–2013 115 101/14 56 NA p16 IHC 90.4% Altenhofen et al. (2020)
      NA 381 332/49 NA NA HPV RNA 79.8% Liu et al. (2020)
      2002–2013 611 517/94 NA NA HPV ISH and p16 IHC 89.0% Elhalawani et al. (2020)
      2007–2018 88 69/19 73 16, 18, 33, 35 PCR amplification of HPV gene loci or p16 IHC 70.5% Dickstein et al. (2020)
      UK
      2010–2016 273 207/66 59 NA p16 IHC 73.3% De Felice et al. (2020)
      Canada
      2005–2017 2039 1668/371 NA NA p16-IHC 48.7% Huang et al. (2020)
      1997–2015 372 0/372a NA NA p16-IHC 56.8% Gazzaz et al. (2019)
      1998–2004 525 381/144 NA NA p16-IHC, HPV DNA ISH 73.5% Hall et al. (2019)
      Australia
      2016–2017 650 284/366 52 16 HPV16 DNA 1.8% Tang et al. (2020)
      2018–2019 910 315/595 37 13, 16, 18, 32 HPV DNA 35.3% Jamieson et al. (2020)
      Australia and New Zealand
      NA 189 NA NA 16, 18 P16 IHC and HPVRNA ISH 88.1% Young et al.(2020a, 2020b)
      Netherlands
      2009–2016 216 143/73 NA NA p16 IHC and HPV DNA 31.9% Chargi et al. (2020)
      1995–2015 168 135/33 NA NA p16 IHC and/or HPV DNA 50.0% Molony et al. (2020)
      Brazil
      1999–2010 346 308/38 55 16 HPV16 and p16 IHC 6.1% Buexm et al. (2020)
      2017–2019 91 78/13 61 NA p16 IHC 20.9% Girardi et al. (2020)
      1984–2014 215 190/25 56 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59, 68, 73, 82 HPV DNA detection and/or p16 IHC 59.1% De Cicco et al. (2020)
      Germany
      2000–2017 102 82/20 57.5 16, 18, 33, 59 HPV genotyping and p16 IHC 40.2% Weiss et al. (2020)
      2000–2011 141 103/38 NA 16 HPV DNA and p16 IHC 34.0% Huebbers et al. (2019)
      2000–2014 323 245/78 58.87 NA HPV DNA and p16 IHC 19.8% Grønhøj et al. (2019a)
      2000–2017 730 NA NA NA HPV DNA and p16 IHC 27.1% Wittekindt et al. (2019)
      2007–2016 92 77/15 NA 16, 18 HPV Genotyping and p16 IHC 71.4% Freitag et al. (2020)
      Denmark
      2000–2017 2169 1564/605 58 16, 18, 33, 25, 59, 26, 31, 45, 56, 11, 58 HPV DNA detection and p16 IHC 55.0% Zamani et al. (2020)
      2000–2014 417 0/417 61.2 NA HPV DNA and p16 IHC 48.7% Christensen et al. (2019)
      2000–2014 993 720/273 59.50 NA HPV DNA and p16 IHC 56.9% Grønhøj et al. (2019a)
      2000–2014 1499 NA NA NA HPV DNA and p16 IHC 55.0% Grønhøj et al. (2019b)
      2000–2014 1243 903/340 60.2 NA HPV DNA and p16 IHC 63.4% Rasmussen et al. (2019)
      Korea
      NA 60 50/10 59 NA HPV DNA detection and p16 IHC 80.0% Suh et al. (2020)
      2004–2013 113 101/12 NA NA HPV Genotyping and p16 IHC 69.9% Kwon et al. (2020)
      Austria
      2014–2019 62 48/14 NA 16, 18, 33, 40, 62, 68 HPV Genotyping or p16 IHC 100% Kofler et al. (2020)
      South Glasgow
      2010–2017 272 NA NA 16. 18. 33. 39. 58 HPV Genotyping 44.0% Zubair et al. (2020)
      Finland
      2000–2016 157 110/47 59.5 NA p16 IHC 9.8% Sievert et al. (2020)
      Spain
      2017–2019 54 43/11 62 NA p16 IHC and HPV DNA 18.5% Viros Porcuna et al. (2020)
      Cameroon
      2014–2015 101 31/70 42 32, 68, 82 p16 IHC and HPV RNA ISH 5.0% Rettig et al. (2019)
      Sweden
      2000–2018 195 123/72 67 NA HPV DNA and p16 IHC 15.9% Hammarstedt et al. (2020)
      Croatia
      2002–2015 99 82/17 60 NA HPV DNA and HPV E6 mRNA 40.4% Božinović et al. (2019)
      Thailand
      2010–2016 110 95/15 59 16, 18, 26, 31, 33, 35, 39, 45, 51, 52, 53, 56, 58, 59, 66, 68, 70, 73, 82 p16 IHC and HPV DNA ISH 14.5% Nopmaneepaisarn et al. (2019)
      Italy
      2010–2017 59 43/16 66 NA HPV DNA 41.2% Ravanelli et al. (2020)
      Studies included in this table meet the following criteria: OPSCC patients; HPV DNA or RNA positive or p16 positive; studies published between January 2019 and October 2020
      aThis study included women with OPSCC only
      IHC: immunohistochemical staining; ISH: in situ hybridization

      Table 1.  Human papillomavirus (HPV) status in relation to oropharyngeal squamous cell carcinoma (OPSCC).

      HPV-driven oncogenic processes are characterized by HPV oncoproteins E6 and E7, which induce p53 and retinoblastoma (Rb) degradation, consequently leading to a deregulation of the cell cycle and an inhibition of apoptosis (Wittekindt et al. 2018). A plethora of data have been recently accumulated for different incidences of gene mutations and chromosomal aberrations between HPV-related and HPV-unrelated HNSCCs (Pickering et al. 2013; The Cancer Genome Atlas Network 2015). For example, TP53 mutations are found in approximately 60%–70% of HNSCCs, and different gain-of-function p53 mutants are related to oncogenesis, especially in HPV-unrelated HNSCC (Zhou et al. 2016). We recently developed a prognostic scoring system including five covariates (age, pT, pN, perineural invasion, and EAp53 score) for HPV-independent HNSCC patients via The Cancer Genome Atlas (TCGA)-based tumor genomic analysis (Qian et al. 2019). In addition to TP53 mutation, the exclusivity of CDKN2A and TERT driver mutations has also been identified in HPV-related HNSCC (Zapatka et al. 2020). Recurrent deletions and truncating mutations of TNF receptor-associated factor 3 (TRAF3), which is involved in innate and acquired antiviral immune responses, were found to be associated with HPV-related HNSCC (The Cancer Genome Atlas Network 2015). Furthermore, recent omics studies on HPV virus-host protein interactions have identified several potential and multiple oncogenesis pathways that can be promoted by HPV interactions, similar to recurrent mutations in cancer (Eckhardt et al. 2018). Moreover, an increase in mutations related to a higher expression of apolipoprotein B mRNA-editing catalytic polypeptide (APOBEC) was found in HPV-related HNSCC compared to HPV-negative tumors (Zapatka et al. 2020). Notably, it is hypothesized that a pathogenetic process is needed for the development of HPV-related cancers (Cui et al. 2019). A recent study demonstrated that genotypes of KRAS mutations and a loss of PTEN with both HPV E6/E7 dependence and independence lead to precipitous cervical cancer development, while HPV E6/E7 alone leads to only carcinoma in situ in a mouse model (Böttinger et al. 2020). A comprehensive understanding of how these mutations are involved in the carcinogenesis of HPV-related cancers remains to be established.

      Another important consideration is antiviral defenses and host–pathogen interactions that help us understand HPV-induced immune evasions in HPV-related cancers (Zhou et al. 2019). E6 and/or E7 seropositivity has been found in the majority of HPV DNA-positive HNSCC patients and is associated with longer recurrence-free survival (Smith et al. 2010; Lang Kuhs et al. 2017). A genome-wide association study on the association between OPSCC and human leukocyte antigen (HLA) loci demonstrated that the class Ⅱ haplotype DRB1*1301-DQA1*0103-DQB1*0603 was associated with a strongly reduced risk of HPV-related OPSCC compared to HPV-negative tumors (Lesseur et al. 2016).

      However, the down-regulation of antigen-processing machinery components against HPV oncoproteins (i.e., HPV16 E7 or E5) has been reported in cervical cancer and HNSCC (Albers et al. 2005). HPV16 E7 can also suppress stimulator interferon gene (STING) complex-induced type Ⅰ interferon (IFN-I) activation, by which effector T cell expansion is limited (Luo et al. 2020). Moreover, higher membranous PD-L1 expression at the tonsils and high levels of PD-1 expression within the majority of CD8 + tumor-infiltrating leukocytes (TILs) indicate adaptive immune resistance in HPV-related HNSCC (Lyford-Pike et al. 2013). In addition, the immune features within the tumor microenvironment (TME) of HPV-related HNSCC can be differentiated from HPV-negative tumors (Cillo et al. 2020). HPV-related OPSCC patients with HPV16-specific type-Ⅰ T cells and type-Ⅰ-oriented TME have a better prognosis than patients lacking HPV immunity (Welters et al. 2018). Enriched germinal center B cells in TILs of HPV-related HNSCC indicate their role through germinal center reactions during virus-driven progression (Cillo et al. 2020). B cells in nongerminal center states are more prominent in HPV-negative tumors (Cillo et al. 2020). Recently, HPV-specific B cell responses, including antigen-specific activated and germinal center B cells and plasma cells, were identified in the TME of samples from HPV-related HNSCC (Wieland et al. 2020). These findings demonstrate more heterogeneous immunity in HPV-driven tumors, which has the potential to refine the risk group.

      A fundamental understanding of the heterogeneity, plasticity and cellular mechanisms of HPV-related HNSCC biology offers an opportunity to uncover therapeutic windows and to separate the small subset of patients with HPV-related cancer at high risk, from whom a de-escalation approach would be appropriate. Here, we review the current knowledge regarding HPV-related HNSCC and the challenges of targeting these cancers. We also discuss potential applications for biomarker-based stratification strategies, which are due to spatial and temporal varieties of conditions, including the broader TME and the underlying pathways of antitumor response and tumor resistance.

    • Worldwide, more than 830, 000 cases of HNSCC are diagnosed each year, with approximately 430, 000 deaths (Bray et al. 2018). The incidence of HPV-related HNSCC is increasing, i.e., by 2.5% per year for OPSCC in the United States (Mifsud et al. 2017; Mourad et al. 2017). In China, the estimated age-standardized incidence rate of HNSCC was 2.7 per 100, 000 and 2.22/100, 000 person-years by the standard population of China in 2000 (ASRIC and ASRMC) for OPSCC (Liu et al. 2018). As seen in recent studies in China, the rates of HR-HPV infection in HNSCC are 7.5% in a case control study and 26.4% in southern Chinese population (Chor et al. 2016; Ni et al. 2019). HPV-related HNSCCs have been found in the oral cavity, oropharynx, pharynx, larynx and salivary glands, with the highest prevalence for OPSCCs (Qian et al. 2016; Wiegand et al. 2018). Table 1 illustrates recent reports of HPV-related OPSCCs in different regions. Squamous cell carcinoma of unknown primary in the head and neck (SCCUPHN) present with neck lymph node metastasis with no evidence of a primary tumor accounts for 4%–5% of HNSCCs (Ren et al. 2019; Schroeder et al. 2020). The prevalence of HPV positivity in SCCUPHN was 49% as reviewed by a recent meta-analysis (Ren et al. 2019). In a prospective study, SCCUPHN, defined as the metastasis of SCC to a neck node, was likely to be HPV-driven OPSCC because of the similarities in risk factor profile and survival with HPV-driven OPSCC (Schroeder et al. 2020). Additionally, the rate of a secondary primary tumor in HNSCC with unknown HPV status is reported to be ~ 14%, and the highest is 36% (Chuang et al. 2008). Patients who develop multiple primary tumors of HPV-related HNSCC (one primary HPV-related OPSCC with a second primary tumor of any head and neck site) were recently reported at a rate of 0.95%–2.64% depending on different datasets (Strober et al. 2020). Notably, those patients tend to be younger and have no neck adenopathy compared to patients with one primary HPV-related tumor (Strober et al. 2020).

      The prevalence of HPV-related OPSCC varies in different age groups. In earlier reports, patients with HPV-related OPSCC tended to be demographically younger. However, an increase in the proportion of patients of older age over time leads to the median age being older (Smith et al. 2004; Fakhry et al. 2020). Among recent studies from different regions, the median age varies from 37 to 73 years with the different time intervals of collected data (Table 1).

      To date, HR-HPV genotypes 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59 are categorized as Group 1 carcinogens, 68 as Group 2A and 26, 30, 34, 53, 66, 67, 69, 70, 73, 82, 85, 97 as Group 2B carcinogen (Bouvard et al. 2009). HPV infections with HR-subtypes (HPV 16, 18, 26, 30, 31, 33, 35, 39, 45, 51, 52, 53, 56, 58, 59, 66, 67, 68 and 69) have been identified in HNSCC (Fig. 1, Table 1) (Castellsagué et al. 2016; Tumban 2019). A recent analysis of 3, 680 samples from 29 countries shows that HPV 16, 33, 35 and 18 are responsible for the majority of HPV-related OPSCCs (Castellsagué et al. 2016). Overall, HPV 16 is predominant and accounts for 90%–97% of HPV-related OPSCCs (Gillison et al. 2015). Human immunodeficiency virus (HIV)-infected individuals have a risk of developing HPV-HIV coinfection, and the incidence of age-standardized HPV-related HNSCC increased from 6.8 to 11.4 per 100, 000 person-years from 1996 to 2009 in North America (Beachler et al. 2014; Dsouza et al. 2014).

      Figure 1.  A The HPV genomic structure (use HPV 16 as an example). The ORFs encoding the early (blue) and late (yellow) genes are marked. B The life cycle of HPV starts with viral infection at the basal cells through trauma and the viral genomes are maintained at low copy number level in undifferentiated cell. Upon cell differentiation, the viral genomes become amplified and virion assembly ensues, resulting in the release of viruses in the cornified layer. C The contribution of different high-risk HPV genotypes to head and neck squamous carcinoma (Tumban 2019).

    • The genetic information of HPVs is carried by their circular, double-stranded DNA genome, which has a size of approximately 8, 000 base pairs (Zheng and Baker 2006). According to the genome data compiled in the Papillomavirus Episteme database (https://pave.niaid.nih.gov/), there are more than 225 distinct genotypes of HPVs, which are classified according to the DNA sequence of the L1 gene. To be recognized as a new genotype, the L1 region sequence has to be more than 10% different from its closest member. Despite the sequence variation, the HPV genome structure is similar for all genotypes and is organized into three regions (Fig. 1). The early gene region contains several overlapping open reading frames (ORFs) denoted E1, E2, E4, E5, E6, E7 and E8 (E5 and E8 not present in all genotypes) that code for nonstructural and regulatory proteins involved in various processes of the virus life cycle, such as viral replication and transactivation of gene expression. Currently, 12-15 genotypes of HPVs are considered oncogenic for certain types of cancers, with the early proteins E6 and E7 playing key roles in oncogenesis and thus are two multifunctional viral oncoproteins (Zheng and Wang 2011; Roman and Munger 2013; Vande Pol and Klingelhutz 2013; Wang et al. 2014). The late gene region encodes structural proteins L1 and L2, the major and minor capsid proteins. In addition, a noncoding region (NCR) or upstream regulation region (URR) between the late and early regions contains a viral replication Ori (Wang et al. 2017) and binding sites for various viral and host transcription and regulation factors, such as viral E1 and E2, the functions of which are essential in viral DNA replication and amplification.

      The transcription of viral genes and viral genome replication are tightly regulated throughout the life cycle of papillomaviruses in a host cell differentiation manner. The virus infects basal layer cells through epithelial layer trauma. In undifferentiated basal cells, the expression of most viral early proteins from viral early promoter-derived RNA transcripts is maintained at low levels to avoid triggering immune responses. As cell differentiation occurs, viral DNA replication and activation of the viral late promoter lead to the expression of viral late genes and the production of infectious virus particles in the granular and cornified layers of the epithelium (Fig. 1B). Although epigenetic modifications, including chromatin remodeling and DNA methylation on both viral and host genomes, play important roles in the control of viral gene expression, with numerous studies being explored for their function at various stages of the viral life cycle, the posttranscriptional regulation, such as RNA splicing, polyadenylation, stability, export and translation, is essential for the expression of each viral protein. Our understanding of these regulations remains minimal. In high-risk HPV16 and HPV18, viral E6 and E7 are expressed as a single bicistronic E6/E7 RNA undergoing extensive RNA splicing (Tang et al. 2006; Wang et al. 2011; Ajiro et al. 2016). While viral E6 is expressed from the unspliced E6 RNA coding region, viral E7 protein can be expressed only from spliced E6*I RNA (Zheng et al. 2004; Tang et al. 2006; Ajiro et al. 2012; Brant et al. 2019). To date, we know very little how viral E1 and E2 are expressed in the context of the entire viral genome during high-risk HPV infection. Papillomaviral E4 is encoded from the viral early region, but it is a viral late protein translated from a viral late transcript derived from a late promoter residing in the E7 coding region (Wang et al. 2011; Xue et al. 2017). By alternative splicing, this late transcript also translates viral L2 and L1 for viral capsid formation (Zheng and Baker 2006). Viral L1 and L2 form the basis for HPV vaccines in preventing HPV infection and the development of HPV-induced cancers (Zhou et al. 1991; Kirnbauer et al. 1992; Schiller et al. 2012). Further elucidation of the molecular mechanisms that control viral DNA replication, transcription and posttranscriptional regulation may provide novel targets for combating HPV and treating HPV-associated cancers. For more detailed discussion on epigenetic regulation of HPV genome transcription, please refer to a recent review by Burley et al. (Burley et al. 2020).

    • The integration of HR-HPV DNA into the host genome has been considered an important biological step in the development of carcinogenesis in invasive cervical cancer and HNSCC (Mesri et al. 2014; Zapatka et al. 2020). Initial studies demonstrated that transcriptionally active integrated and/or episomal viral DNA in HNSCC cell lines was independent of viral copy number and integration sites (Akagi et al. 2014; Olthof et al. 2015). HPV integration can lead to host genomic instability, such as deletions, inversions, and chromosomal translocations (Akagi et al. 2014). A number of viral integration sites in the host genome were found in intergenic regions as well as cancer-associated genes such as TP63, ETS2, RUNX1, FOXA1 and ERBB2 (Olthof et al. 2014, 2015; Walline et al. 2016; Koneva et al. 2018). Moreover, viral integration into cellular genes was commonly identified in recurrent HPV16-positive OPSCC patients, and these cellular genes are related to cancer-associated signaling pathways or mechanisms (Walline et al. 2016). Integrated viral DNA copies could be in tandem. Viral DNA integration through the disruption of the viral E2 region leads to increased transcription of viral E6 and E7. Tumors with HPV DNA integration differ from HPV integration-negative tumors by different patterns of DNA methylation and gene expression profiles (Parfenov et al. 2014). Recently, Zapatka et al. found that HPV 16 and HPV 18 integration events in cervical cancer and HNSCC were associated with local variations and genomic rearrangements based on the Pan-Cancer Analysis of Whole Genomes Consortium (Zapatka et al. 2020).

      HPV integration inducing genome instability is hypothesized to be a secondary genetic event in the carcinogenesis of HPV-associated HNSCC. HPV infection is associated with increased expression of the APOBEC genes APOBEC3A and APOBEC3B but exclusively with known driver genes such as TP53, CDKN2A and TERT (Kondo et al. 2017; Zapatka et al. 2020). These findings suggest a possible role of APOBECs in HPV-induced carcinogenesis, i.e., the activity of APOBECs as C-to-U RNA editing enzymes contributes to alterations in host genome expression, and APOBEC3A increases tumorigenesis in vivo (Burns et al. 2013; Wallace and Münger 2018; Law et al. 2020). In addition, as part of the immune defense system, APOBEC3A can sensitize cancer cells to cisplatin treatment by activating base excision repair and mediating the repair of cisplatin interstrand crosslinks (Conner et al. 2020). These results suggest a role of impaired antiviral defense in driving the carcinogenesis of HPV-related HNSCC. HPV16 insertions also lead to the amplification of the PIM1 serine/threonine kinase gene in HNSCC cell lines (Broutian et al. 2020). The inhibition of PIM family kinases successfully decreased cell proliferation in vitro and in vivo in an HNSCC model (Broutian et al. 2020).

      Notably, viral integration can be found in both tumors that respond to treatment and recurrent tumors with more complex integration patterns in host genes (Walline et al. 2016). By analyzing viral-host fusion transcripts, Koneva et al. showed that the HPV-positive but HPV integration-negative subgroup had better survival than the HPV integration-positive subgroup and HPV-unrelated HNSCC (Koneva et al. 2018). Moreover, HPV-positive but HPV integration-negative tumors had enhanced tumor infiltrates of immune cells and upregulated immune-related genes. Consistently, another study indicated that HPV-related HNSCC can be subdivided into an immune cell enrichment phenotype and a phenotype with higher proliferation (Koneva et al. 2018). Thus, the enhanced immune profile in patients with HPV-positive but HPV integration-negative tumors may be attributed to better survival for these patients. However, potential mechanisms for HPV integration-induced oncogenesis of HNSCC remain elusive.

    • Recent landmark sequencing studies have demonstrated gene expression profiles and somatic mutations such as TP53, CDKN2A, PTEN, PIK3CA, EGFR, HRAS, FBXW7 and NOTCH1 in diverse anatomical sites of HNSCC (Agrawal et al. 2011; Stransky et al. 2011; Pickering et al. 2013; The Cancer Genome Atlas Network 2015). Importantly, diversity in the number of mutations and gene profiles was seen in patients with a history of tobacco use and between HPV-related and HPV-unrelated tumors. The mutation rate of HPV-related tumors was almost half that of HPV-unrelated tumors (Stransky et al. 2011). Thus, two etiologies may result in the alteration of oncogenes and tumor suppressor genes that have tumorigenic effects involved in multistep biological processes.

      HPV-related HNSCC harbors mutations in the oncogene PIK3CA encoding PI3K catalytic p110 subunit alpha, a loss of TRAF3 and the amplification of E2F1 (The Cancer Genome Atlas Network 2015). A recent comprehensive analysis on oral squamous cell carcinoma (OSCC) identified secondary genetic alterations, including PIK3CA, ZNF750 and EP300 as candidate cancer driver genes (Gillison et al. 2014; Gillison et al. 2019a). As we discussed above, virus-host interactions, as seen by the interaction between HPV integration with APOBEC and others, may shape genomic alterations and facilitate tumorigenesis.

      Notably, PIK3CA mutations (2.6% to 19%) lead to the activation of the PI3K-AKT-mTOR1 signaling pathway necessary for the viral life cycle (Lui et al. 2013; Surviladze et al. 2013). HPV oncoproteins E6 and E7 also increase PI3K-AKT-mTOR signaling (Pim et al. 2005; Contreras-Paredes et al. 2009). Moreover, both HPV-related and HPV-unrelated HNSCCs harbor PIK3CA mutations, and higher expression of PIK3CA in primary tumors is associated with tumor recurrence and chemo- and radioresistance (García-Escudero et al. 2018; Marquard and Jücker 2020). Thus, inhibitors targeting the PI3K-AKT-mTOR pathway have been developed for cancer therapies (Marquard and Jücker 2020). However, the clinical response rates remain modest in these studies and warrant further investigation (Marquard and Jücker 2020). Studies in patient-derived xenograft (PDX) models demonstrate that EGFR, AKT1 and CSMD1 copy number aberrations are related to the effect of PI3-kinase inhibition regardless of the status of PIK3CA mutation (Ruicci et al. 2020). Adaptive resistance to PI3K inhibition is also seen in mTORC2-mediated Akt reactivation following PI3K inhibition. The knockdown of RICTOR, a subunit of mTORC2, can sensitize HNSCC cells to PI3K inhibition in vitro (Ruicci et al. 2019b).

      TRAF3 functions as a tumor suppressor negatively to regulate NF-κB pathway activation in HPV-related HNSCC (Zhang et al. 2018). TRAF3 is involved in the innate and acquired antiviral immune responses (The Cancer Genome Atlas Network 2015). The re-expression of TRAF3 can enhance TP53 and RB tumor suppressor proteins and decrease HPV E6 oncoprotein in HPV + HNSCC cell lines (Zhang et al. 2018). Thus, regulating TRAF3 and aberrantly activating the alternative NF-kB pathway warrant further investigation as therapeutic targets in cancer treatment.

      Another interesting aspect of HPV-induced HNSCC oncogenesis is epigenetic alterations, including DNA methylation and histone modifications (Kostareli et al. 2013; Hatano et al. 2017; Papillon-Cavanagh et al. 2017; Guo et al. 2020; Mac and Moody 2020). For example, higher DNA methylation levels are more common in HPV-related OPSCC than in HPV-unrelated tumors and normal tissues (Ren et al. 2018). Furthermore, candidates for DNA differentially methylated regions (DMRs) can discriminate HPV-related OPSCC from normal controls with good receiver operating characteristic (ROC) performances (Ren et al. 2018). Moreover, these changes are involved in different stages of the HPV life cycle in HPV-related OPSCC and other HPV-related malignancies (Boscolo-Rizzo et al. 2017; Mac and Moody 2020). HPV oncoproteins E6 and E7 may confer histone methylation and acetylation on targeted genes (Boscolo-Rizzo et al. 2017; Gaździcka et al. 2020). Histone methylation, such as elevated trimethylation at lysine 27 of histone H3 (H3K27me3), in HPV-related HNSCC is associated with tumorigenesis (Lindsay et al. 2017). Targeting zeste homolog 2 (EZH2), a histone methyltransferase, can reduce H3K27me3 and has the potential to sensitize cells to chemotherapy (Lindsay et al. 2017). Evidence also suggests that histone acetylation and deacetylation may deregulate the transcription of various genes in malignancy development in HNSCC (reviewed in (Boscolo-Rizzo et al. 2017; Gaździcka et al. 2020). Recently, Liu et al. demonstrated that HR-HPV oncogenes induce the long noncoding RNA (IncRNA), Inc-FANCI-2, mediated by E7 and E6, which is independent of p53/E6AP and pRb/E2F in cervical carcinogenesis (Liu et al. 2021). The differential regulation of lncRNAs between HPV-related and HPV-unrelated HNSCCs has also been demonstrated (Nohata et al. 2016; Haque et al. 2018; Song et al. 2019; Kopczyńska et al. 2020). However, their role in the oncogenesis of HNSCC remains elusive. In conclusion, epigenetic alterations might be useful in identifying subgroups of tumors, predicting clinical outcomes and providing potential targets (Kostareli et al. 2013, 2016; Ren et al. 2018; Shen et al. 2020).

      In particular, research on single-cell molecular profiling can provide spatial and temporal characteristics of HNSCC in regard to intratumor heterogeneity (Puram et al. 2017; Qi et al. 2019). Tumor genetic heterogeneity determined by mutant-allele tumor heterogeneity is associated with worse outcome of patients with HNSCC (Mroz et al. 2013). Furthermore, a single-cell transcriptomic analysis identified subtypes as atypical, mesenchymal, basal, and classic phenotypes of OSCC (Puram et al. 2017). Recently, Cillo et al. identified a differential spectrum of immune lineages (helper CD4+ T cells and B cells) between HPV and HPV+ HNSCCs by single-cell transcriptional profiling (Cillo et al. 2020). Thus, with the advantage of single-cell technologies, a more profound understanding of genetic, epigenetic and transcriptional differences for both HPV-related and HPV-unrelated tumors would be possible (Qi et al. 2019).

    • A wide exploration of the tumor-escape mechanisms of HNSCC leads to immunological approaches against tumors, including cancer vaccination and ICB treatment (Fig. 2A) (Albers et al. 2010; Xu et al. 2018; Cramer et al. 2019). The anti-PD-L1 monoclonal antibody atezolizumab has also shown its effect in patients with previously treated, advanced HNSCC in a phase I trial (Colevas et al. 2018). However, the overall response rates for both anti-PD1 and anti-PD-L1 treatments are only approximately 20% (Colevas et al. 2018; Qian et al. 2020). Virally mediated tumors such as HPV-related HNSCC demonstrate HPV-specific T cell immunity, and there is significant interest in developing a combination therapy to improve ICB treatment efficacy (Bhatt et al. 2020).

      Figure 2.  A Immune escape of HPV-related head and neck squamous cell carcinoma (HNSCC) and potential approaches to improve immunotherapeutic effects against HPV-related tumor. HLA: human leukocyte antigen; TCR: T-cell receptor; PD-1: programmed cell death protein 1; PDL-1: programmed death-ligand 1; LMP2: low molecular weight protein 2; TAP1: antigen processing subunit 1; STING: stimulator of interferon genes. B The platform of risk stratification for HPV-related HNSCC. IHC: immunohistochemical staining.

      High PD-1 expression was significantly associated with HPV-positive HNSCC in an analysis of a TCGA dataset (Lyu et al. 2019). PDL1 expression in HPV-related HNSCC ranges from ~49.2% to ~75% according to a report (Outh-Gauer et al. 2020). Increased PD1 expression on T cells is correlated with HR-HPV infection in the pathogenesis of cervical intraepithelial neoplasias (CINs) (Yang et al. 2013, 2017). Additionally, a higher HPV 16 viral load is correlated with high CD8 + and PD-1 + TIL expression in anal squamous cell cancer (ASCC) (Balermpas et al. 2017). PDL1 expression is significantly enhanced in relation to HPV positivity (i.e., HPV16 E7 oncoprotein) in CINs and cervical cancer compared to normal cervical epithelia (Yang et al. 2013; Mezache et al. 2015). Taken together, these findings suggest that PD1/PDL1 pathways may be activated by HR-HPV (i.e., E5, E6 and E7 oncoproteins) for HPV-related cancers. HPV E6/E7-induced master regulators (ENO1, PRDM1, OVOL1, and MNT) are positively correlated with PD-L1 and TGFB1 expressions in cervical cancer (Qin et al. 2017). The activation of the PD1/PDL1 pathway by HR-HPV can further inhibit Th1 cytokine IFN-c and IL-12 expressions and upregulate TH2-type cytokine and IL-10 expressions, consequently leading to immunosuppression and CIN progression (Wakabayashi et al. 2019; Zhou et al. 2019). Remarkably, the reactivity of nonviral tumor antigen-specific T cells, including mutated neoantigen or cancer germline antigen, together with HPV oncoprotein-reactive T cells was observed in HPV-associated metastatic cervical cancer patients with complete regression after tumor-infiltrating adoptive T cell therapy (Stevanović et al. 2017). Thus, it would be interesting to understand the crosstalk of HPV-mediated immunosuppression and immune checkpoint activities for further development of therapeutic strategies.

      Efforts to define HPV-related HNSCC responses to ICB treatments were initially assessed based on recent clinical trials (Bauml et al. 2017; Ferris et al. 2018; Burtness et al. 2019). In CheckMate 141, a better median overall survival among patients receiving nivolumab treatment was found to be associated with HPV-positive tumors with PDL1-positive expression (Ferris et al. 2018). In KEYNOTE-012, the response rate to pembrolizumab treatment varied among HNSCC patients with PD-L1 positivity, with a higher response rate in HPV-positive tumors than in HPV-negative tumors (Seiwert et al. 2016). A recent meta-analysis demonstrated an improved response rate to ICB treatment among patients with HPV-positive HNSCC and a higher OS in patients with PDL1-positive HNSCC (Galvis et al. 2020). However, the treatment efficacy of atezolizumab in a phase I trial was found independent of HPV status and PD-L1 expression level (Colevas et al. 2018). Because of the limited size of each study, the findings should be interpreted with caution.

      The development of combination therapy by employing different immune priming approaches has the potential to improve effective immune responses in HPV-related cancer patients who receive immunotherapy. For example, targeting HPV16/18 E6/E7 by a DNA vaccine with an IL12 adjuvant is able to generate durable HPV16/18 antigen-specific cytotoxic T cells and tumor immune responses in patients with p16 + locally advanced HNSCC (Aggarwal et al. 2019). One patient who developed metastatic disease received anti-PD1 nivolumab treatment and had a complete response (Aggarwal et al. 2019). This phase Ⅰ/Ⅱa clinical trial, in line with other vaccination strategies targeted to HR-HPV E6/E7 oncoproteins, demonstrates a complementary approach to improve immunotherapy outcomes (Aggarwal et al. 2019; Massarelli et al. 2019; Xu et al. 2020). NLRX1 depletion leads to a turnover of HPV16 E7, potentiating STING/IFN-I suppression and consequently improving tumor control (Luo et al. 2020). Clinical trials (NCT02675439, NCT03172936, and NCT03010176) utilizing a combination of a STING agonist plus anti-CTLA-4 or anti-PD1 ICB treatments are underway (Luo et al. 2020). In addition, it has been demonstrated that HPV E5 mediates resistance to anti-PD-L1 immunotherapy, which is due to acquired loss of major histocompatibility complex (MHC) expression (Sharma et al. 2017; Miyauchi et al. 2020). Rimantadine, an FDA-approved antiviral drug to treat influenza that was recently found to induce an antitumor response, could increase MHC expression in HPV E5-expressing HNSCC (Miyauchi et al. 2020) (Fig. 2A). However, rimantadine in combination with radiation and PD-L1 checkpoint blockade treatment did not show a synergistic effect (Miyauchi et al. 2020). E2-derived CD8 T cell epitopes were found in patients with HPV-related HNSCC (Krishna et al. 2018). Wieland et al. demonstrated that the E2 protein is a major target of the humoral immune response in the TME of HPV-related HNSCC (Wieland et al. 2020). Thus, a combination of HPV E2, E6 and E7 as targets needs to be explored for future immunotherapeutic approaches. Alternatively, innovations in nanotechnology will likely synergize with immunotherapy to elicit a robust treatment response in HNSCC (Xu et al. 2017). Another example is an HR-HPV nanovaccine formulated with CL 1, 2-dioleoyl-3-trimethyl-ammonium-propane (DOTAP), and long HR-HPV peptides can successfully boost Ag-specific CD8 T cell responses, induce complete tumor regression through a type Ⅰ IFN response in HPV-E6/E7 TC-1 tumor models and synergize with an anti-PD1 checkpoint inhibitor (Gandhapudi et al. 2019). It should be noted that chemoradiotherapy decreases HPV-specific T cell responses and increases PD-1 expression on CD4 + T cells in patients with HPV-related oropharyngeal cancer (Parikh et al. 2014). Accordingly, a future analysis of patients with HPV-related HNSCC who have and have not responded to ICB treatment would provide additional insights for targeted immunotherapy as well as deintensified treatments.

    • To formulate a diagnosis for HPV-related HNSCC, methods based on p16 immunohistochemical (IHC) staining, quantitative polymerase chain reaction (qPCR)-based HR-HPV DNA or RNA testing, and HPV DNA or RNA in situ hybridization (ISH) have been established. In 2018, the College of American Pathologists (CAP) and American Society of Clinical Oncology (ASCO) released guidelines that HR-HPV testing should be performed for OPSCC and cervical nodal metastases of unknown primary tumors (Fakhry et al. 2018; Lewis et al. 2018). Protein p16 IHC staining was recommended prior to other HPV testing with a cutoff of 70% nuclear and cytoplasmic positivity (Fig. 2B). For HPV-related tumors, the HR-HPV E7 oncoprotein increases expression of histone lysine demethylase 6B (KDM6B) and induces pRb degradation, thereby leading to H3K27-specific demethylation (derepression) of p16 promoter to enhance high level of p16 expression for proliferation of the cancer cells lacking the G1 checkpoint due to viral E7-induced loss of pRb (McLaughlin-Drubin et al. 2013; Pal and Kundu 2019). However, Albers et al. demonstrated that a subgroup of HNSCC with HPV-DNA+/p16 had poor survival compared to subgroups with HPV-DNA+/p16+ and HPV-DNA/p16+, which suggests a biologically different subtype independent of HPV status (Albers et al. 2017). A recent survival analysis further revealed that 60.6% of p16+ HPV-DNA OPSCCs did not resemble HPV16-driven but HPV-negative tumors (Wagner et al. 2020). Moreover, gene signatures such as TP53 mutations are indistinguishable between tumors with HPV DNA+RNA and those with HPV DNA (Wichmann et al. 2015). As indicated above, some patients who received deintensified treatments failed to show an improvement in clinical trials. Thus, pathology-based HPV testing beyond p16 IHC staining remains to be established. HPV RNA ISH, by which viral E6/E7 mRNA has been detected, showed a better survival prediction and a higher specificity than p16 IHC staining (Lewis 2020). Additionally, the sensitivity of HPV RNA ISH is higher than that of HPV DNA ISH (91% vs. 65%) (Kerr et al. 2015). The performance of HPV RNA ISH testing in stratifying OPSCC patients is worth further validation (Lewis 2020).

      While current HPV-specific testing warrants further improvement, a better understanding of the biology of HPV-related HNSCC is necessary for us to identify new biomarkers correlated with disease outcomes and to predict treatment response signatures, especially for immunotherapy. For example, the latency effect of tobacco smoking exposure appears more profound in HPV-related HNSCC individuals who start smoking before sexual activity when compared to those who start after (Madathil et al. 2020). High levels of TILs that reflect the immune response can stratify HPV-related OPSCC patients into high-risk and low-risk groups for survival (Ward et al. 2014). Interestingly, a prognostic model was further developed, including three covariates (TIL levels, heavy smoking, and T-stage) in which low TIL levels, heavy smoking, and late T-stage were related to poor outcome for HPV-related OPSCC (Ward et al. 2014). HPV integration status based on RNA-seq data can differentiate survival between HPV integration-positive and HPV-negative HNSCC patients (Koneva et al. 2018). Most importantly, a set of immune-related gene signatures enriched in HPV-positive but HPV integration-negative tumors are distinguishable from those in HPV integration-positive tumors (Koneva et al. 2018). The lower E2F target gene expression predicted by reduced E7 levels is associated with disease recurrence through patient-derived xenograft (PDX) models for HPV-related HNSCC (Facompre et al. 2020). HPV + tumors have been related to an inflamed/mesenchymal phenotype. By analyzing RNA-sequencing data from TCGA, studies show that HPV+ HNSCC exhibited an upregulation of MHC-Ⅰ- and MHC-Ⅱ-related genes, which may be induced by IFN-gamma, a strong Th1 response and higher expression of the T cell "exhaustion markers" LAG3, PD1, TIGIT, and TIM3 with coordinately expressed CD39 compared to HPV tumors at the transcription level (Gameiro et al. 2018, 2019; Ruicci et al. 2015). In addition, cancer stem cells (CSC), a subpopulation within the bulk tumor entity of HNSCC, have shown their characteristics such as less antigen expression, processing, and presentation to induce the immunogenicity and immunosuppression (Qian et al. 2020). Further, studies demonstrate that HNSCC patients with the HPV+/CSClow phenotype had better outcomes than HPV/CSChigh group (Reid et al. 2019). Given the paucity of molecular biomarkers, combined insights into genetic, epigenetic and transcriptional alterations can provide robust candidate biomarkers to stratify patients further and to predict treatment outcomes.

    • The molecular landscapes of HNSCC have largely been defined during the past decade. The intratumor heterogeneity (e.g., genetic, epigenetic and histopathologic) of HPV-related HNSCC has been identified as contributing to the pathophysiology of the disease. This variability contributes to the range of treatment responses observed for both established clinical practice and clinical trials. The clinical translation of these findings may help for dynamic risk restratification of HPV-related HNSCC patients with new molecular biomarkers. The management of HPV-related HNSCC is changing, and substantial research focusing on discovery approaches to cancer diagnostics and prognostic evaluations is required (Bigelow et al. 2020).

    • This work was supported by grants from the Medical and the Health Science Project of Zhejiang Province (2019KY327) and Guangji Talents Foundation Award (E) of Zhejiang Cancer Hospital.

    • The authors declare no conflict of interest.

    • This article does not contain any studies with human or animal subjects performed by any of the authors.

    Figure (2)  Table (1) Reference (199) Relative (20)

    目录

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return