Therapeutic Vaccines for Human Papillomavirus-Associated Malignancies: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Prophylactic vaccines against human papillomavirus (HPV) have proven efficacy in those who have not been infected by the virus. As for therapeutic vaccines, they require the differentiated modulation of the immune system as both chronic infections and cancers are associated with specific immunosuppression and impairment of the immune surveillance system.

  • carries
  • cancer
  • therapeutic
  • DNA
  • RNA

1. Introduction

According to the World Health Organization (WHO), cancer-related diseases represent the world’s second leading cause of death, resulting in approximately 19.3 million new cases and approximately 10 million deaths in 2020 [1]. In addition to genetic factors and habits such as smoking, alcoholism, and poor diet, infectious diseases—especially those arising from viral infections—stand out as responsible for approximately 13% of human cancers [2].
Among viruses, the human papillomavirus (HPV), belonging to the Papillomaviridae family, is linked to squamous cell carcinomas and adenocarcinomas, and is considered the second most prevalent etiologic agent, with 5% of the global burden (approximately 690,000 new cases) [2][3]. Currently, 229 types of HPV are described by the International HPV Reference Center (www.hpvcenter.se; accessed on 1 November 2022) [4][5]. All HPVs can induce benign proliferative lesions (such as warts), but 12 genotypes (HPVs types 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59) are considered high-risk because they are capable of inducing malignant transformations [6]. Of these, HPV16 and HPV18 comprise the most oncogenic and prevalent types [7]. Although the distribution of these genotypes varies by geographic area [8], they represent a considerable burden worldwide, especially in developing countries.
These HPVs infect both the cutaneous and mucosal epithelium and are tissue-specific, in which different subtypes preferentially infect each tumor subsite, and there may be specific immune barriers in each microenvironment [9][10]. With a high risk of infection, approximately 80% of people will be infected with HPV at some point in their lives [11]. However, while the immune system usually clears the virus, for some people, the infection remains and can lead to precancerous changes [12]. HPV infection is a major cause of cervical, anogenital, and oropharyngeal neoplasms [13], and more recently, although less prevalently, has been associated with cases of skin cancer [14], lung [15], and esophageal adenocarcinoma (EAC) [16].
Current viral infection control strategies are based on routine screening and prophylactic vaccination, implemented as a worldwide program against low-risk genotypes and the most prevalent oncogenic types [17]. Currently, three different HPV vaccines (Cervarix®, Gardasil®, and Gardasil®-9) have been or are being used worldwide to prevent HPV-related cancers [18]. More recently, the Cecolin vaccine was licensed in China, developed against types 16 and 18 and is currently under review by the WHO [19]. HPV vaccines prevent genital warts, cervical cancer, and most HPV-related cancers, including anogenital cancers [9]. In addition, it reduces the risk of most associated throat and penile cancers [20].
However, progress towards prevention is sometimes discouraging due to the limited access to vaccination and restrictions on screening for HPV-positive malignancies, particularly in developing countries [21][22]. Furthermore, despite the high efficacy linked to a significant reduction in the rate of cervical cancer, acceptance is limited, and they do not benefit patients with established tumors [23]. Traditional treatment options for these individuals, whether in advanced or recurrent stages, include chemotherapy, surgery, and radiation therapy, which are associated with a short survival and substantial adverse effects [24]. As a result, developing therapeutic options for HPV-related malignancies is imperative. Nucleic acid vaccines are safe, quick, and easy to implement platforms capable of evoking efficient adaptive responses [25][26]. However, strategies to increase the immunogenicity of DNA vaccines, minimizing the degradation of mRNA molecules, and enable their acquisition by immune cells are commonly necessary [27]. One way to overcome this limitation is to use delivery systems that allow for the uptake of specific antigens and help modulate the immune response [28].

2. Therapeutic Vaccines for HPV-Associated Malignancies

Prophylactic vaccines aim to prevent infection by a pathogen, and thus protect the individual from future contact through immunological memory [29]. The application of prophylactic vaccines induces an adaptive immune response primarily focused on the humoral immune response [30]. This immunity is achieved through B cells, but to be successful, it requires the participation of CD4+ T cells, hence depending on effective cellular immunity [31].
Capsid proteins from different high-risk HPV strains are used in preventive HPV vaccinations to generate a neutralizing antibody response that prevents recurrent HPV infection [10]. These vaccines are based on viral capsid protein L1 virus-like particles (VLPs) produced and expressed by yeast (such as Saccharomyces cerevisiae) and baculovirus in insect cells [32]. From the spontaneous self-assembly of the L1 protein, a highly immunogenic structure similar to the native conformation of virions is formed, which is then recognized by the immune system cells, inducing the production of neutralizing antibodies [33]. For these vaccines, the significance of the humoral response is evident; nevertheless, studies suggest that B cells may also have an indirect effect in modulating immune responses against HPV-related malignancies [10]. Recent studies show that additional diagnostics and therapies targeting B cells can help predict patients with a better prognosis who would benefit from less invasive treatments [34][35]. In the study by Kim et al. (2020), for example, it was found that in HPV-associated squamous cell carcinomas, B cells improved the overall survival and were activated by radiation and PD-1 blockade. Additionally, Hladíková et al. (2019) found that tumor-infiltrating B cells affect the progression of oropharyngeal squamous cell carcinoma through cell-to-cell interactions with CD8+ T cells.
As for therapeutic vaccines, they require the differentiated modulation of the immune system as both chronic infections and cancers are associated with specific immunosuppression and impairment of the immune surveillance system [36]. This vaccine aims to eliminate the disease by increasing, modulating, or redirecting the immune response, thus forcing the immune system to recognize pathogens and abnormal cells [37][38][39]. Since the pathogenic genesis of HPV is linked to the persistent expression of oncogenic viral proteins, most of them are considered as therapeutic targets against HPV-related malignancies [23].
It is currently recognized that the oncogenic causative role of high-risk HPV types is fundamentally attributed to the action of the main viral oncoproteins, E6 and E7, which, respectively, inhibit the tumor suppressors p53 and pRB, which are involved in the development of malignancies [40]. These oncoproteins act synergistically, targeting various cellular pathways involved in regulating cell cycle control, promoting cellular immortalization, and facilitating invasion and malignant progression in the host [41]. In addition to these, the HPV E5 protein has been considered an attractive therapeutic target to prevent the progression of precancerous lesions into invasive cervical cancer since it is considered a putative oncogene that acts in the first stage of carcinogenesis, is responsible for regulating the MHC-I, and mediates immune evasion [23][42]. Oncoproteins are constitutively produced at high levels in tumor cells and are not found elsewhere in the human body, which distinguishes HPV-associated cancers [41][43]. This constitutive production makes them an excellent target for the therapeutic vaccines designed to provoke a specific antitumor response, targeting cells that express the antigens and limiting the danger of harming healthy tissue [44].
Unlike prophylactic ones, they are primarily focused on cell-mediated immunity and involve the interaction between antigen-presenting cells (APCs) and naive T cells that will become CD4+ effector cells (via MHC-II) or CD8+ (via MHC-I) [45]. CD4+ cells differentiate into helper T cells that secrete effector molecules, such as cytokines, and may act by increasing the CTL (cytotoxic T lymphocyte) immune response, activating antibody-producing B cells, and modulating regulatory or inflammatory profiles [23]. As with cancer vaccines, CD4+ and CD8+ T cells travel to the tumor site and, upon finding corresponding antigens, kill tumor cells by cytotoxicity and cytokine production [45]. Therapeutic vaccine platforms against HPV-associated cancers in clinical and preclinical stages include bacterial and viral vectors, peptides, proteins, nucleic acids, and, more recently, whole cell-based vaccines [43][46]. Most platforms consisting of attenuated or inactivated pathogens generate a sufficient signal to the immune system to produce memory cells and antibodies [47]. However, they present problems associated with safety, such as virulence reversal, limitations related to efficacy against rapidly evolving pathogens, and demand for production systems with high levels of biosafety [48]. Nucleic acid vaccines, on the other hand, are third-generation vaccines that focus on the synthesis of an antigen of interest and presentation by MHC molecules, allowing the specific induction of cellular responses, which are important in the treatment of cancer, allergies, and autoimmune diseases [49][50]. Furthermore, the production of nucleic acid-based vaccines makes them quick to develop since there is no large-scale growth of highly pathogenic organisms, reducing the cost and the risk of contamination and infection [50].

This entry is adapted from the peer-reviewed paper 10.3390/pathogens11121444

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249.
  2. de Martel, C.; Georges, D.; Bray, F.; Ferlay, J.; Clifford, G.M. Global Burden of Cancer Attributable to Infections in 2018: A Worldwide Incidence Analysis. Lancet Glob. Health 2020, 8, e180–e190.
  3. Araldi, R.P.; Sant’Ana, T.A.; Módolo, D.G.; de Melo, T.C.; Spadacci-Morena, D.D.; de Cassia Stocco, R.; Cerutti, J.M.; de Souza, E.B. The Human Papillomavirus (HPV)-Related Cancer Biology: An Overview. Biomed. Pharmacother. 2018, 106, 1537–1556.
  4. de Villiers, E.-M.; Fauquet, C.; Broker, T.R.; Bernard, H.-U.; zur Hausen, H. Classification of Papillomaviruses. Virology 2004, 324, 17–27.
  5. Bernard, H.-U.; Burk, R.D.; Chen, Z.; van Doorslaer, K.; Hausen, H.Z.; de Villiers, E.-M. Classification of Papillomaviruses (PVs) Based on 189 PV Types and Proposal of Taxonomic Amendments. Virology 2010, 401, 70–79.
  6. Münger, K.; Baldwin, A.; Edwards, K.M.; Hayakawa, H.; Nguyen, C.L.; Owens, M.; Grace, M.; Huh, K. Mechanisms of Human Papillomavirus-Induced Oncogenesis. J. Virol. 2004, 78, 11451–11460.
  7. Li, N.; Franceschi, S.; Howell-Jones, R.; Snijders, P.J.F.; Clifford, G.M. Human Papillomavirus Type Distribution in 30,848 Invasive Cervical Cancers Worldwide: Variation by Geographical Region, Histological Type and Year of Publication. Int. J. Cancer 2011, 128, 927–935.
  8. Clifford, G.; Gallus, S.; Herrero, R.; Muñoz, N.; Snijders, P.; Vaccarella, S.; Anh, P.; Ferreccio, C.; Hieu, N.; Matos, E.; et al. Worldwide Distribution of Human Papillomavirus Types in Cytologically Normal Women in the International Agency for Research on Cancer HPV Prevalence Surveys: A Pooled Analysis. Lancet 2005, 366, 991–998.
  9. Hirth, J. Disparities in HPV Vaccination Rates and HPV Prevalence in the United States: A Review of the Literature. Hum. Vaccines Immunother. 2019, 15, 146–155.
  10. Shamseddine, A.A.; Burman, B.; Lee, N.Y.; Zamarin, D.; Riaz, N. Tumor Immunity and Immunotherapy for HPV-Related Cancers. Cancer Discov. 2021, 11, 1896–1912.
  11. Chesson, H.W.; Dunne, E.F.; Hariri, S.; Markowitz, L.E. The Estimated Lifetime Probability of Acquiring Human Papillomavirus in the United States. Sex. Transm. Dis. 2014, 41, 660–664.
  12. Best, S.R.; Niparko, K.J.; Pai, S.I. Biology of Human Papillomavirus Infection and Immune Therapy for HPV-Related Head and Neck Cancers. Otolaryngol. Clin. N. Am. 2012, 45, 807–822.
  13. Ghebre, R.; Berry-Lawhorn, J.M.; D’Souza, G. State of the Science: Screening, Surveillance, and Epidemiology of HPV-Related Malignancies. Am. Soc. Clin. Oncol. Educ. Book 2021, 41, 377–388.
  14. Chen, M.-L.; Wang, S.-H.; Wei, J.C.-C.; Yip, H.-T.; Hung, Y.-M.; Chang, R. The Impact of Human Papillomavirus Infection on Skin Cancer: A Population-Based Cohort Study. Oncologist 2021, 26, e473–e483.
  15. de Oliveira, T.H.A.; do Amaral, C.M.; de França São Marcos, B.; Nascimento, K.C.G.; de Miranda Rios, A.C.; Quixabeira, D.C.A.; Muniz, M.T.C.; da Silva Neto, J.C.; de Freitas, A.C. Presence and Activity of HPV in Primary Lung Cancer. J. Cancer Res. Clin. Oncol. 2018, 144, 2367–2376.
  16. Guo, L.; Liu, S.; Zhang, S.; Chen, Q.; Zhang, M.; Quan, P.; Sun, X.-B. Human Papillomavirus-Related Esophageal Cancer Survival: A Systematic Review and Meta-Analysis. Medicine 2016, 95, e5318.
  17. Lei, J.; Ploner, A.; Elfström, K.M.; Wang, J.; Roth, A.; Fang, F.; Sundström, K.; Dillner, J.; Sparén, P. HPV Vaccination and the Risk of Invasive Cervical Cancer. N. Engl. J. Med. 2020, 383, 1340–1348.
  18. Cheng, L.; Wang, Y.; Du, J. Human Papillomavirus Vaccines: An Updated Review. Vaccines 2020, 8, 391.
  19. Markowitz, L.E.; Schiller, J.T. Human Papillomavirus Vaccines. J. Infect. Dis. 2021, 224, S367–S378.
  20. Ornellas, P.; Ornellas, A.A. HPV Vaccination Is Fundamental for Reducing or Erradicate Penile Cancer | Opinion: NO. Int. Braz. J. Urol. 2018, 44, 862–864.
  21. Bewley, S. HPV Vaccination and Cervical Cancer Screening. Lancet 2022, 399, 1939.
  22. Catarino, R.; Petignat, P.; Dongui, G.; Vassilakos, P. Cervical Cancer Screening in Developing Countries at a Crossroad: Emerging Technologies and Policy Choices. World J. Clin. Oncol. 2015, 6, 281.
  23. Boilesen, D.R.; Nielsen, K.N.; Holst, P.J. Novel Antigenic Targets of HPV Therapeutic Vaccines. Vaccines 2021, 9, 1262.
  24. Khairkhah, N.; Bolhassani, A.; Najafipour, R. Current and Future Direction in Treatment of HPV-Related Cervical Disease. J. Mol. Med. 2022, 100, 829–845.
  25. Lee, J.; Kumar, S.A.; Jhan, Y.Y.; Bishop, C.J. Engineering DNA Vaccines against Infectious Diseases. Acta Biomater. 2018, 80, 31–47.
  26. Vetter, V.; Denizer, G.; Friedland, L.R.; Krishnan, J.; Shapiro, M. Understanding Modern-Day Vaccines: What You Need to Know. Ann. Med. 2018, 50, 110–120.
  27. Li, L.; Petrovsky, N. Molecular Adjuvants for DNA Vaccines. Curr. Issues Mol. Biol. 2017, 22, 17–40.
  28. Schijns, V.; Fernández-Tejada, A.; Barjaktarović, Ž.; Bouzalas, I.; Brimnes, J.; Chernysh, S.; Gizurarson, S.; Gursel, I.; Jakopin, Ž.; Lawrenz, M.; et al. Modulation of Immune Responses Using Adjuvants to Facilitate Therapeutic Vaccination. Immunol. Rev. 2020, 296, 169–190.
  29. Rosales, C.; Rosales, R. Prophylactic and Therapeutic Vaccines against Human Papillomavirus Infections. In Vaccines; Afrin, F., Hemeg, H., Ozbak, H., Eds.; InTech: London, UK, 2017; ISBN 978-953-51-3475-6.
  30. Tanner, R.; Villarreal-Ramos, B.; Vordermeier, H.M.; McShane, H. The Humoral Immune Response to BCG Vaccination. Front. Immunol. 2019, 10, 1317.
  31. Pulendran, B.; Arunachalam, P.S.; O’Hagan, D.T. Emerging Concepts in the Science of Vaccine Adjuvants. Nat. Rev. Drug Discov. 2021, 20, 454–475.
  32. Wang, J.W.; Roden, R.B. Virus-like Particles for the Prevention of Human Papillomavirus-Associated Malignancies. Expert Rev. Vaccines 2013, 12, 129–141.
  33. Buck, C.B.; Day, P.M.; Trus, B.L. The Papillomavirus Major Capsid Protein L1. Virology 2013, 445, 169–174.
  34. Hladíková, K.; Koucký, V.; Bouček, J.; Laco, J.; Grega, M.; Hodek, M.; Zábrodský, M.; Vošmik, M.; Rozkošová, K.; Vošmiková, H.; et al. Tumor-Infiltrating B Cells Affect the Progression of Oropharyngeal Squamous Cell Carcinoma via Cell-to-Cell Interactions with CD8+ T Cells. J. Immunother. Cancer 2019, 7, 261.
  35. Kim, S.S.; Shen, S.; Miyauchi, S.; Sanders, P.D.; Franiak-Pietryga, I.; Mell, L.; Gutkind, J.S.; Cohen, E.E.W.; Califano, J.A.; Sharabi, A.B. B Cells Improve Overall Survival in HPV-Associated Squamous Cell Carcinomas and Are Activated by Radiation and PD-1 Blockade. Clin. Cancer Res. 2020, 26, 3345–3359.
  36. Kamphorst, A.O.; Araki, K.; Ahmed, R. Beyond Adjuvants: Immunomodulation Strategies to Enhance T Cell Immunity. Vaccine 2015, 33, B21–B28.
  37. Boukhebza, H.; Bellon, N.; Limacher, J.M.; Inchauspé, G. Therapeutic Vaccination to Treat Chronic Infectious Diseases: Current Clinical Developments Using MVA-Based Vaccines. Hum. Vaccines Immunother. 2012, 8, 1746–1757.
  38. Eggenhuizen, P.J.; Ng, B.H.; Ooi, J.D. Treg Enhancing Therapies to Treat Autoimmune Diseases. Int. J. Mol. Sci. 2020, 21, 7015.
  39. Hu, Z.; Ott, P.A.; Wu, C.J. Towards Personalized, Tumour-Specific, Therapeutic Vaccines for Cancer. Nat. Rev. Immunol. 2018, 18, 168–182.
  40. Yim, E.-K.; Park, J.-S. The Role of HPV E6 and E7 Oncoproteins in HPV-Associated Cervical Carcinogenesis. Cancer Res. Treat. 2005, 37, 319.
  41. Yeo-Teh, N.; Ito, Y.; Jha, S. High-Risk Human Papillomaviral Oncogenes E6 and E7 Target Key Cellular Pathways to Achieve Oncogenesis. Int. J. Mol. Sci. 2018, 19, 1706.
  42. Paolini, F.; Curzio, G.; Cordeiro, M.N.; Massa, S.; Mariani, L.; Pimpinelli, F.; de Freitas, A.C.; Franconi, R.; Venuti, A. HPV 16 E5 Oncoprotein Is Expressed in Early Stage Carcinogenesis and Can Be a Target of Immunotherapy. Hum. Vaccines Immunother. 2017, 13, 291–297.
  43. Rumfield, C.S.; Roller, N.; Pellom, S.T.; Schlom, J.; Jochems, C. Therapeutic Vaccines for HPV-Associated Malignancies. ImmunoTargets Ther. 2020, 9, 167–200.
  44. Donà, M.G.; Di Bonito, P.; Chiantore, M.V.; Amici, C.; Accardi, L. Targeting Human Papillomavirus-Associated Cancer by Oncoprotein-Specific Recombinant Antibodies. Int. J. Mol. Sci. 2021, 22, 9143.
  45. Melssen, M.; Slingluff, C.L. Vaccines Targeting Helper T Cells for Cancer Immunotherapy. Curr. Opin. Immunol. 2017, 47, 85–92.
  46. Brinkman, J.A.; Hughes, S.H.; Stone, P.; Caffrey, A.S.; Muderspach, L.I.; Roman, L.D.; Weber, J.S.; Kast, W.M. Therapeutic Vaccination for HPV Induced Cervical Cancers. Dis. Markers 2007, 23, 337–352.
  47. Pan, C.; Yue, H.; Zhu, L.; Ma, G.; Wang, H. Prophylactic Vaccine Delivery Systems against Epidemic Infectious Diseases. Adv. Drug Deliv. Rev. 2021, 176, 113867.
  48. Qin, F.; Xia, F.; Chen, H.; Cui, B.; Feng, Y.; Zhang, P.; Chen, J.; Luo, M. A Guide to Nucleic Acid Vaccines in the Prevention and Treatment of Infectious Diseases and Cancers: From Basic Principles to Current Applications. Front. Cell Dev. Biol. 2021, 9, 633776.
  49. Zhang, N.; Nandakumar, K.S. Recent Advances in the Development of Vaccines for Chronic Inflammatory Autoimmune Diseases. Vaccine 2018, 36, 3208–3220.
  50. Zhang, C.; Maruggi, G.; Shan, H.; Li, J. Advances in MRNA Vaccines for Infectious Diseases. Front. Immunol. 2019, 10, 594.
More
This entry is offline, you can click here to edit this entry!
ScholarVision Creations