365
Views
6
CrossRef citations to date
0
Altmetric
Editorial

Promises and challenges of human papillomavirus vaccines for cervical cancer

Pages 1-5 | Published online: 10 Jan 2014

Cervical cancer is the leading cause of death in women living in developing countries, mostly in Asia, South America, Africa and Europe. Owing to improved personal hygiene and regular Pap smear screening programs it is becoming less of a threat to women’s health in developed countries. However, it is still the second most common cancer in women worldwide. In terms of etiology, infection in high-risk groups of human papillomavirus (HPV) has been known to be a major cause of cervical cancer incidence.

The burden of this disease includes high medical, financial and psychological costs. Fortunately, present advances in the understanding of HPV biology and molecular technology have allowed us to develop prophylactic vaccines against HPV infection. For instance, Gardasil™ (Merck, NJ, USA) composed of L1-based virus-like particles (VLPs) of HPV6, 11, 16 and 18, and Cervarix™ (VLP of HPV16 and 18; GlaxoSmithKline, Hertfordshire, UK) have been licensed as a prophylactic vaccine. These vaccines are estimated to reduce the incidence of cervical cancer and its precancerous diseases, thus improving the wellbeing of women. While these prophylactic vaccines appear very promising, some issues still remain unclear, for example, the duration of protective immunity after vaccination, availability of the vaccines owing to their high costs in developing countries, the extent of crossprotection against infection with nonvaccine HPV types, who and when to vaccinate, ideal combination of HPV subtypes, how vaccination affects screening programs and safety issues.

It has been well known that less than 5% of women infected with HPV develop dysplasia, cervical intraepithelial neoplasia (CIN) I, II and III, and carcinoma in situ in sequence prior to cervical cancer. What determines the malignancy versus clearance of HPV infection is believed to mainly be the host’s immune status. Despite current successes in preventing HPV infection through the vaccination protocols, their therapeutic efficacy is thought to be lacking in women with persistent HPV infection. This is due to the nature of HPV biology. For example, HPV expresses its regulatory proteins but not L1 coat proteins in the basal epithelial cells. Under this circumstance, L1 coat protein-specific antibodies generated by prophylactic vaccination are irrelevant to control of viral replication. Among the viral regulatory proteins, E6 and E7 are expressed constantly in cervical cancer cells and are also required for tumorigenesis and maintenance of cervical cancer Citation[1–4], therefore making them a potential therapeutic target for immunotherapy against cervical cancer and its precancerous diseases.

Cytotoxic T-lymphocytes as a positive factor for cervical cancer treatment

Concurrent chemoradiation therapy has been the current standard therapy in locally advanced cervical cancer. A large benefit of concurrent chemoradiation for survival, progression-free survival and local/distant control rates in cervical cancer patients has been suggested Citation[5,6]. In some of these patients, however, recurrence of the disease has been problematic. Both tumor regression and tumor response to conventional therapy (i.e., radical surgery, chemotherapy and radiotherapy) seem to be greatly influenced by the host immune status. For example, E6- and E7-specific cytotoxic T-lymphocyte (CTL) responses were detected more commonly in HPV16-positive women without CIN, as opposed to HPV16-positive women with CIN Citation[7]. CTLs also exerted a suppressive effect on development of squamous intraepithelial lesions in patients Citation[8]. Furthermore, the magnitude of HPV E6- and E7-specific T-cell responses correlates inversely with recurrence of diseases in CIN patients following ablative or excisional treatment Citation[9]. In addition, a high number of CD8+ tumor-infiltrating T cells were associated with a lack of lymph node metastasis in cervical cancer patients Citation[10]. These clinical findings correlate well with those of numerous animal studies, demonstrating a major effector role of CTL in either protecting or removing tumors in the experimental model systems Citation[11–19]. These animal and clinical studies collectively suggest that both induction and enhancement of E6- or E7-specific CTL responses, as well as an increase in CTL infiltration to tumor tissues, are critically important for obtaining a better response in patients with cervical cancer and its precancerous disease. Thus, immune therapy against cervical cancer should focus on how to augment tumor-specific CTL responses and increase CTL infiltration to cancer tissues.

Strategies for enhancing CTL responses

There have been numerous approaches to augmenting CTL responses to various vaccine types. In particular, an inverse correlation was observed between tumor size and the therapeutic efficacy of therapeutic vaccine-driven CTL responses in an HPV E6/E7-expressing tumor model Citation[18], suggesting that the curability of tumors of a larger size needs more vigorous tumor antigen-specific CTL activity. In this regard, there are two major strategies, direct and indirect, to obtain better CTL activity for tumor regression. These have been tested mostly in an animal model system.

Direct enhancement of CTL responses

Novel strategies have been tested to directly enhance tumor antigen-specific CTL responses in an HPV E6/E7-expressing tumor animal model. The use of substances (such as CpG-oligodeoxynucleotides [ODN], Quil-A, liposome-polycation DNA particle, dsRNA [poly(I:C)]) as an adjuvant for HPV16 E7 protein and peptide vaccines is one example Citation[12,20–23]. Conjugation of a lipid tail or helper T-cell epitope to E7 CTL peptides was also shown to improve an antigen-specific CTL response Citation[24,25]. In this regard, potent helper T-cell functions are required for the high frequency and long duration of CTL activity in vivoCitation[26]. Moreover, direct targeting of E7 antigens to dendritic cells by utilizing the E7/adenylate cyclase fusion proteins was shown to enhance anti-tumor CTL responses Citation[27].

In E7 DNA vaccines, modification of intracellular targeting patterns of E7 antigens, as well as use of molecular adjuvants (encoding anti-apoptotic proteins and serine protease inhibitors) Citation[15,17,28–33], is a typical example of modulating CTL responses. In the viral and bacterial vector delivery system, conjugation of E7 antigens to a costimulatory molecule CD40 ligand Citation[14,34] or addition of E7 antigens with IL-12 Citation[13,35,36] were both effective for antigen-specific CTL augmentation. In adoptive CTL therapy, infusion of cytokines is probably more effective for improving CTL activity in cervical cancer patients, This is based on the data of other tumor model studies showing that adoptive T-cell therapy was more successful with infusion with IL-2 or IL-15 Citation[37,38]. These direct CTL-enhancing approaches are applicable for treating patients with cervical cancer. Presently, more research efforts focusing on direct augmentation of CTL responses are ongoing.

Indirect enhancement of CTL activity

Indirect strategies for enhancing CTL activity include the methods of increasing antigen-specific CTL activity by removing any negative factors responsible for suppression of CTL induction and maintenance. The feasibility of using antibodies specific for Treg cells (known to inhibit CTL responses) for enhancing E7 vaccine-induced CTL responses for anti-tumor resistance has been demonstrated in an HPV animal tumor model (Sin J-I, Unpublished Data). These data correlate well with that of other tumor model studies in which either depletion or blockade of CD4+CD25+ Treg cells positively influenced vaccine-specific CTL responses and anti-tumor resistance Citation[39–43]. Similarly, the anti-tumor effectiveness of CTLA-4 (known to suppress CTL induction) blockade by delivering anti-CTLA-4 antibodies was demonstrated in melanoma cell vaccine models Citation[42,43]. Thus, antibody treatments for either removing Tregs or blocking production of inhibitory signals for CTL activation appear promising for indirectly influencing the anti-tumor CTL responses of therapeutic vaccines.

Clinical aspects of HPV vaccines in cervical cancer & its precancerous disease

Precancerous cervical cancer lesions are divided into several stages, such as dysplasia, CIN I, II and III, and carcinoma in situ, depending on histologic observation (disturbed epithelial architecture and progressive cellular atypia). Therapeutic vaccines (E7 peptide and heat-shock protein-conjugated E7 protein vaccines, viral vector delivery of E2 proteins and E7-fused VLP) have been tested for their effectiveness in treating the precancerous lesions Citation[44–47]. In these studies, the vaccines were successful for inducing antigen-specific CTL responses, as well as displaying objective clinical benefits, such as regression of HPV-associated precancerous lesions. These findings offer some optimism for the utility of therapeutic vaccines in clearing precancerous HPV lesions.

However, in cervical cancer patients, the efficacy of therapeutic vaccines was found to be lacking. It has been thought that 5-year survival rates decrease with the increasing cancer stages. In addition, primary tumor size, lymph node involvement and metastasis to other body parts are a limiting factor in treating cervical cancer. In these patients, therapeutic vaccines (e.g., peptide vaccines, viral vector delivery and dendritic cell vaccines) were well tolerated without any clinical benefits, even in the presence of HPV-specific CTL in some patients Citation[48–52]. In particular, some of these patients were immune-compromised (e.g., a lack or loss of MHC expression in cancer cells and an induction of immune tolerance). Thus, downregulation of the host immune system is a definite negative factor for treating cervical cancer patients by therapeutic vaccination. Presently, treating patients with cervical cancer by immunotherapy alone appears less promising.

Implications of HPV vaccines for cervical cancer treatment

After clinical practice of radical surgery, chemotherapy and radiation, cervical cancer cells tend to be metastasized to other parts of the body and/or form resistant cancer cells against the previous practice. These are the main causes of tumor recurrence. Tumor recurrence might be prevented by the addition of therapeutic vaccines to conventional therapy protocols. In this regard, therapeutic vaccines can induce systemic tumor-specific immune surveillance, which can control the occurrence of any resistant and metastatic tumor cell clones. This possibility was tested in an animal HPV16 E6/E7-expressing tumor model system. For instance, a dramatic therapeutic synergy between a cytotoxic drug, cisplatin, and HPV16 E7 subunit vaccines was observed Citation[19]. The combination strategy increased tumor cure and response rates, and decreased tumor recurrence through induction of long-term anti-tumor memory responses. A similar anti-tumor therapeutic effect was also observed when radiation and HPV16 E7 subunit vaccines were combined Citation[53]. In these two studies, therapeutic synergy was mediated through increased tumor cell sensitivity of drug-treated or radiated tumor cells to CTL-mediated killing. In particular, both chemotherapy and radiation increased the expression of tumor cell surface markers, which have been known to be important for facilitating CTL-tumor cell contacts (Sin J-I, Unpublished Data; Citation[53]). Chemotherapy and radiation also kill tumor cells directly through induction of apoptosis, allowing antigen-presenting cells to crosspresent and then generate tumor-specific CTL responses. This possibility was demonstrated in an HPV animal model Citation[54]. Finally, chemotherapy and radiation probably make cancer tissues more inflammatory, thereby inducing a greater infiltration of lymphocytes to the tissues, as previously shown by our group Citation[19,53]. Thus, it is reasonable to integrate therapeutic vaccines to conventional therapy modalities for a better cervical cancer intervention. However, it is still unclear how these animal study results translate to clinical cases. When used in combination with chemotherapeutic drugs (usually suppressing the host’s immune system), the timing and schedule of delivering therapeutic vaccines (as an active immune therapy) need to be carefully designed for adequate induction of an antigen-specific CTL response. However, in the case of adoptive transfer of CTLs, which are already activated in vitro, immune suppression by chemotherapeutic drugs might be beneficial, as the drugs tend to inhibit the host immune cells, including Tregs (known to counteract CTL activity). In this case, the fully activated and then adoptively transferred CTLs are unlikely to be affected by the drugs. This possibility was tested by our group using an HPV E6/E7-expressing tumor model [Sin J-I, Unpublished Data]. Therefore, a careful evaluation of the effects of drugs or radiation on the tumor, tumor microenvironment and the immune system is essential for obtaining optimal therapeutic benefits in cervical cancer patients.

In summary, presently available prophylactic vaccines are expected to reduce HPV infection rates dramatically, thus, protecting women from cervical cancer incidence. Therapeutic vaccines, however, present us with far more challenges than prophylactic vaccines. These challenges include the immune-compromised state of cervical cancer patients and the immune-evasive nature of cancer cells themselves. In spite of these challenges, there have been many promising preclinical data demonstrating an improved anti-tumor efficacy of therapeutic vaccines in combination with chemotherapy and radiotherapy. Although the ultimate outcome of these preclinical results in clinical situations is not yet known, there is no doubt that HPV vaccines can contribute to the eradication of cervical cancer and, eventually, the wellbeing of women worldwide.

Financial & competing interests disclosure

This work was supported by the Korea Research Foundation Grant (KRF-2004–041-E00094). The author has no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

References

  • Munger K, Phelps WC, Bubb V, Howley PM, Schlegel R. The E6 and E7 genes of the human papillomavirus type 16 together are necessary and sufficient for transformation of primary human keratinocytes. J. Virol.63, 4417–4421 (1989).
  • Dyson N, Howley PM, Munger K, Harlow E. The human papillomavirus-16 E7 oncoprotein is able to bind the retinoblastoma gene product. Science243, 934–937 (1989).
  • Scheffner M, Munger K, Bryne JC, Howley PM. The state of the p53 and retinoblastoma genes in human cervical carcinoma cell lines. Proc. Natl Acad. Sci. USA88, 5523–5527 (1991).
  • Werness BA, Levine AJ, Howley PM. Association of HPV type 16 and 18 E6 protein with p53. Science248, 76–79 (1990).
  • Rose P, Bundy BN, Watkins EB et al. Concurrent cisplatin-based radiotherapy and chemotherapy for locally advanced cervical cancer. N. Engl. J. Med.340, 1144–1153 (1999).
  • Green JA, Kirwan JM, Tierney JF et al. Survival and recurrence after concomitant chemotherapy and radiotherapy for cancer of the uterine cervix: a systematic review and meta-analysis. Lancet358, 781–786 (2001).
  • Nakagawa M, Stites DP, Farhat S et al. Cytotoxic T lymphocyte responses to E6 and E7 proteins of human papillomavirus type 16: relationship to cervical intraepithelial neoplasia. J. Infect. Dis.175, 927–931 (1997).
  • Nakagawa M, Stites DP, Patel S et al. Persistence of human papillomavirus type 16 infection is associated with lack of cytotoxic T lymphocyte response to the E6 antigens. J. Infect. Dis.182, 595–598 (2000).
  • Sarkar AK, Tortolero-Luna G, Follen M, Sastry KJ. Inverse correlation of cellular immune responses specific to synthetic peptides from the E6 and E7 oncoproteins of HPV-16 with recurrence of cervical intraepithelial neoplasia in a cross-sectional study. Gynecol. Oncol.99, S251–S261 (2005).
  • Piersma SJ, Jordanova ES, van Poelgeest MI et al. High number of intraepithelial CD8+ tumor-infiltrating lymphocytes is associated with the absence of lymph node metastases in patients with large early-stage cervical cancer. Cancer Res.67, 354–361 (2007).
  • Cheng WF, Hung CF, Hsu KF et al. Cancer immunotherapy using Sindbis virus replicon particles encoding a VP22-antigen fusion. Hum. Gene Ther.13, 553–568 (2002).
  • Kim TY, Myoung HJ, Kim JH et al. Both E7 and CpG-ODN are required for protective immunity against challenge with human papillomavirus 16 (E6/E7)-immortalized tumor cells: involvement of CD4+ and CD8+ T cells in protection. Cancer Res.62, 7234–7240 (2002).
  • Ahn WS, Bae SM, Kim TY et al. A therapy modality using recombinant IL-12 adenovirus plus E7 protein in a human papillomavirus 16 E6/E7-associated cervical cancer animal model. Hum. Gene Ther.14, 1389–1399 (2003).
  • Zhang L, Tang Y, Akbulut H, Zelterman D, Linton PJ, Deisseroth AB. An adenoviral vector cancer vaccine that delivers a tumor-associated antigen/CD40-ligand fusion protein to dendritic cells. Proc. Natl Acad. Sci. USA100, 15101–15106 (2003).
  • Kim TW, Hung CF, Boyd DA et al. Enhancement of DNA vaccine potency by coadministration of a tumor antigen gene and DNA encoding serine protease inhibitor-6. Cancer Res.64, 400–405 (2004).
  • Kim TW, Hung CF, Kim JW et al. Vaccination with a DNA vaccine encoding herpes simplex virus type 1 VP22 linked to antigen generates long-term antigen-specific CD8-positive memory T cells and protective immunity. Hum. Gene Ther.15, 167–177 (2004).
  • Kim MS, Sin JI. Both antigen optimization and lysosomal targeting are required for enhanced anti-tumour protective immunity in a human papillomavirus E7-expressing animal tumour model. Immunol.116, 255–266 (2005).
  • Sin JI, Hong SH, Park YJ, Park JB, Choi YS, Kim, MS. Anti-tumor therapeutic effects of E7 subunit and DNA vaccines in an animal cervical cancer model: anti-tumor efficacy of E7 therapeutic vaccines is dependent on tumor sizes, vaccine doses, and vaccine delivery routes. DNA Cell Biol.25, 277–286 (2006).
  • Bae SH, Park YJ, Choi YS, Park JB, Kim MS, Sin JI. Therapeutic synergy of human papillomavirus E7 subunit vaccines plus cisplatin in an animal tumor model: causal involvement of increased sensitivity of cisplatin-treated tumors to CTL-mediated killing in therapeutic synergy. Clin. Cancer Res.13, 341–349 (2007).
  • Fernando GJP, Murray B, Zhou J, Frazer IH. Expression, purification and immunological characterization of the transforming protein E7, from cervical cancer-associated human papillomavirus type 16. Clin. Exp. Immunol.115, 397–403 (1999).
  • Cui Z, Huang L. Liposome-polycation-DNA (LPD) particle as a carrier and adjuvant for protein-based vaccines: therapeutic effect against cervical cancer. Cancer Immunol. Immunother.54, 1180–1190 (2005).
  • Zwaveling S, Ferreira Mota SC, Nouta J et al. Established human papillomavirus type 16-expressing tumors are effectively eradicated following vaccination with long peptides. J. Immunol.169, 350–358 (2002).
  • Cui Z, Qiu F, Synthetic double-stranded RNA poly(I:C) as a potent peptide vaccine adjuvant: therapeutic activity against human cervical cancer in a rodent model. Cancer Immunol. Immunother.54, 1180–1190 (2005).
  • Livingston BD, Crimi C, Grey H et al. The hepatitis B virus-specific CTL responses induced in humans by lipopeptide vaccination are comparable to those elicited by acute viral infection. J. Immunol.159, 1383–1392 (1997).
  • Gahery-Segard H, Pialoux G, Charmeteau B et al. Multiepitopic B- and T-cell responses induced in humans by a human immunodeficiency virus type 1 lipopeptide vaccine. J. Virol.74, 1694–1703 (2000).
  • Mortara L, Gras-Masse H, Rommens C, Venet A, Guillet JG, Bourgault-Villada I. Type 1 CD4+ T-cell help is required for induction of antipeptide multispecific cytotoxic T lymphocytes by a lipopeptidic vaccine in rhesus macaques. J. Virol.73, 4447–4451 (1999).
  • Preville X, Ladant D, Timmerman B, Leclerc C. Eradication of established tumors by vaccination with recombinant Bordetella pertussis adenylate cyclase carrying the human papillomavirus 16 E7 oncoprotein. Cancer Res.65, 641–649 (2005).
  • Ji H, Wang TL, Chen CH et al. Targeting human papillomavirus type 16 E7 to the endosomal/lysosomal compartment enhances the anti-tumor immunity of DNA vaccines against murine human papillomavirus type 16 E7-expressing tumors. Hum. Gene Ther.10, 2727–2740 (1999).
  • Hung CF, Cheng WF, Hsu KF et al. Cancer immunotherapy using a DNA vaccine encoding the translocation domain of a bacterial toxin linked to a tumor antigen. Cancer Res.61, 3698–3703 (2001).
  • Cheng WF, Hung CF, Chai CY et al. Tumor-specific immunity and anti-angiogenesis generated by a DNA vaccine encoding calreticulin linked to a tumor antigen. J. Clin. Invest.108, 669–678 (2001).
  • Hung CF, He L, Juang J, Lin TJ, Ling M, Wu TC. Improving DNA vaccine potency by linking Marek’s disease virus type 1 VP22 to an antigen. J. Virol.76, 2676–2682 (2002).
  • Hung CF, Cheng WF, He L et al. Enhancing major histocompatibility complex class I antigen presentation by targeting antigen to centrosomes. Cancer Res.63, 2393–2398 (2003).
  • Kim TW, Hung CF, Ling M et al. Enhancing DNA vaccine potency by coadministration of DNA encoding anti-apoptotic proteins. J. Clin. Invest.112, 109–117 (2003).
  • Tang Y, Zhang L, Yuan J et al. Multistep process through which adenoviral vector vaccine overcomes anergy to tumor-associated antigens. Blood104, 2704–2713 (2004).
  • Kim TG, Kim CH, Won EH et al. CpG-ODN-stimulated dendritic cells act as a potent adjuvant for E7 protein delivery to induce antigen-specific anti-tumor immunity in a HPV 16 (E6/E7)-associated tumor animal model. Immunology112, 117–125 (2004).
  • Bermudez-Humaran LG, Cortes-Perez NG, Lefevre F et al. A novel mucosal vaccine based on live Lactococci expressing E7 antigen and IL-12 induces systemic and mucosal immune responses and protects mice against human papillomavirus type 16-induced tumors. J. Immunol.175, 7297–7302 (2005).
  • Roychowdhury S, May KF Jr, Tzou KS et al. Failed adoptive immunotherapy with tumor-specific T cells: reversal with low-dose interleukin 15 but not low-dose interleukin 2. Cancer Res.64, 8062–8067 (2004).
  • Klebanoff CA, Finkelstein SE, Surman DR et al. IL-15 enhances the in vivo antitumor activity of tumor-reactive CD8+ T cells. Proc. Natl Acad. Sci. USA101, 1969–1974 (2004).
  • Comes A, Rosso O, Orengo AM et al. CD25+ Regulatory T cell depletion augments immunotherapy of micrometastases by an IL-21-secreting cellular vaccine. J. Immunol.176, 1750–1758 (2006).
  • Viehl CT, Moore TT, Liyanage UK et al. Depletion of CD4+CD25+ regulatory T cells promotes a tumor-specific immune response in pancreas cancer-bearing mice. Ann. Surg. Oncol.13, 1252–1258 (2006).
  • Prasad SJ, Farrand KJ, Matthews SA, Chang JH, McHugh RS, Ronchese F. Dendritic cells loaded with stressed tumor cells elicit long-lasting protective tumor immunity in mice depleted of CD4+CD25+ regulatory T cells. J. Immunol.174, 90–98 (2005).
  • van Elsas A, Hurwitz AA, Allison JP. Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation. J. Exp. Med.190, 355–366 (1999).
  • Quezada SA, Peggs KS, Curran MA, Allison JP. CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells. J. Clin. Invest.116, 1935–1945 (2006).
  • Muderspach L, Wilczynski S, Roman L et al. A Phase I trial of a human papillomavirus (HPV) peptide vaccine for women with high-grade cervical and vulvar intraepithelial neoplasia who are HPV 16 positive. Clin. Cancer Res.6, 3406–3416 (2000).
  • Garcia-Hernandez E, Gonzalez-Sanchez JL, Andrade-Manzano A et al. Regression of papilloma high-grade lesions (CIN 2 and CIN 3) is stimulated by therapeutic vaccination with MVA E2 recombinant vaccine. Cancer Gene Ther.13, 592–597 (2006).
  • Roman L, Wilczynski S, Muderspach LI et al. A Phase II study of hsp-7 (SGN-00101) in women with high-grade cervical intraepithelial neoplasia. Gynecol. Oncol.106, 558–566 (2007).
  • Kaufmann, A., Nieland, JD, Jochmus I et al. Vaccination trial with HPV16 L1E7 chimeric virus-like particles in women suffering from high grade cervical intraepithelial neoplasia (CIN 2/3). Int. J. Cancer121, 2794–2800 (2007).
  • van Driel WJ, Ressing ME, Kenter GG et al. Vaccination with HPV 16 peptides of patients with advanced cervical carcinoma: clinical evaluation of a Phase I–II trial. Eur. J. Cancer35, 946–952 (1999).
  • Ferrara A, Nonn M, Sehr P et al. Dendritic cell-based tumor vaccine for cervical cancer II: results of a clinical pilot study in 15 individual patients. J. Cancer Res. Clin. Oncol.129, 521–530 (2003).
  • Santin AD, Bellone S, Palmieri M et al. HPV 16/18 E7-pulsed dendritic cell vaccination in cervical cancer patients with recurrent disease refractory to standard treatment modalities. Gynecol. Oncol.100, 469–478 (2006).
  • Borysiewicz LK, Fiander A, Nimako M et al. A recombinant vaccinia virus encoding human papillomavirus types 16 and 18, E6 and E7 proteins as immunotherapy for cervical cancer. Lancet347, 1523–1527 (1996).
  • Kaufmann AM, Stern PL, Rankin EM et al. Safety and immunogenicity of TA-HPV, a recombinant vaccinia virus expressing modified human papillomavirus (HPV)-16 and HPV-18 E6 and E7 genes, in women with progressive cervical cancer. Clin. Cancer Res.8, 3676–3685 (2002).
  • Ye GW, Park JB, Park YJ, Choi YS, Sin JI. Increased sensitivity of radiated murine cervical cancer tumors to E7 subunit vaccine-driven CTL-mediated killing induces synergistic anti-tumor activity. Mol. Ther.15, 1564–1570 (2007).
  • Tseng C, Monie A, Wu CY et al. Treatment with proteasome inhibitor bortezomib enhances antigen-specific CD8+ T-cell-mediated anti-tumor immunity induced by DNA vaccination. J. Mol. Med.86, 899–908 (2008).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.