Skip to main content

Immunologic treatments for precancerous lesions and uterine cervical cancer

Abstract

Development of HPV-associated cancers not only depends on efficient negative regulation of cell cycle control that supports the accumulation of genetic damage, but also relies on immune evasion that enable the virus to go undetected for long periods of time. In this way, HPV-related tumors usually present MHC class I down-regulation, impaired antigen-processing ability, avoidance of T-cell mediated killing, increased immunosuppression due to Treg infiltration and secrete immunosuppressive cytokines. Thus, these are the main obstacles that immunotherapy has to face in the treatment of HPV-related pathologies where a number of different strategies have been developed to overcome them including new adjuvants. Although antigen-specific immunotherapy induced by therapeutic HPV vaccines was proved extremely efficacious in pre-clinical models, its progression through clinical trials suffered poor responses in the initial trials. Later attempts seem to have been more promising, particularly against the well-defined precursors of cervical, anal or vulvar cancer, where the local immunosuppressive milieu is less active. This review focuses on the advances made in these fields, highlighting several new technologies (such as mRNA vaccine, plant-derived vaccine). The most promising immunotherapies used in clinical trials are also summarized, along with integrated strategies, particularly promising in controlling tumor metastasis and in eliminating cancer cells altogether.

After the early promising clinical results, the development of therapeutic HPV vaccines need to be implemented and applied to the users in order to eradicate HPV-associated malignancies, eradicating existing perception (after the effectiveness of commercial preventive vaccines) that we have already solved the problem.

Introduction

Cervical cancer is the third most common cancer in women and the fifth most common overall cancer worldwide as age standardized incidence rate in both sexes combined [1]. The prime causal factor of the disease is a persistent infection with high-risk human papillomavirus (HPV), with individuals failing to mount a sufficient immune response against the virus. The high-risk HPV genome encodes three oncoproteins, E5, E6 and E7, the last two oncoproteins are constitutively expressed in high-grade lesions and cancer, and are required for the onset and maintenance of the malignant phenotype. About 170 HPV genotypes have been identified and 40 can infect the ano-genital area: uterine cervix, vulva, vaginal wall, penis and anus. HPVs are classified as high-risk types, commonly associated with cancer, and low-risk, mostly identified in condyloma acuminatum. The International Agency for Research on Cancer (IARC) conducted a study on over 30,000 cervical cancers that showed HPV 16, 18, 58, 33, 45, 31, 52, 35, 59, 39, 51, 56, to be the most common types associated with invasive cervical cancer with HPV 16 accounting for over 50% and HPV 16 and 18 for >70% worldwide [2]. Epidemiological data report that HPV infection occurs at least once during lifespan in about 75% of U.S. women [3], and natural history shows that most HPV infections resolve spontaneously, while in some women infection persists and progresses to cervical cancer. The incidence of high-grade cervical intraepithelial neoplasia (CIN 3) is about 1-2 per 10 females with low-grade CIN, and without treatment about one third progresses to cervical cancer [4, 5].

Cervical carcinogenesis is a multi-step process, which starts with viral infection and requires the establishment of persistent HPV infection. Papillomavirus persistence is favored by at least three major factors: the virus life cycle that takes place away from dermal immune cells, the absence of virus-induced cell lysis with, in turn, no or weak inflammatory response and finally the altered immune response induced by viral proteins. Thus, cancer development depends not only on efficient negative regulation of cell cycle control supporting the accumulation of genetic damage, but also on immune evasion that enables the virus to lie undetected for a long time. Many studies confirm that persistent HPV infection is the most important risk factor, mainly with high-risk subtypes [5]. Studies in HIV women or in patients treated with immunosuppressive agents reported an increased incidence of CIN lesions, suggesting an important role of cell-mediated immune response against HPV antigens [6, 7]. The role of systemic and local mucosal immune responses to HPV antigens is controversial. Some studies suggest a positive association between systemic cell-mediated immune responses and the regression of CIN [8]. Moreover, antibody responses to the major viral capsid protein, L1, can be detected by about 6 months after infection and may be observed up to 5 years later in women who have been cleared from infection. Type specific L1 antibody responses have also been detected in persistent disease and cancer in about half of the patients [9, 10].

A number of escape factors may affect the natural immune response against HPV proteins, together with the loss of correct signals from immune system to activate adaptive immune system. Indeed, optimal activation of adaptive immunity and generation of specific CD4 T helper 1 type immunity supporting development of CD8 cytotoxic T cells against viral early proteins, like E2, E6, E7, is critical for virus clearance in basal epithelial cells. T helper cells also support optimal activation of B cells, with secreting HPV capsid type specific neutralizing antibodies, which can protect against subsequent infections at mucosal and systemic levels [11]. Spontaneous regression mainly occurs in lesions infiltrated by CD4+ and cytotoxic CD8+ T cells and it is also associated with circulating HPV early antigen-specific CD4+ and CD8+ T cells, which also have a favorable prognostic significance [1215].

In the early phases of carcinogenesis the three oncogenes of the virus E5, E6 and E7 play an important role in immune evasion. In particular, the E5 protein [16] seems to further facilitate the virus-induced immune escape by down-regulating MHC/HLA class I and II [17, 18] and inducing a reduction in recognizing CD8+ T cells, at least in vitro[19]. This down regulation does not affect the HLA molecules (HLA-C/E) whose presence on the cell surface to avoid natural killer action is essential [20, 21]. Several pieces of experimental evidence show that neither synthesis nor transport to the cell surface of HLA-C/E is affected by E5 expression, leading to the conclusion that E5 selectively inhibits surface expression of HLA-A and HLA-B [17]. In this manner, high risk HPVs are potentially capable of avoiding both CTL and NK cell killing, favoring persistent infection. In the late phases, when integration of the virus (frequently with loss of E2 and E5 genes) into the host genome blocks the productive life cell cycle, favoring immortalization and mutation acquisition, the E6 and E7 still play a fundamental role: i) high-risk E6 reduces the surface expression of CDH1 by epithelial cells; ii) E6 and E7 inhibit the transcription of toll-like receptor (TLR) 9, necessary to activate antigen-presenting cells as part of innate immune response; iii) E7 reduces expression of transporter associated with antigen processing 1 (TAP1), a component of the presentation and processing pathway, so blocking the activation of specific T lymphocytes; iv) high-risk HPVs down-regulates the expression of pro-inflammatory cytokines [22].

In addition, therapeutic T cell effector mechanisms are limited due to the following: changes in local immunity; the production of cytokines such as Interleukin (IL)-10 or transforming growth factor (TGF-beta); increased number of regulatory T cells (Tregs) and to immunosuppressive myeloid cells that are commonly observed in HPV related cancers. Moreover, frequent mutational events in cancer include HLA loss of expression, with subsequent escape of tumor cells [23, 24].

In this manner, HPV-related tumors usually present MHC class I down-regulation, impaired antigen-processing ability, avoidance of T-cell mediated killing, increased immunosuppression due to Treg infiltration, and secrete immunosuppressive cytokines [25].

These are the main obstacles faced when achieving a valid immunotherapy against HPV-related pathologies where a number of different strategies have been developed in order to overcome them including adjuvants. Adjuvant is a term derived from the Latin word, adjuvare, which means to aid or to help. Historical boosting humoral immune response was the goal of the developed adjuvants and, as a result, many commonly used adjuvants are effective in elevating serum antibody titers, but do not elicit significant Th1 responses or cytotoxic T lymphocytes (CTLs). However, certain adjuvants have recently been demonstrated to be able to induce cellular immunity which are summarized in Table 1 according to their mechanism of action [26].

Table 1 List of adjuvants by their dominant mechanism of action

Conceptually, immunity can be utilized in a therapeutic setting in two ways: first, by using specific natural o synthetic antibodies against defined targets, or second, by inducing an immune response in the organism against specific antigens (preventive and therapeutic vaccines). In particular, for HPV-induced lesions and cancer viral antigens or/and virus-induced host antigens can be targeted by these approaches. Indeed, once a patient is infected with HPV there is no effective way to cure persistent HPV infection which is the first step towards the development of pre-cancerous lesions. It was estimated that even with mass vaccination through commercially highly effective preventive quadrivalent or bivalent HPV vaccines [2731], it will take at least 20 years before the incidence and prevalence of cervical cancer significantly decreases due to the slow rate of carcinogenesis. Furthermore, during this period of time many people will be infected by high risk HPV that will consequently increase the burden of established HPV infections and HPV-associated diseases that lie undetected, untreated and slowly progressing toward malignant transformation.

Thus, there is great need for the development of novel therapeutic intervention to control HPV-associated disease and cancers. Given that existing treatments [3234] are partially effective in cancers or pre-neoplastic lesions, and useless in persistent infections, immune therapies may represent a valid tool.

The following paragraphs summarize these issues and focus on the treatment (clinical trials) of already established infections (Table 2) or cancer (Table 3) where preventive intervention by vaccine has not effect.

Table 2 Clinical trials for HPV-associated pre-neoplastic lesions
Table 3 Clinical trials for HPV-associated cancer

Therapeutic antibodies

The use of intracellular antibodies (intrabodies) to inhibit protein function holds promise for the treatment of human diseases. The difficulties associated with the development of intrabody-based therapies are similar to those that hamper the development of protein inhibitors in general, and reflect problems in generating reagents that are effective and specific. This effectiveness and specificity can be achieved by using intrabodies to combat intracellular parasites like viruses. Indeed, viruses express proteins that are directly involved in causing disease and are clearly diverse from those of the host cell. For viruses that cause only local infections, such as HPV, the intrabody approach may be more appropriate. Furthermore, not only the infected cells but also the transformed cells require the continuous expression of some HPV proteins, particularly the oncogenes E6 and E7. This is most evident in Hela cells, derived from an HPV-associated malignancy in the 1950s, which still almost 50 years later require the expression of these oncogenes for their growth in tissue culture [35, 36].

Intrabodies against the E6 [37] and E7 [38] of HPV have been produced and proved effective in in vitro cancer cell models. More recently, an intrabody against the E7 of HPV 16 was proved to block tumor growth in animal models [39]. Given the accessibility of HPV-associated lesions to topical therapy, preclinical results suggest that large interfering molecules, such as intrabodies, may be useful inhibitors of viral protein–protein interactions and particularly appropriate for the treatment of HPV-associated diseases. However, no clinical study has been published so far.

Instead of using intrabodies, the utilization of monoclonal antibodies against membrane-expressed antigens is suggested, that may be induced by the HPV, i.e. Epidermal Growth Factor Receptor (EGFR). Monoclonal antibodies anti EGFR are already in clinical use and have been fully reviewed elsewhere [32]. However, beside HPV induction other membrane-associated antigens can be found in transformed cervical cells and may be targeted by monoclonal antibodies. An example of these antibodies is the adecatumum (MT201), a humanized monoclonal antibody targeting epithelial cell adhesion molecules, showing activity in cervical cancer cell lines over-expressing epithelial cell adhesion molecule (EpCAM) [40]. This activity may suggest a hypothetical clinical employment of this monoclonal antibody which has already been performed with monoclonal antibodies against EGFR.

Therapeutic vaccines

Therapeutic vaccines aim to eradicate or reduce already infected cells by stimulating cytotoxic T cells against target infected cells and up-regulating MHC Class I expression. Vaccine-mediated immune strategies could be directed toward at least two different stages of the oncogenic infection: firstly, infection and then secondly, the established infection. By eliciting neutralizing antibody responses the prophylactic vaccines challenge the first infection by inhibiting the HPV to bind to the cell or the early phases of viral entry. These vaccines are already in clinical use and will not be discussed in this review.

The therapeutic vaccines should fight already infected cells, and could be tailored based on the presence of episomal replicating virus or integrated viral sequences. In the first case, the vaccine targets could be all the early proteins, in the second case only the E6-E7 proteins appear to be a realistic target of intervention [41]. In addition, as HPV 16 accounts for over 50% of invasive cancer worldwide, the clinical studies mainly focused on the E6 and E7 proteins of this HPV [2].

In the experimental model an effective immunotherapy administered before tumor challenge includes an antigen-specific component, whereas an effective immunotherapy after tumor challenge can be achieved through the enhancement of either innate or adaptive immunity, and seems to be optimal with both. Therefore, valid therapeutic vaccines must achieve this goal. Trials of immunotherapy in patients with HPV associated pre-malignancy are expected to be more effective than in cancer patients, since the impaired antigen presentation by cervical cancer cells due to mutations in MHC and TAP genes may render the immunotherapy less effective. Although there are potential immune-evasive mechanisms that are attributable to the HPV infection itself [42].

Examples of those therapeutic vaccines, which have reached phases I and II clinical trials, are presented in the subsequent paragraphs according to the different formulations.

Protein/peptide-based vaccines

To date, several protein or peptide-based vaccines are either undergoing clinical evaluation or are in development. A major limitation to peptide-based vaccines is the HLA restriction that can be overcome by the use of whole protein-based vaccines, which harbor multiple immunogenic epitopes, binding various allelic HLA molecules. On the other hand, protein-based vaccines are generating predominantly antibody responses rather than CTL responses because proteins are processed through the endocytic/MHC class II pathway. In addition, both peptides and proteins are poorly immunogenic. Therefore, most of the research in this area was focused on the co-administration of adjuvant immune-enhancing agents such as chemokines, cytokines, and co-stimulatory molecules to enhance the potency of the vaccine. In particular saponin-based [43] or liposome–based (LPD) formulations [44], or TLR agonists [45] were employed as adjuvants for protein vaccines. Recently, the fusion of the beta-1,3-1,4-glucanase (LicKM) of Clostridium thermocellum bacterial protein to the HPV E7 protein produced an antigen with strong intrinsic adjuvating activity, indicating that this manipulation of the antigen may elicit some unknown helpful functions [46, 47]. Many other fusion proteins were reported to elicit some adjuvating activities such as Mycobacteria-derived heat-shock proteins (Hsp) [48, 49], truncated Pseudomonas aeruginosa exotoxin A [50], Bordetella pertussis adenylate cyclase [51], and the cell penetrating peptide Limulus polyphemus protein [52]. In addition, alternative delivery system such as electroporation can further improve the activity of this adjuvating protein vaccine [45].

Peptide-based vaccines need to increase not only the poor immunogenicity level but also the obstacle of MHC restriction. TLR agonists have also been explored as adjuvants for peptide-based HPV vaccines because of their capability to activate both innate and adaptive immunity. Vaccines consisting in CTL and or TH epitope adjuvated with TLR 9 [53], TLR4 or [54] and TLR3 [55] agonists demonstrated their efficacy in mouse models. This activity was demonstrated also by utilizing a CTL epitope fused to a T-helper epitope, pan-DR epitope (PADRE) [56]. These results suggest that adjuvants targeting dendritic cells are useful in peptide-based vaccines. Indeed, a new strategy (TriVax) based on the administration of co-stimulatory anti-CD40 monoclonal, TLR agonist Polyinosinic-polycytidylic acid [Poly(I:C)] and CD8+ T-cell epitope HPV 16 E7 (aa49-57) was able to induce tumor clearance in two HPV-induced murine cancer models [57]. Many of these protein/peptide-based vaccines moved to clinical trials where all of them indicated low toxicity and a good safety profile, but a strong discordance exists between immune and clinical responses, reinforcing the need of further improvement to the vaccination.

However, among the protein-based vaccine candidates, SGN-00101 vaccine, a fusion protein consisting of Hsp from Mycobacterium bovis and HPV 16 E7, has generated considerable interest. As a single-agent therapy, in both phase I and phase II clinical trials, fusion protein was able to induce regression of lesions in anal high-grade squamous intraepithelial lesions [58], recurrent respiratory papillomatosis [59], and CIN 2-3 [6062]. In addition, phase II clinical trial with TA-CIN, a fusion protein-based vaccine expressing HPV 16 L2-E6-E7 conjugated proteins, in conjunction with topical application of TLR agonist Imiquimod, an imidazoquinoline amine, showed high levels of CD4+ and CD8+ T cells locally in patients with high-grade vulvar intra-epithelial neoplasia (VIN) [63]. One year after treatment 63% patients showed complete clinical regression of VIN lesions, associated with HPV clearance in 36% of the subjects. More recently Genticel’s vaccine candidate Procervix utilizing the adenylate cyclase (cyaa) technology, a protein vector that delivers the E7 antigens from HPV 16 and HPV 18 was proved safe in phase I trial. The company is now (March 2014) launching its phase II trial in women infected with high-risk HPV before the appearance of high- grade cervical lesions. This trial is the first ever recruiting HPV infected women with no cervical lesions and it would give information about a vaccine that may close the gap between preventive vaccines and later stage therapeutic options [64].

Since peptide-based vaccines are stable, easy to produce and have a high safety profile, many clinical trials have been reported. Research has focused on addressing the main limitations of peptide-based vaccines, namely their low immunogenicity, and the enhancement of the immunogenicity of peptide-based vaccines has been explored in clinical studies. The PADRE universal T-helper peptide was utilized to increase the activity of CTL epitopes encoding HPV 16 E7 that was presented by HLA-A*0201 (50% of the general population). These vaccines failed to mount a valid immune response in women with late stage cervical cancer [65, 66]. More promising results were obtained in HLA-A2-positive patients with CIN/VIN 2/3 [67], where HPV E7 lipopeptide (aa 86-93)/PADRE was able to stimulate an immune response and led to complete regression of CIN lesions in 3 of 17 valuable patients. However, these vaccines presented limitations due to the HLA restriction. Therefore, this limitation was avoided by using long peptide which prompted their utilization in clinical trials. In cervical cancer patients who had undergone resection, the use of immunization with 13 overlapping long peptides spanning the entire sequence of HPV 16 E6 and E7 mixed with Montanide ISA 51 clearly revealed immunization-driven IFN-gamma production in enzyme-linked immunospot (ELISPOT) assay after completing the protocol [68]. When this same platform was tested in immunizing cervical cancer patients with active disease, both CD4+ and CD8+ T-cell IFN-gamma responses were detected toward both antigens [69]. Moreover, significant increases in proliferative capacity were also noted in responding T cells, reminiscent of the type of response noted in spontaneous regression [69]. This vaccine was well tolerated with few side effects: minor swelling at the injection site and flu-like symptoms. Phase II clinical trials of this vaccine in histologically confirmed HPV 16-positive high-grade VIN patients had a complete regression of their lesion after 3 or 4 vaccinations with HPV 16 E6/E7 overlapping peptide vaccine [70]. In the non-responders to the vaccine, an increased number of HPV 16-specific CD4 + CD25 + Foxp3+ Treg cells was ascertained [71].

The presence of these Foxp3+ T cells is linked to impaired immunity in malignancies. The efficacy of this vaccine was also demonstrated in a placebo-controlled randomized Phase II study showing an increased number of HPV 16-specific T cells in patients with HPV 16+ high squamous intraepithelial lesion (HSIL) [72].

Plant-derived/produced vaccines

Plant molecular pharming represents a well-established biotechnology area that includes the production of protein biopharmaceuticals such as enzymes, hormones, antibodies, and vaccine antigens in plant systems. Plant-produced proteins represent a significant fraction of pharmaceuticals in advanced preclinical and clinical trial status. However, plant platforms present several drawbacks: time-consuming in generating stable transgenic lines, non homogeneous protein production in different tissues, impact of pests and diseases even in controlled conditions (greenhouses) and, more importantly, growth in non-sterile conditions. This last point may affect the good manufacturing practices (GMP) necessary for the production of pharmaceuticals. Transient expression or in vitro culture have emerged as alternative platforms to circumvent some of these drawbacks. Food and Drug Administration (FDA) has recently approved the first plant-made drug for human use, an enzyme produced in genetically engineered carrot cells for treating type 1 Gaucher’s disease. Plant production of candidate prophylactic and therapeutic HPV vaccines is proven, with evidence of efficacy in animals. There are data showing that an adjuvant-like effect was obtained in immunizations with crude tobacco plant extracts containing the E7 protein of HPV 16 [73, 74]. The recombinant plant-derived vaccines as ‘in planta formulation’ without adjuvants were able to elicit also a protective Th1 cell response in mice. Similar adjuvating activity was seen in another tobacco plant-produced fusion protein of the HPV 16 E7; this antigen preparation was able to induce a specific CD8+ T stimulation that elicited a therapeutic effect on experimental tumors [46, 47]. Finally, the possibility to produce E7 with high immunological activity in microalgae opens the way to producing antigens at affordable price, retaining the adjuvating activity of these plant-derived antigens [75]. An FDA-approved clinical trial for non-Hodgkin’s lymphoma with plant-produced single-chain variable fragment (scFv) was able to establish the safety and immunogenicity of plant made human vaccines [76, 77], thus indicating the feasibility of this approach for human anticancer therapies. However, to our knowledge, no clinical trial with plant derived anti-HPV vaccine has been carried out yet.

DNA/RNA based vaccines

DNA vaccines have been used in the clinical arena to elicit antigen-specific immune responses. Although nucleic acid vaccines do not appear to induce as vigorous immune responses as live viral vaccine vectors, they have several advantages, mainly naked DNA is relatively safe, stable, cost efficient, and able to sustain reasonable levels of antigen expression within cells. DNA-based plasmid vectors remain stable in a wide range of conditions over great lengths of time (longer periods than RNA vaccines), and they can be delivered with little risk to individuals who are immunosuppressed. In addition, since DNA vaccines do not elicit neutralizing antibodies in the vaccinated patient, they can be repeatedly administered with similar efficacy. Many strategies have been employed to produce an efficient delivery of targeted antigen-to-antigen presenting cells (APC) such as dendritic cells (DCs), an enhancement of antigen processing and presentation in DCs, and an augmentation of DC and T cell interaction [78]. Recently, it has been reported that the fusion of the E7 gene of HPV 16 with a plant virus coat protein produced strong antitumor activity in a mouse model activating both CD4+ and CD8+ T cells [46, 79], as well as a fusion of E7 gene to a gene encoding a mutated form of the immunotoxin from saponaria officinalis, the saporine [80]. The latter should enhance the activity of the vaccine by the immunomodulant activity of this mutated saporine that has lost its toxic activity. The possibility to utilize enhanced delivery methods like electroporation, microencapsulation, and gene gun has further enhanced the targeting of DNA vaccine to DCs.

A dose-escalation trial of plasmid DNA encoding a transgene that produced E7 linked to Hsp70 showed limited efficacy at the highest dose, with low induction of responses in the IFN-gamma ELISPOT assay and a resolution rate of 33% [81].

A plasmid DNA encoding a 13-amino acid sequence of E7 encapsulated in biodegradable poly (D,L-lactide-co-glycolide) micro-particles was utilized to develop the ZYC101 vaccine expressing a HPV 16 E7 HLA-A2 restricted peptide. Two different phase I clinical trials examining the potential treatment of patients with anal dysplasia or with high-grade CIN, respectively, demonstrated a high number of immunological response i.e. circulating HPV-specific T cells and histological regression/improvement in 1/3 of the patients [82, 83]. The improved version ZYC101a, that includes in addition to the HPV-encoding sequences of HPV 16 E7 the regions encoding segments of HPV 16 and HPV 18 E6 and E7 viral proteins, is one of the few therapeutic vaccines reaching the phase II/III in clinical trials involving subjects with high-grade CIN. In a prospectively defined population of women younger than 25 years CIN resolution was significantly higher in the ZYC101a groups compared to placebo [84]. Its activity in the treatment of patients with CIN 2/3 was also evaluated in a double-blinded, randomized, placebo-controlled clinical trial where half of 21 patients receiving the vaccine showed HPV 16/18-specific T cell responses but only 6 patients recovered from the high grade CIN [85].

VGX-3100, a DNA vaccine incorporating plasmids targeting HPV 16 and 18 E6 and E7 proteins was utilized in clinical trials employing electroporation technology. This technology was demonstrated particularly effective in animal models where the candidate vaccine is delivered via intramuscular injection followed by electroporation using various devices to deliver a small electrical charge. In a phase I clinical trial, 78% of the VGX-3100 vaccinated high-grade CIN subjects showed T cell and antibody responses [86]. On the bases of these findings, a double-blinded, randomized, placebo-controlled phase II clinical trial is ongoing on high grade CIN (NCT01304524). Further trials will be developed associating this vaccination with IL-12, used as an adjuvant, in order to significantly increase the CD4+ T cell response and with a vaccination against telomerase (hTERT), an antigen that is known to be related to a great number of human tumors and whose immunogenicity was demonstrated by the presence of existing naturally occurring T cell responses (Bagarazzi M. Personal Communication).

Other DNA vaccines have also been associated with other adjuvating treatments. In particular, the TLR7 agonist, Imiquimod promoting the activation of antigen-presenting cells and, in turn, leading to the production of cytokines IFN-alpha, IL-6, and TNF alpha [87] was shown to be active in mouse models [88]. In particular, the Imiquimod treatment affected the tumor microenvironment by reducing the number of myeloid-derived suppressor cells that have an immunosuppressive role and increasing natural killer (NK) and NKT cells that may play a role in tumor volume reduction. Thus this approach was utilized in an ongoing phase I clinical trial, which investigates a prime-boost strategy, combined with topical Imiquimod in treating patients with CIN3. The prime-boost strategy consisted of a DNA vaccine encoding an endoplasmic reticulum signal sequence (Sig), linked to an attenuated form of HPV 16 E7 fused to Hsp70 (pNGVL4a-Sig/E7(Detox)/Hsp70) boosted with a recombinant vaccinia virus encoding E6 and E7 of HPV 16 and 18 (TA-HPV) [NCT00788164].

Finally, the use of RNA replicons is a potentially interesting strategy for HPV vaccination. RNA replicons are naked RNA molecules derived from alpha-viruses, such as Sindbis virus [89, 90], Semliki Forest virus [9193], and Venezuelan equine encephalitis (VEE) [94] viurs. These RNA vaccines are self-replicating and self-limiting, and may be administered as either RNA or DNA, which is then transcribed into RNA replicons. RNA replicon-based vectors can replicate in a wide range of cell types and can be used to produce sustained levels of antigen expression in cells, making them more immunogenic than conventional DNA vaccines. However, RNA replicons are less stable than DNA. To combine the benefits of DNA and RNA replicon, DNA-launched RNA replicon, termed ‘suicidal’ DNA was utilized for HPV vaccine development in preclinical models [94, 95].

This ‘suicidal DNA’ is transcribed into RNA within the transfected cell and provides a stable and efficient way to express tumor antigen but the cells may undergo apoptosis. Another replicon system is derived from the flavivirus Kunjin (KUN) [96]. The new generation of KUN replicon vectors, which allows for the synthesis of replicon RNA from plasmid DNA did not induce cellular apoptosis and was able to elicit specific T cell responses protecting mice from tumor challenge [97]. However, despite the general success of RNA replicons in preclinical models, RNA replicon-based vaccines have had limited clinical testing.

The newest mRNA-based vaccines have also been developed by Novartis as a non-viral delivery system for self-amplifying RNA that has been successfully tested in preclinical models. Another mRNA-based vaccine is the RNActive® vaccine platform from CureVac (Tübingen, Germany) that is based on a more stable modified mRNA sequence with increased immunogenicity by complexation with protamine. This mRNA vaccine exploits both the antigenic and the adjuvant properties of mRNAs to activate the adaptive and innate immune system. Two ongoing clinical trials on patients with prostate and non-small cell lung cancer show that RNActive® vaccines are safe and effective in inducing long lasting, humoral and cellular immune responses. No information is available on the possible utilization on HPV-associated cancers.

Bacterial/viral vectors

Bacteria, such as Listeria monocytogenes (LM) [98, 99], Lactococcus lactis [100], Lactobacillus casei [101], Salmonella and Bacillus Calmette-Guerin, and several viral vectors, including vaccinia virus (VV), adenovirus, adeno-associated virus, alpha-virus, and its derivative vectors, have been used to deliver genes or proteins of interest to elicit antigen-specific immunotherapy. Among the bacterial vectors, LM has emerged as a promising vector, because in animal models it is able to induce both CD8+ and CD4+ immune responses, to elicit regression of established tumors, and to overcome central tolerance by expanding low avidity CD8+ T cells specific for E7 [98]. ADXS11-001 a live, attenuated LM bacterial vector secreting HPV 16 E7 fused to listeriolysin O (LLO) LLO was utilized in clinical trials. Promising results of this vaccine in phase I trials for safety and immunological responses [102, 103] were further assessed in phase II clinical trials. At least three trials are ongoing involving women with persistent or recurrent cervical carcinoma (NCT01266460), with CIN 2/3 with surgical indication (NCT01116245) [104], and patients (including male) with HPV-associated oropharyngeal cancer (NCT01598792).

Among viral vectors employed for the expression of HPV antigens, like adenoviruses [105, 106], alpha-viruses [107109] and VV [110112], the last virus was historically one of the first viral vectors employed in clinical trials on therapeutic vaccines against HPV-associated cancer [113]. More recently Avipox viruses have been developed as novel vectors for the development of vaccines. Although their replication is restricted to avian they are permissive for entry and transgenic expression in most mammalian cells, and immunologically non cross-reactive with vaccinia, which avoids pre-existing immunity in smallpox experienced humans. Avipox viruses might therefore represent safer immunogens that have demonstrated their activity in inhibiting the growth of HPV16 E7 expressing tumor in C57 Bl6 mice with a HPV16 E7 DNA-prime/Fowlpox HPV16 E7-boost schedule [114].

To date many VV vaccines have been employed in clinical trials to deliver genes and antigens of interest efficiently. Phase I/II clinical trials in patients with vulvar or vaginal and early or late -stage cervical cancer were conducted with a vaccinia vector encoding HPV 16 and HPV 18 E6 and E7 antigens (TA-HPV) recombinant VV [113, 115117]. In particular, in a phase II clinical trial, 29 patients with stage I or II cervical cancer were vaccinated twice via scarification with TA-HPV; although clinical outcomes were not measured due to surgical intervention in all patients, induction of CTL responses were detected in a number of patients in the form of target cell lysis by isolated peripheral bone marrow cells (PBMCs) [117].

In another study, a recombinant VV expressing E6 and E7 antigen together with IL-2 (TG4001/R3484) was administered to CIN 2/3 patients, with very promising clinical results. Ten patients (48%) were evaluated as clinical responders at month 6. At month 12, 7 out of 8 patients without conization reported neither suspicion of CIN 2/3 relapse nor HPV 16 infection [118]. Interim results phase IIb trial on patients with HPV-related CIN 2/3 lesions demonstrated the activity of vaccine in monotherapy, but the trial did not reach its primary endpoint of six-month resolution of CIN 2/3 and will not move on to a phase III trial [119]. A recombinant modified vaccinia Ankara vector was also utilized to express papillomavirus protein other than E6 or E7, namely the bovine papillomavirus E2 (MVA-E2). E2 is a transcriptional repressor of E6 and E7 oncogenes, it was assumed that in low grade lesions, where the virus is not integrated, the MVA-E2 expressed protein could bind to the repressor region of the viral genome. However, there is no evidence for E2 expression direct contribution to the therapeutic effect seen in both patients with CIN [120, 121] and genital warts [122]. The vaccine could be active such as a non-specific “pro-inflammatory stimulation” in the cervico-vaginal tract that induces an immune response.

Finally, in alternative to viral vectors, synthetic viral vector like virus like particle (VLP) can be utilized because they are easy to manufacture and have the capacity for compacting DNA, and targeting specific cell receptors. Thus, the same technology used for producing anti-HPV prophylactic vaccines was employed for producing chimeric VLPs. An L1–E7 fusion protein has been shown to self-assemble into chimeric VLPs (CVLP) that can induce E7-specific cellular immunity in mice [123]. A randomized, double blind, placebo-controlled clinical trial has been conducted in CIN 2/3 patients with CVLP. Antibodies with high titers against HPV 16 L1 and low titers against HPV 16 E7 as well as cellular immune responses against both proteins were induced. Although not statistically significant a trend for histological improvement to CIN I or normal histology was seen in 39% of the patients [124].

Dendritic cells (DCs) based vaccines

The immune response to initial stages of infection causes inflammatory responses that trigger innate effector cells, such as NK and NKT cells. This inflammatory response, driving the innate immunity, is initiated through pathogen-associated molecular pattern (PAMP) sensors including TLRs 1–9. These receptors in response to specific bacterial or viral components activate APCs via the transcription factor nuclear factor KB (NF-KB). In addition, infection may alter the local metabolic and cellular microenvironment activating danger-associated molecular pattern (DAMP) sensors, particularly nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs), as components of multiprotein complexes termed the inflammasomes, inducing maturation and releasing members of the IL-1 family. In particular, the produced IL-1b and IL-18 mediate repair responses such as angiogenesis and, via upregulation of cytokines and chemokines, induce the recruitment of inflammatory cells to the site of infection. The observed slow clearance of HPV infection and weak immune responses to viral proteins is a likely consequence of the nonlytic nature of HPV infection and a consequent delay in induction of PAMP- and DAMP-induced inflammatory responses through TLRs and the inflammasomes. Thus, in the absence of inflammation a number of events take place (i.e. IL-10 production by Th cells and mast cells; IFN-gamma production by CD-1d-activated NKT cells, increased TGF-beta) inducing negative regulatory signals that in turn can change the state of the APC by altering costimulatory molecule expression, thus inhibiting induction of cytotoxic effector T cells. Therefore a therapy aimed to reactivate these APCs could be a valid tool for clinical intervention.

Among specialized APCs the most potent are DCs, because they express high levels of MHC and co-stimulatory molecules. Therefore, DCs were the research focus of many investigators and a variety of methods for generating DCs, loading them with tumor antigens, and administering them to patients. The success of Provenge, a DC vaccine incorporating prostatic acid phosphatase, in patients with advanced prostate cancer has generated great interest for DC-based vaccines [125, 126]. Although expensive, DC-based vaccines have been tested in patients with HPV-associated cervical cancer by successfully transducing genes coding for E6 and E7 into DCs. In a clinical study, autologous DCs were pulsed with HPV 16 or HPV 18 E7 recombinant proteins and E7-specific CD8+ T cell responses were observed in 4 out of 11 late stage cervical cancer patients [127]. In another clinical study, stage IB or IIA cervical cancer patients were vaccinated with autologous DC pulsed with recombinant HPV 16/18 E7 antigens and keyhole limpet hemocyanin 1 (KLH), an immunological carrier protein. This vaccine generated E7-specific T cell responses in 8 out of 10 patients and antibody responses in all patients [128]. Another ongoing clinical trial is being conducted in Taiwan National University in recurrent cervical cancer (NCT00155766), using DCs pulsed with HPV 16 E7 antigen. However, DC-based vaccines could be used to treat advanced cervical cancers but are unlikely to be used to treat CIN lesions because the procedures involved in this kind of treatment are expensive and labor-intensive.

Combinational immunotherapy

Given the importance of local microenvironment in the persistence of HPV lesions, strategies aiming to alter local immunity have shown some positive results, therefore therapeutic HPV vaccine strategies have shifted toward combinatorial approaches with radiotherapy and chemotherapy. Low-dose radiation in combination with HPV vaccination was effective in the treatment of tumors in preclinical models [129]. Radiation therapy seems to be a useful method in stabilizing tumor cell growth when applied with immunotherapy by inducing apoptosis in tumor cells.

A chemotherapeutic agent in combination with DNA-based vaccines was proven an effective HPV therapy in preclinical models [130136]. Low cyclophosphamide doses, altering local immunity, exerted some positive effects in persistent low-risk HPV lesions [135]. A randomized trial comparing chemotherapy versus a combination with the HPV 16 Synthetic Long Peptide (SLP) in advanced cervical cancer is planned. A pilot study is ongoing with the aim to investigate the optimal time (“window”) for the vaccination with the HPV 16 SLP vaccine after standard treatment of 6 cycles Carboplatin-Paclitaxel every three weeks (EudraCT2010-018841-76).

A randomized study was carried out in 110 recurrent/refractory cervical cancer patients with cisplatin and different doses of HPV bacterial vector-based vaccine ADXS11-001, and preliminary results showed efficacy and manageable toxicity [137].

In addition, other compounds affecting the immunological environment like COX-2 inhibitors, through the prevention of the production of prostaglandin E2 or antibodies to IL-6 [138] or IL-10 [139] or the TLR agonist Imiquimod could be effective.

In particular, Imiquimod is already in clinical use against warts stimulating local innate immunity and potentiating adaptive immune response by activating tissue antigen presenting cells. A number of studies with topical Imiquimod have been reported with favorable results mainly in vulvar intraepithelial neoplasia (VIN) lesions [63, 140] and combination therapy with HPV DNA vaccines described in the previous paragraphs of this review.

Cytokines-based therapies in combination with HPV therapeutic vaccine showed promising results in preclinical models. Treatment with IL-12 gene, administered as gene therapy, viral gene therapy, by adenovirus, and in combination with E6-E7 oncogenes, determined tumor growth suppression [141, 142].

Recently, programmed death-1 receptor (PD-1) is emerging as a pivotal target for combination therapy by affecting immune suppression.

Indeed, PD-1 is expressed on T cells following T-cell receptor (TCR) activation and binding of this receptor to its cognate ligands, programmed death ligand (PDL)-1 and PDL-2, down-regulates TCR signals, promoting T-cell anergy and apoptosis, thus leading to immune suppression.

An anti-PD-1 antibody (CT-011) with Treg-cell depletion by low-dose cyclophosphamide (CPM), combined with HPV 16 E7 peptide vaccine, produced synergistic antigen-specific immune responses inducing complete regression of established tumors in a significant percentage of treated animals, with prolonging survival [143].

Expanded phase I clinical studies with anti-PD-1 (BMS-936558) and anti-PDL -1 (BMS-936559) showed objective clinical responses in renal cell carcinoma, melanoma, and non–small cell lung cancer, and a relationship between tumor cell surface PD-L1 expression and objective responses to anti-PD1 therapy [144, 145]. In addition, a recent study showed that PD-1: PDL-1 pathway may create an “immune-privileged” site for initial viral infection in the tonsils and subsequent adaptive immune resistance once tumors are established suggesting a rationale for therapeutic blockade of this pathway in patients with HPV + oropharyngeal squamous cell carcinoma [146]. Other strategies trying to inhibit the suppressive tumor microenvironment utilize monoclonal antibodies such as Ipilimumab. This antibody is a fully human monoclonal antibody against the cytotoxic T-lymphocyte antigen-4 (CTLA-4), an immune-inhibitory molecule expressed in activated T cells and in suppressor T regulatory cells. The interaction between the monoclonal antibody and CTLA-4 blocks inhibitory signals and enhances T cell activation, leading to increased antitumor responses [147]. Ipilimumab has been approved for melanoma, but ongoing trials are testing the drug in other tumors, among them locally advanced cervical cancer, in a sequential regimen following chemoradiation (NCT01711515).

Conclusions

The development of cervical cancer depends not only on efficient negative regulation of cell-cycle control supporting the accumulation of genetic damage, but also on a sophisticated viral mechanism of immune evasion [2022]. Despite this complex interplay, most HPV infections are cleared within one year, and cell-mediated immunity plays an important role in this process. Thus, a better understanding of immunological path involved in cervical cancer carcinogenesis is an important goal in cervical cancer knowledge, and the development of new treatment modalities involving immune system may represent an innovative strategy in this complex disease. Although antigen-specific immunotherapy induced by therapeutic HPV vaccines was proved extremely efficacious in preclinical models, its progression through clinical trials suffered of the early poor responses. Such initial clinical trials were conducted in advanced cancer patients to evaluate the safety of the respective vaccine. Because of the immunosuppressive milieu generated by established tumors, it appears that (likewise to cancer immunotherapy in general) in cases of advanced disease, an immunological antitumor effect will, if at all, only be successful along with standard therapy. Later attempts took advantage of the unique situation in HPV-related carcinogenesis, that is, the existence of well-defined precursors to cervical, anal or vulvar cancer.

Several clinical studies on these precursors lesions have so far been completed with promising effects, even if these trials were not powered to detect small effects in the vaccine versus the placebo groups. Indeed, there was no follow-up study with the same strategy, perhaps as a result that these trials were investigator-initiated or sponsored by small biotech companies.

Large pharmaceutical corporations that could have ensured continuation of the individual program and guarantee the prosecution in more powered phase II/III trials were completely lacking. Even the partnership announced in 2007 between a large pharmaceutical firm (Roche) and a biotech company (Transgene) on a promising vaccine TG4001 was recently discontinued.

In conclusion immune escape and immune suppression are active in cancer and to some extent also in premalignant lesions and, therefore one might consider targeting persistent infections before they become clinically apparent. This ‘paradigm shift’ in vaccination of cancer patients is a quite logical step in HPV-related cervical cancer. Treating this early condition with post-exposure prophylaxis might be more successful than therapy at more advanced stages. Several well-defined cohorts of women with persistent infections do exist, and the design and execution of clinical trials aiming to clear of HPV DNA should be straightforward. However, it is important to take into consideration that patients with HSIL are generally asymptomatic and can be treated by minor surgical procedures. A clinical trial on long overlapping peptide vaccine in CIN 2/3 patients was stopped prematurely because motivational problems of the patients and the local/systemic side effects of the vaccine. Thus, motivational problems must be taken into account when considering studies in patients with premalignant lesions for whom an effective treatment is available [72]. Future developments should consider the disparities of side effects between standard of care and new therapies to maximize the potential benefits of therapeutic vaccination. In addition, therapeutic efficacy would receive beneficial effects by addressing the immunosuppressive tumor microenvironment. Combinational strategies particularly for cancer patients are a promising tool to control tumor metastasis and eliminate cancer cells altogether.

The development of prophylactic and therapeutic HPV vaccines must be continued in order to come close to the eradication of HPV-associated malignancies even in other localizations [148, 149], getting rid of the perception (after the effectiveness of commercial preventive vaccines) that the problem has been solved.

References

  1. Ferlay J, Soerjomataram I, Ervik M, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F: GLOBOCAN 2012 v1.0, Cancer Incidence and Mortality Worldwide. IARC CancerBase No. 11 [Internet]. 2013, Lyon, France: International Agency for Research on Cancer, Available from: http://globocan.iarc.fr, accessed on day/month/year

    Google Scholar 

  2. Li N, Franceschi S, Howell-Jones R, Snijders PJF, Clifford GM: Human papillomavirus type distribution in 30,848 invasive cervical cancers worldwide: variation by geographical region, histological type and year of publication. Int. J Cancer. 2010, 128: 927-935.

    Article  CAS  Google Scholar 

  3. Bosch FX, De Sanjosé S: Chapter 1: Human papillomavirus and cervical cancer–burden and assessment of causality. J Natl Cancer Inst Monogr. 2003, 31: 3-13.

    Article  PubMed  Google Scholar 

  4. Trottier H, Franco EL: The epidemiology of genital human papillomavirus infection. Vaccine. 2006, 24 (Suppl 1): S1-S15.

    PubMed  Google Scholar 

  5. Gravitt PE: The known unknowns of HPV natural history. J Clin Invest. 2011, 121: 4593-4599. 10.1172/JCI57149.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  6. Ellerbrock TV, Chiasson MA, Bush TJ, Sun XW, Sawo D, Brudney K, Wright TC: Incidence of cervical squamous intraepithelial lesions in HIV-infected women. JAMA. 2000, 283: 1031-1037. 10.1001/jama.283.8.1031.

    Article  CAS  PubMed  Google Scholar 

  7. Ognenovski VM, Marder W, Somers EC, Johnston CM, Farrehi JG, Selvaggi SM, McCune WJ: Increased incidence of cervical intraepithelial neoplasia in women with systemic lupus erythematosus treated with intravenous cyclophosphamide. J Rheumatol. 2004, 31: 1763-1767.

    CAS  PubMed  Google Scholar 

  8. Nakagawa M, Gupta SK, Coleman HN, Sellers MA, Banken JA, Greenfield WW: A favorable clinical trend is associated with CD8 T-cell immune responses to the human papillomavirus type 16 e6 antigens in women being studied for abnormal pap smear results. J Low Genit Tract Dis. 2010, 14: 124-129. 10.1097/LGT.0b013e3181c6f01e.

    Article  PubMed  Google Scholar 

  9. Wang SS, Schiffman M, Herrero R, Carreon J, Hildesheim A, Rodriguez AC, Bratti MC, Sherman ME, Morales J, Guillen D, Alfaro M, Clayman B, Burk RD, Viscidi RP: Determinants of human papillomavirus 16 serological conversion and persistence in a population-based cohort of 10 000 women in Costa Rica. Br J Cancer. 2004, 91: 1269-1274. 10.1038/sj.bjc.6602088.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  10. Carter JJ, Madeleine MM, Shera K, Schwartz SM, Cushing-Haugen KL, Wipf GC, Porter P, Daling JR, McDougall JK, Galloway DA: Human papillomavirus 16 and 18 L1 serology compared across anogenital cancer sites. Cancer Res. 2001, 61: 1934-1940.

    CAS  PubMed  Google Scholar 

  11. Stern PL, van der Burg SH, Hampson IN, Broker TR, Fiander A, Lacey CJ, Kitchener HC, Einstein MH: Therapy of human papillomavirus-related disease. Vaccine. 2012, 30 (Suppl 5): F71-F82.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  12. De Jong A, Van Poelgeest MI, van der Hulst JM, Drijfhout JW, Fleuren GJ, Melief CJ, Kenter G, Offringa R, van der Burg SH: Human papillomavirus type 16-positive cervical cancer is associated with impaired CD4+ T-cell immunity against early antigens E2 and E6. Cancer Res. 2004, 64: 5449-5455. 10.1158/0008-5472.CAN-04-0831.

    Article  CAS  PubMed  Google Scholar 

  13. Kim KH, Greenfield WW, Cannon MJ, Coleman HN, Spencer HJ, Nakagawa M: CD4+ T-cell response against human papillomavirus type 16 E6 protein is associated with a favorable clinical trend. Cancer Immunol Immunother. 2012, 61: 63-70. 10.1007/s00262-011-1092-5.

    Article  PubMed Central  PubMed  Google Scholar 

  14. Farhat S, Nakagawa M, Moscicki AB: Cell-mediated immune responses to human papillomavirus 16 E6 and E7 antigens as measured by interferon gamma enzyme-linked immunospot in women with cleared or persistent human papillomavirus infection. Int J Gynecol Cancer. 2009, 19: 508-512. 10.1111/IGC.0b013e3181a388c4.

    Article  PubMed Central  PubMed  Google Scholar 

  15. Heusinkveld M, Welters MJ, Van Poelgeest MI, van der Hulst JM, Melief CJ, Fleuren GJ, Kenter GG, van der Burg SH: The detection of circulating human papillomavirus-specific T cells is associated with improved survival of patients with deeply infiltrating tumors. Int J Cancer. 2011, 128: 379-389. 10.1002/ijc.25361.

    Article  CAS  PubMed  Google Scholar 

  16. Venuti A, Paolini F, Nasir L, Corteggio A, Roperto S, Campo MS, Borzacchiello G: Papillomavirus E5: the smallest oncoprotein with many functions. Mol Cancer. 2011, 10: 140-10.1186/1476-4598-10-140. Review

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  17. Ashrafi GH, Haghshenas MR, Marchetti B, O’Brien PM, Campo MS: The E5 protein of human papillomavirus type 16 selectively down-regulates surface HLA class. Int J Cancer. 2005, 113: 276-283. 10.1002/ijc.20558.

    Article  CAS  PubMed  Google Scholar 

  18. Zhang B, Li P, Wang E, Brahmi Z, Dunn KW, Blum JS, Roman A: The E5 protein of human papillomavirus type 16 perturbs MHC class II antigen maturation in human foreskin keratinocytes treated with interferon-γ. Virology. 2003, 310: 100-108. 10.1016/S0042-6822(03)00103-X.

    Article  CAS  PubMed  Google Scholar 

  19. Campo MS, Graham SV, Cortese MS, Ashrafi GH, Araibi EH, Dornan ES, Miners K, Nunes C, Man S: HPV-16 E5 down-regulates expression of surface HLA class I and reduces recognition by CD8 T cells. Virology. 2010, 407: 137-142. 10.1016/j.virol.2010.07.044.

    Article  CAS  PubMed  Google Scholar 

  20. Stanley MA, Pett MR, Coleman N: HPV: from infection to cancer. Biochem Soc Trans. 2007, 35: 1456-1460. 10.1042/BST0351456.

    Article  CAS  PubMed  Google Scholar 

  21. O’Brien PM, Campo MS: Evasion of host immunity directed by papillomavirus encoded proteins. Virus Res. 2002, 1: 103-118.

    Article  Google Scholar 

  22. Crosbie EJ, Einstein MH, Franceschi S, Kitchener HC: Human papillomavirus and cervical cancer. Lancet. 2013, 382: 889-899. 10.1016/S0140-6736(13)60022-7.

    Article  PubMed  Google Scholar 

  23. Scott ME, Ma Y, Kuzmich L, Moscicki AB: Diminished IFN-gamma and IL-10 and elevated Foxp3 mRNA expression in the cervix are associated with CIN 2 or 3. Int J Cancer. 2009, 124: 1379-1383. 10.1002/ijc.24117.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  24. Gooden M, Lampen M, Jordanova ES, Leffers N, Trimbos JB, van der Burg SH, Nijman H, Van Hall T: HLA-E expression by gynecological cancers restrains tumor-infiltrating CD8+ T lymphocytes. Proc Natl Acad Sci USA. 2011, 108: 10656-10661. 10.1073/pnas.1100354108.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  25. Piersma SJ: Immunosuppressive tumor microenvironment in cervical cancer patients. Cancer Microenviron. 2011, 4: 361-375. 10.1007/s12307-011-0066-7.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  26. O’Hagan DT, Rappuoli R: Novel approaches to vaccine delivery. Pharm Res. 2004, 21: 1519-1530.

    Article  PubMed  Google Scholar 

  27. Koutsky LA, Ault KA, Wheeler CM, Brown DR, Barr E, Alvarez FB, Chiacchierini LM, Jansen KU, Proof of Principle Study Investigators: A controlled trial of a human papillomavirus type 16 vaccine. N Engl J Med. 2002, 347: 1645-1651. 10.1056/NEJMoa020586.

    Article  CAS  PubMed  Google Scholar 

  28. Harper DM, Franco EL, Wheeler C, Ferris DG, Jenkins D, Schuind A, Zahaf T, Innis B, Naud P, De Carvalho NS, Roteli-Martins CM, Teixeira J, Blatter MM, Korn AP, Quint W: Dubin G; GlaxoSmithKline HPV Vaccine Study Group: Efficacy of a bivalent L1 virus-like particle vaccine in prevention of infection with human papillomavirus types 16 and 18 in young women: a randomised controlled trial. Lancet. 2004, 364: 1757-1765. 10.1016/S0140-6736(04)17398-4.

    Article  CAS  PubMed  Google Scholar 

  29. Lowy DR, Schiller JT: Reducing HPV-associated cancer globally. Cancer Prev Res. 2012, 5: 18-23. 10.1158/1940-6207.CAPR-11-0542.

    Article  Google Scholar 

  30. Harper DM, Williams KB: Prophylactic HPV vaccines: current knowledge of impact on gynecologic premalignancies. Discov Med. 2010, 10: 7-17.

    PubMed  Google Scholar 

  31. Campo MS, Roden RB: Papillomavirus prophylactic vaccines: established successes, new approaches. J Virol. 2010, 84: 1214-1220. 10.1128/JVI.01927-09.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  32. Vici P, Mariani L, Pizzuti L, Sergi D, Di Lauro L, Vizza E, Tomao F, Tomao S, Mancini E, Vincenzoni C, Barba M, Maugeri-Saccà M, Giovinazzo G, Venuti A: Emerging Biological Treatments for Uterine Cervical Carcinoma. J Cancer. 2014, 5: 86-97. 10.7150/jca.7963.

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  33. Wright TC, Cox JT, Massad LS: Consensus guidelines for the management of women with cervical cytological abnormalities. JAMA. 2001, 2002 (287): 2120-2129.

    Google Scholar 

  34. Ma B, Maraj B, Tran NP, Knoff J, Chen A, Alvarez RD, Hung CF, Wu TC: Emerging human papillomavirus vaccines. Expert Opin Emerg Drugs. 2012, 17: 469-492. 10.1517/14728214.2012.744393.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  35. Hall AH, Alexander KA: RNA interference of human papillomavirus type 18 E6 and E7 induces senescence in HeLa cells. J Virol. 2003, 77: 6066-6069. 10.1128/JVI.77.10.6066-6069.2003.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  36. Qi Z, Xu X, Zhang B, Li Y, Liu J, Chen S, Chen G, Huo X: Effect of simultaneous silencing of HPV-18 E6 and E7 on inducing apoptosis in HeLa cells. Biochem Cell Biol. 2010, 88: 697-704. 10.1139/O10-005.

    Article  CAS  PubMed  Google Scholar 

  37. Griffin H, Elston R, Jackson D, Ansell K, Coleman M, Winter G, Doorbar J: Inhibition of papillomavirus protein function in cervical cancer cells by intrabody targeting. J Mol Biol. 2006, 355: 360-378. 10.1016/j.jmb.2005.10.077.

    Article  CAS  PubMed  Google Scholar 

  38. Accardi L, Donà MG, Di Bonito P, Giorgi C: Intracellular anti-E7 human antibodies in single-chain format inhibit proliferation of HPV16-positive cervical carcinoma cells. Int J Cancer. 2005, 116: 564-570. 10.1002/ijc.21052.

    Article  CAS  PubMed  Google Scholar 

  39. Accardi L, Paolini F, Mandarino A, Percario Z, Bonito PD, Carlo VD, Affabris E, Giorgi C, Amici C, Venuti A: In vivo antitumor effect of an intracellular single-chain antibody fragment against the E7 oncoprotein of Human Papillomavirus 16. Int J Cancer. 2013, in press

    Google Scholar 

  40. Richter CE, Cocco E, Bellone S, Bellone M, Casagrande F, Todeschini P, Rüttinger D, Silasi DA, Azodi M, Schwartz PE, Rutherford TJ, Pecorelli S, Santin AD: Primary cervical carcinoma cell lines overexpress epithelial cell adhesion molecule (EpCAM) and are highly sensitive to immunotherapy with MT201, a fully human monoclonal anti-EpCAM antibody. Int J Gynecol Cancer. 2010, 20: 1440-1447.

    PubMed Central  PubMed  Google Scholar 

  41. Badaracco G, Venuti A: Human papillomavirus therapeutic vaccines in head and neck tumors. Expert Rev Anticancer Ther. 2007, 7: 753-766. 10.1586/14737140.7.5.753.

    Article  PubMed  Google Scholar 

  42. Venuti A: Progress and challenges in the vaccine-based treatment of head and neck cancers. J Exp Clin Cancer Res. 2009, 28: 69-10.1186/1756-9966-28-69.

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  43. Stewart TJ, Drane D, Malliaros J: ISCOMATRIX adjuvant: an adjuvant suitable for use in anticancer vaccines. Vaccine. 2004, 22: 3738-3743. 10.1016/j.vaccine.2004.03.026.

    Article  CAS  PubMed  Google Scholar 

  44. Cui Z, Huang L: Liposome-polycation-DNA (LPD) particle as a carrier and adjuvant for proteinbased vaccines: therapeutic effect against cervical cancer. Cancer Immunol Immunother. 2005, 54: 1180-1190. 10.1007/s00262-005-0685-2.

    Article  CAS  PubMed  Google Scholar 

  45. Kang TH, Monie A, Wu LS: Enhancement of protein vaccine potency by in vivo electroporation mediated intramuscular injection. Vaccine. 2011, 29: 1082-1089. 10.1016/j.vaccine.2010.11.063.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  46. Venuti A, Massa S, Mett V, Vedova LD, Paolini F, Franconi R, Yusibov V: An E7-based therapeutic vaccine protects mice against HPV16 associated cancer. Vaccine. 2009, 27: 3395-3397. 10.1016/j.vaccine.2009.01.068.

    Article  CAS  PubMed  Google Scholar 

  47. Massa S, Franconi R, Brandi R, Muller A, Mett V, Yusibov V, Venuti A: Anti-cancer activity of plant-produced HPV16 E7 vaccine. Vaccine. 2007, 25: 3018-3021. 10.1016/j.vaccine.2007.01.018.

    Article  CAS  PubMed  Google Scholar 

  48. Chu NR, Wu HB, Wu T, Boux LJ, Siegel MI, Mizzen LA: Immunotherapy of a human papillomavirus (HPV) type 16 E7-expressing tumour by administration of fusion protein comprising Mycobacterium bovis bacilli Calmette-Guerin (BCG) hsp65 and HPV16 E7. Clin Exp Immunol. 2000, 121: 216-225. 10.1046/j.1365-2249.2000.01293.x.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  49. Liu H, Wu BH, Rowse GJ, Emtage PC: Induction of CD4-independent E7-specific CD8+ memory response by heat shock fusion protein. Clin Vaccine Immunol. 2007, 14: 1013-1023. 10.1128/CVI.00029-07.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  50. Liao CW, Chen CA, Lee CN: Fusion protein vaccine by domains of bacterial exotoxin linked with a tumor antigen generates potent immunologic responses and antitumor effects. Cancer Res. 2005, 65: 9089-9098. 10.1158/0008-5472.CAN-05-0958.

    Article  CAS  PubMed  Google Scholar 

  51. 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. 2005, 65: 641-649.

    CAS  PubMed  Google Scholar 

  52. Granadillo M, Vallespi MG, Batte A, Mendoza O, Soria Y, Lugo VM, Torrens I: A novel fusion protein-based vaccine comprising a cell penetrating and immunostimulatory peptide linked to human papillomavirus (HPV) type 16 E7 antigen generates potent immunologic and anti-tumor responses in mice. Vaccine. 2011, 290: 920-930.

    Article  CAS  Google Scholar 

  53. Zwaveling S, Ferreira Mota SC, Nouta J: Established human papillomavirus type 16-expressing tumors are effectively eradicated following vaccination with long peptides. J Immunol. 2002, 169: 350-358.

    Article  CAS  PubMed  Google Scholar 

  54. Zhang YQ, Tsai YC, Monie A, Hung CF, Wu TC: Carrageenan as an adjuvant to enhance peptide-based vaccine potency. Vaccine. 2010, 28: 5212-5219. 10.1016/j.vaccine.2010.05.068.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  55. Wu CY, Yang HY, Monie A: Intraperitoneal administration of poly (I:C) with polyethylenimine leads to significant antitumor immunity against murine ovarian tumors. Cancer Immunol Immunother. 2011, 60: 1085-1096. 10.1007/s00262-011-1013-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Daftarian P, Mansour M, Benoit AC: Eradication of established HPV 16-expressing tumors by a single administration of a vaccine composed of a liposome-encapsulated CTL-T helper fusion peptide in a water-in-oil emulsion. Vaccine. 2006, 24: 5235-5244. 10.1016/j.vaccine.2006.03.079.

    Article  CAS  PubMed  Google Scholar 

  57. Barrios K, Celis E: TriVax-HPV: An improved peptide-based therapeutic vaccination strategy against human papillomavirus-induced cancers. Cancer Immunol Immunother. 2012, 61: 1307-1317. 10.1007/s00262-012-1259-8.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  58. Goldstone SE, Palefsky JM, Winnett MT: Activity of HspE7, a novel immunotherapy, in patients with anogenital warts. Dis Colon Rectum. 2002, 45: 502-507. 10.1007/s10350-004-6229-6.

    Article  PubMed  Google Scholar 

  59. Derkay CS, Smith RJ, McClay J: HspE7 treatment of pediatric recurrent respiratory papillomatosis: final results of an open-label trial. Ann Otol Rhinol Laryngol. 2005, 114: 730-737.

    Article  PubMed  Google Scholar 

  60. Roman LD, Wilczynski S, Muderspach LI: A phase II study of Hsp-7 (SGN-00101) in women with high-grade cervical intraepithelial neoplasia. Gynecol Oncol. 2007, 106: 558-566. 10.1016/j.ygyno.2007.05.038.

    Article  CAS  PubMed  Google Scholar 

  61. Einstein MH, Kadish AS, Burk RD: Heat shock fusion protein-based immunotherapy for treatment of cervical intraepithelial neoplasia III. Gynecol Oncol. 2007, 106: 453-460. 10.1016/j.ygyno.2007.04.038.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  62. Van Doorslaer K, Reimers LL, Studentsov YY, Einstein MH, Burk RD: Serological response to an HPV16 E7 based therapeutic vaccine in women with high-grade cervical dysplasia. Gynecol Oncol. 2010, 116: 208-212. 10.1016/j.ygyno.2009.05.044.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  63. Daayana S, Elkord E, Winters U: Phase II trial of imiquimod and HPV therapeutic vaccination in patients with vulval intraepithelial neoplasia. Br J Cancer. 2010, 102: 1129-1236. 10.1038/sj.bjc.6605611.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  64. Genticel: Genticel reaches an important milestone by launching its phase II trial in women infected with high - risk HPV before the appearance of high grade cervical lesions. 2014, Available from http://www.genticel.com/

    Google Scholar 

  65. Steller MA, Gurski KJ, Murakami M: Cell-mediated immunological responses in cervical and vaginal cancer patients immunized with a lipidated epitope of human papillomavirus type 16 E7. Clin Cancer Res. 1998, 4: 2103-2109.

    CAS  PubMed  Google Scholar 

  66. Van Driel WJ, Ressing ME, Kenter GG: Vaccination with HPV16 peptides of patients with advanced cervical carcinoma: clinical evaluation of a phase I-II trial. Eur J Cancer. 1999, 35: 946-952. 10.1016/S0959-8049(99)00048-9.

    Article  CAS  PubMed  Google Scholar 

  67. Muderspach L, Wilczynski S, Roman L, Bade L, Felix J, Small LA, Kast WM, Fascio G, Marty V, Weber J: 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. 2000, 6: 3406-3416.

    CAS  PubMed  Google Scholar 

  68. Welters MJ, Kenter GG, Piersma SJ: Induction of tumor-specific CD4+ andeCD8+ T-cell immunity in cervical cancerpatients by a human papillomavirustype 16 E6 and E7 long peptides vaccine. Clin. Cancer Res. 2008, 14: 178-187. 10.1158/1078-0432.CCR-07-1880.

    Article  CAS  PubMed  Google Scholar 

  69. Kenter GG, Welters MJ, Valentijn AR: Phase I immunotherapeutic trial with longpeptides spanning the E6 and E7 sequencesof high-risk human papillomavirus 16 inend-stage cervical cancer patients shows low toxicity and robust immunogenicity. Clin Cancer Res. 2008, 14: 169-177. 10.1158/1078-0432.CCR-07-1881.

    Article  CAS  PubMed  Google Scholar 

  70. Kenter GG, Welters MJ, Valentijn AR: Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med. 2009, 361: 1838-1847. 10.1056/NEJMoa0810097.

    Article  CAS  PubMed  Google Scholar 

  71. Welters MJ, Kenter GG, De Vos Van Steenwijk PJ: Success or failure of vaccination for HPV16-positive vulvar lesions correlates with kinetics and phenotype of induced T-cell responses. Proc Natl Acad Sci USA. 2010, 107: 11895-11899. 10.1073/pnas.1006500107.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  72. De Vos Van Steenwijk PJ, Ramwadhdoebe TH, Lowik MJ: A placebo-controlled randomized HPV16 synthetic long-peptide vaccination study in women with high-grade cervical squamous intraepithelial lesions. Cancer Immunol Immunother. 2012, 61: 1485-1492. 10.1007/s00262-012-1292-7.

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  73. Franconi R, Massa S, Illiano E, Mullar A, Cirilli A, Accardi L, Di Bonito P, Giorgi C, Venuti A: Exploiting the plant secretory pathway to improve the anticancer activity of a plant-derived HPV16 E7 vaccine. Int J Immunopathol Pharmacol. 2006, 19: 187-197.

    CAS  PubMed  Google Scholar 

  74. Franconi R, Di Bonito P, Dibello F, Accardi L, Muller A, Cirilli A, Simeone P, Donà MG, Venuti A, Giorgi C: Plant-derived human papillomavirus 16 E7 oncoprotein induces immune response and specific tumor protection. Cancer Res. 2002, 62: 3654-3658.

    CAS  PubMed  Google Scholar 

  75. Demurtas OC, Massa S, Ferrante P, Venuti A, Franconi R, Giuliano G: A Chlamydomonas-derived Human Papillomavirus 16 E7 vaccine induces specific tumorprotection. PLoS One. 2013, 8: e61473-10.1371/journal.pone.0061473.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  76. Franconi R, Demurtas OC, Massa S: Plant-derived vaccines and other therapeutics produced in contained systems. Expert Rev Vaccines. 2010, 9: 877-892. 10.1586/erv.10.91.

    Article  CAS  PubMed  Google Scholar 

  77. McCormick AA: Tobacco derived cancer vaccines for non-Hodgkin’s lymphoma: perspectives and progress. Hum Vaccin. 2011, 7: 305-312. 10.4161/hv.7.3.14163.

    Article  CAS  PubMed  Google Scholar 

  78. Lin K, Roosinovich E, Ma B, Hung CF, Wu TC: Therapeutic HPV DNA vaccines. Immunol Res. 2010, 47: 86-112. 10.1007/s12026-009-8141-6.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  79. Massa S, Simeone P, Muller A, Benvenuto E, Venuti A, Franconi R: Antitumor activity of DNA vaccines based on the human papillomavirus-16 E7 protein genetically fused to a plant virus coat protein. Hum Gene Ther. 2008, 19: 354-364. 10.1089/hum.2007.122.

    Article  CAS  PubMed  Google Scholar 

  80. Massa S, Paolini F, Spanò L, Franconi R, Venuti A: Mutants of plant genes for developing cancer vaccines. Hum Vaccin. 2011, 7 (Suppl147): 55-

    Google Scholar 

  81. Trimble CL, Peng S, Kos F, Gravitt P, Viscidi R, Sugar E, Pardoll D, Wu TC: A Phase I trial of a human papillomavirus DNA vaccine for HPV16+ cervical intraepithelial neoplasia 2/3. Clin Cancer Res. 2009, 15: 361-367. 10.1158/1078-0432.CCR-08-1725.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  82. Klencke B, Matijevic M, Urban RG, Lathey JL, Hedley ML, Berry M, Thatcher J, Weinberg V, Wilson J, Darragh T, Jay N, Da Costa M, Palefsky JM: Encapsulated plasmid DNA treatment for human papillomavirus 16-associated anal dysplasia: a phase I study of ZYC101. Clin Cancer Res. 2002, 8: 1028-1037.

    CAS  PubMed  Google Scholar 

  83. Sheets EE, Urban RG, Crum CP, Hedley ML, Politch JA, Gold MA, Muderspach LI, Cole GA, Crowley-Nowick PA: Immunotherapy of human cervical high-grade cervical intraepithelial neoplasia with microparticle-delivered human papillomavirus 16 E7 plasmid DNA. Am J Obstet Gynecol. 2003, 188: 916-926. 10.1067/mob.2003.256.

    Article  CAS  PubMed  Google Scholar 

  84. Garcia F, Petry KU, Muderspach L, Gold MA, Braly P, Crum CP, Magill M, Silverman M, Urban RG, Hedley ML, Beach KJ: ZYC101a for treatment of high-grade cervical intraepithelial neoplasia: a randomized controlled trial. Obstet Gynecol. 2004, 103: 317-326. 10.1097/01.AOG.0000110246.93627.17.

    Article  CAS  PubMed  Google Scholar 

  85. Matijevic M, Hedley ML, Urban RG, Chicz RM, Lajoie C, Luby TM: Immunization with a poly (lactide co-glycolide) encapsulated plasmid DNA expressing antigenic regions of HPV 16 and 18 results in an increase in the precursor frequency of T cells that respond to epitopes from HPV 16, 18, 6 and 11. Cell Immunol. 2011, 270: 62-69. 10.1016/j.cellimm.2011.04.005.

    Article  CAS  PubMed  Google Scholar 

  86. Bagarazzi ML, Yan J, Morrow MP, Shen X, Parker RL, Lee JC, Giffear M, Pankhong P, Khan AS, Broderick KE, Knott C, Lin F, Boyer JD, Draghia-Akli R, White CJ, Kim JJ, Weiner DB, Sardesai NY: Immunotherapy against HPV16/ 18 generates potent TH1 and cytotoxic cellular immune responses. Sci Transl Med. 2012, 4: 155ra38-

    Article  CAS  Google Scholar 

  87. Bilu D, Sauder DN: Imiquimod: modes of action. Br J Dermatol. 2003, 149 (Suppl 66): 5-8.

    PubMed  Google Scholar 

  88. Chuang CM, Monie A, Hung CF, Wu TC: Treatment with imiquimod enhances antitumor immunity induced by therapeutic HPV DNA vaccination. J Biomed Sci. 2010, 17: 32-10.1186/1423-0127-17-32.

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  89. Hariharan MJ, Driver DA, Townsend K, Brumm D, Polo JM, Belli BA, Catton DJ, Hsu D, Mittelstaedt D, McCormack JE, Karavodin L, Dubensky TW, Chang SM, Banks TA: DNA immunization against herpes simplex virus: enhanced efficacy using a Sindbis virus-based vector. J Virol. 1998, 72: 950-958.

    PubMed Central  CAS  PubMed  Google Scholar 

  90. Brandsma JL, Shylankevich M, Su Y, Roberts A, Rose JK, Zelterman D, Buonocore L: Vesicular stomatitis virus-based therapeutic vaccination targeted to the E1, E2, E6, and E7 proteins of cottontail rabbit papillomavirus. J Virol. 2007, 81: 5749-5758. 10.1128/JVI.02835-06.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  91. Daemen T, Riezebos-Brilman A, Bungener L, Regts J, Dontje B, Wilschut J: Eradication of established HPV16-transformed tumours after immunisation with recombinant Semliki Forest virus expressing a fusion protein of E6 and E7. Vaccine. 2003, 21: 1082-1088. 10.1016/S0264-410X(02)00558-3.

    Article  CAS  PubMed  Google Scholar 

  92. Berglund P, Quesada-Rolander M, Putkonen P, Biberfeld G, Thorstensson R, Liljeström P: Outcome of immunization of cynomolgus monkeys with recombinant Semliki Forest virus encoding human immunodeficiency virus type 1 envelope protein and challenge with a high dose of SHIV-4 virus. AIDS Res Hum Retroviruses. 1997, 13: 1487-1495. 10.1089/aid.1997.13.1487.

    Article  CAS  PubMed  Google Scholar 

  93. Berglund P, Smerdou C, Fleeton MN, Tubulekas I, Liljeström P: Enhancing immune responses using suicidal DNA vaccines. Nat Biotechnol. 1998, 16: 562-565. 10.1038/nbt0698-562.

    Article  CAS  PubMed  Google Scholar 

  94. Hsu KF, Hung CF, Cheng WF, He L, Slater LA, Ling M, Wu TC: Enhancement of suicidal DNA vaccine potency by linking Mycobacterium tuberculosis heat shock protein 70 to an antigen. Gene Ther. 2001, 8: 376-383. 10.1038/sj.gt.3301408.

    Article  CAS  PubMed  Google Scholar 

  95. Kim TW, Hung CF, Juang J, He L, Hardwick JM, Wu TC: Enhancement of suicidal DNA vaccine potency by delaying suicidal DNA-induced cell death. Gene Ther. 2004, 11: 336-342. 10.1038/sj.gt.3302164.

    Article  CAS  PubMed  Google Scholar 

  96. Herd KA, Harvey T, Khromykh AA, Tindle RW: Recombinant Kunjin virus replicon vaccines induce protective T-cell immunity against human papillomavirus 16 E7-expressing tumour. Virology. 2004, 319: 237-248. 10.1016/j.virol.2003.10.032.

    Article  CAS  PubMed  Google Scholar 

  97. Varnavski AN, Young PR, Khromykh AA: Stable high-level expression of heterologous genes in vitro and in vivo by noncytopathic DNA-based Kunjin virus replicon vectors. J Virol. 2000, 74: 4394-4403. 10.1128/JVI.74.9.4394-4403.2000.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  98. Souders NC, Sewell DA, Pan ZK, Hussain SF, Rodriguez A, Wallecha A, Paterson Y: Listeria-based vaccines can overcome tolerance by expanding low avidity CD8 + T cells capable of eradicating a solid tumor in a transgenic mouse model of cancer. Cancer Immun. 2007, 7: 2-

    PubMed Central  PubMed  Google Scholar 

  99. Sewell DA, Pan ZK, Paterson Y: Listeria-based HPV-16 E7 vaccines limit autochthonous tumor growth in a transgenic mouse model for HPV-16 transformed tumors. Vaccine. 2008, 26: 5315-5320. 10.1016/j.vaccine.2008.07.036.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  100. Quistian-Martinez D, Villatoro-Hernandez J, Loera-Arias MJ, Rangel-Colmenero BR, Zavala-Flores LM, Sepúlveda-Saavedra J, Guzmán-López S, Elizondo-Omaña RE, Montes-de-Oca-Luna R, Saucedo-Cárdenas O: Efficient secretion of a modified E7 protein from human papilloma virus type-16 by Lactococcus lactis. Lett Appl Microbiol. 2010, 51: 383-387. 10.1111/j.1472-765X.2010.02905.x.

    Article  CAS  PubMed  Google Scholar 

  101. Adachi K, Kawana K, Yokoyama T, Fujii T, Tomio A, Miura S, Tomio K, Kojima S, Oda K, Sewaki T, Yasugi T, Kozuma S, Taketani Y: Oral immunization with a Lactobacillus casei vaccine expressing human papillomavirus (HPV) type 16 E7 is an effective strategy to induce mucosal cytotoxic lymphocytes against HPV16 E7. Vaccine. 2010, 28: 2810-2817. 10.1016/j.vaccine.2010.02.005.

    Article  CAS  PubMed  Google Scholar 

  102. Maciag PC, Radulovic S, Rothman J: The first clinical use of a live-attenuated Listeria monocytogenes vaccine: a phase I safety study of Lm-LLO-E7 in patients with advanced carcinoma of the cervix. Vaccine. 2009, 27: 3975-3983. 10.1016/j.vaccine.2009.04.041.

    Article  CAS  PubMed  Google Scholar 

  103. Radulovic S, Brankovic-Magic M, Malisic E, Jankovic R, Dobricic J, Plesinac-Karapandzic V, Maciag PC, Rothman J: Therapeutic cancer vaccines in cervical cancer: phase I study of Lovaxin-C. J BUON. 2009, 14 (Suppl 1): S165-S168.

    PubMed  Google Scholar 

  104. Wallecha A, French C, Petit R, Singh R, Amin A, Rothman J: Lm-LLO-based immunotherapies and HPV-associated disease. J Oncol. 2012, 2012: 542851-

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  105. Lee DW, Anderson ME, Wu S, Lee JH: Development of an adenoviral vaccine against E6 and E7 oncoproteins to prevent growth of human papillomavirus-positive cancer. Arch Otolaryngol Head Neck Surg. 2008, 134: 1316-1323. 10.1001/archoto.2008.507.

    Article  PubMed  Google Scholar 

  106. Gomez-Gutierrez JG, Elpek KG, Montes De Oca-Luna R, Shirwan H, Sam Zhou H, McMasters KM: Vaccination with an adenoviral vector expressing calreticulin-human papillomavirus 16 E7 fusion protein eradicates E7 expressing established tumors in mice. Cancer Immunol Immunother. 2007, 56: 997-1007. 10.1007/s00262-006-0247-2.

    Article  CAS  PubMed  Google Scholar 

  107. Daemen T, Riezebos-Brilman A, Regts J, Dontje B, van der Zee A, Wilschut J: Superior therapeutic efficacy of alpha virus mediated immunization against human papilloma virus type 16 antigens in a murine tumour model: effects of the route of immunization. Antivir Ther. 2004, 9: 733-742.

    PubMed  Google Scholar 

  108. Cassetti MC, McElhiney SP, Shahabi V, Pullen JK, Le Poole IC, Eiben GL, Smith LR, Kast WM: Antitumor efficacy of Venezuelan equine encephalitis virus replicon particles encoding mutated HPV16 E6 and E7 genes. Vaccine. 2004, 22: 520-527. 10.1016/j.vaccine.2003.07.003.

    Article  CAS  PubMed  Google Scholar 

  109. Velders MP, McElhiney S, Cassetti MC, Eiben GL, Higgins T, Kovacs GR, Elmishad AG, Kast WM, Smith LR: Eradication of established tumors by vaccination with Venezuelan equine encephalitis virus replicon particles delivering human papillomavirus 16 E7 RNA. Cancer Res. 2001, 61: 7861-7867.

    CAS  PubMed  Google Scholar 

  110. Hibbitts S: TA-CIN, a vaccine incorporating a recombinant HPV fusion protein (HPV16 L2E6E7) for the potential treatment of HPV16-associated genital diseases. Curr Opin Mol Ther. 2010, 12: 598-606.

    CAS  PubMed  Google Scholar 

  111. Zurkova K, Babiarova K, Hainz P, Krystofova J, Kutinova L, Otahal P, Nemeckova S: The expression of the soluble isoform of hFlt3 ligand by recombinant vaccinia virus enhances immunogenicity of the vector. Oncol Rep. 2009, 21: 1335-1343.

    CAS  PubMed  Google Scholar 

  112. Hsieh CJ, Kim TW, Hung CF, Juang J, Moniz M, Boyd DA, He L, Chen PJ, Chen CH, Wu TC: Enhancement of vaccinia vaccine potency by linkage of tumor antigen gene to gene encoding calreticulin. Vaccine. 2004, 22: 3993-4001. 10.1016/j.vaccine.2004.03.057.

    Article  CAS  PubMed  Google Scholar 

  113. Borysiewicz LK, Fiander A, Nimako M, Man S, Wilkinson GW, Westmoreland D, Evans AS, Adams M, Stacey SN, Boursnell ME, Rutherford E, Hickling JK, Inglis SC: A recombinant vaccinia virus encoding human papillomavirus types 16 and 18, E6 and E7 proteins as immunotherapy for cervical cancer. Lancet. 1996, 347: 1523-1527. 10.1016/S0140-6736(96)90674-1.

    Article  CAS  PubMed  Google Scholar 

  114. Radaelli A, De Giuli Morghen C, Zanotto C, Pacchioni S, Bissa M, Franconi R, Massa S, Paolini F, Muller A, Venuti A: A prime/boost strategy by DNA/fowlpox recombinants expressing a mutant E7 protein for the immunotherapy of HPV-associated cancers. Virus Res. 2012, 170: 44-52. 10.1016/j.virusres.2012.08.007.

    Article  CAS  PubMed  Google Scholar 

  115. Davidson EJ, Boswell CM, Sehr P, Pawlita M, Tomlinson AE, McVey RJ, Dobson J, Roberts JS, Hickling J, Kitchener HC, Stern PL: Immunological and clinical responses in women with vulval intraepithelial neoplasia vaccinated with a vaccinia virus encoding human papillomavirus 16/18 oncoproteins. Cancer Res. 2003, 63: 6032-6041.

    CAS  PubMed  Google Scholar 

  116. Baldwin PJ, van der Burg SH, Boswell CM, Offringa R, Hickling JK, Dobson J, Roberts JS, Latimer JA, Moseley RP, Coleman N, Stanley MA, Sterling JC: Vaccinia-expressed human papillomavirus 16 and 18 e6 and e7 as a therapeutic vaccination for vulval and vaginal intraepithelial neoplasia. Clin Cancer Res. 2003, 9: 5205-5213.

    CAS  PubMed  Google Scholar 

  117. Kaufmann AM, Stern PL, Rankin EM, Sommer H, Nuessler V, Schneider A, Adams M, Onon TS, Bauknecht T, Wagner U, Kroon K, Hickling J, Boswell CM, Stacey SN, Kitchener HC, Gillard J, Wanders J, Roberts JS, Zwierzina H: 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. 2002, 8: 3676-3685.

    CAS  PubMed  Google Scholar 

  118. Brun JL, Dalstein V, Leveque J, Mathevet P, Raulic P, Baldauf JJ, Scholl S, Huynh B, Douvier S, Riethmuller D, Clavel C, Birembaut P, Calenda V, Baudin M, Bory JP: Regression of high-grade cervical intraepithelial neoplasia with TG4001 targeted immunotherapy. Am J Obstet Gynecol. 2011, 204: 169e1-169e8.

    Article  Google Scholar 

  119. Transgene: Transgene reports randomized phase 2b data with its therapeutic HPV vaccine TG4001 in women with CIN2/3 intraepithelial cervical neoplasia. 2012, Available from http://www.transgene.fr/

    Google Scholar 

  120. Corona Gutierrez CM, Tinoco A, Navarro T, Contreras ML, Cortes RR, Calzado P, Reyes L, Posternak R, Morosoli G, Verde ML, Rosales R: Therapeutic vaccination with MVA E2 can eliminate precancerous lesions (CIN 1, CIN 2, and CIN 3) associated with infection by oncogenic human papillomavirus. Hum Gene Ther. 2004, 15: 421-431. 10.1089/10430340460745757.

    Article  CAS  PubMed  Google Scholar 

  121. Garcia-Hernandez E, Gonzalez-Sanchez JL, Andrade-Manzano A, Contreras ML, Padilla S, Guzmán CC, Jiménez R, Reyes L, Morosoli G, Verde ML, Rosales R: Regression of papilloma high-grade lesions (CIN 2 and CIN 3) is stimulated by therapeutic vaccination with MVA E2 recombinant vaccine. Cancer Gene Ther. 2006, 13: 592-597. 10.1038/sj.cgt.7700937.

    Article  CAS  PubMed  Google Scholar 

  122. Albarran YCA, de la Garza A, Cruz Quiroz BJ, Vazquez Zea E, Díaz Estrada I, Mendez Fuentez E, López Contreras M, Andrade-Manzano A, Padilla S, Varela AR, Rosales R: MVA E2 recombinant vaccine in the treatment of human papillomavirus infection in men presenting intraurethral flat condyloma: a phase I/II study. BioDrugs. 2007, 21: 47-59. 10.2165/00063030-200721010-00006.

    Article  Google Scholar 

  123. Sharma C, Dey B, Wahiduzzaman M, Singh N: Human papillomavirus 16 L1–E7 chimeric virus like particles show prophylactic and therapeutic efficacy in murine model of cervical cancer. Vaccine. 2012, 30: 5417-5424. 10.1016/j.vaccine.2012.06.010.

    Article  CAS  PubMed  Google Scholar 

  124. Kaufmann AM, Nieland JD, Jochmus I, Baur S, Friese K, Gabelsberger J, Gieseking F, Gissmann L, Glasschröder B, Grubert T, Hillemanns P, Höpfl R, Ikenberg H, Schwarz J, Karrasch M, Knoll A, Küppers V, Lechmann M, Lelle RJ, Meissner H, Müller RT, Pawlita M, Petry KU, Pilch H, Walek E, Schneider A: Vaccination trial with HPV16 L1E7 chimeric virus-like particles in women suffering from high grade cervical intraepithelial neoplasia (CIN 2/3). Int J Cancer. 2007, 121: 2794-2800. 10.1002/ijc.23022.

    Article  CAS  PubMed  Google Scholar 

  125. Cheever MA, Higano CS: PROVENGE (sipuleucel-T) in prostate cancer: the first FDA 7 approved therapeutic cancer vaccine. Clin Cancer Res. 2011, 17: 3520-3526. 10.1158/1078-0432.CCR-10-3126.

    Article  PubMed  Google Scholar 

  126. McKarney I, Sipuleucel T: Provenge: active cellular immunotherapy for advanced prostate 9 cancer. Issues Emerg Health Technol. 2007, 10: 1-4.

    Google Scholar 

  127. Ferrara A, Nonn M, Sehr P, Schreckenberger C, Pawlita M, Durst M, Schneider A, Kaufmann AM: Dendritic cell-based tumor vaccine for cervical cancer II: results of a clinical 12 pilot study in 15 individual patients. J Cancer Res Clin Oncol. 2003, 129: 521-530. 10.1007/s00432-003-0463-5.

    Article  CAS  PubMed  Google Scholar 

  128. Santin AD, Bellone S, Palmieri M, Zanolini A, Ravaggi A, Siegel ER, Roman JJ, Pecorelli S, Cannon MJ: Human papillomavirus type 16 and 18 E7-pulsed dendritic cell 15 vaccination of stage IB or IIA cervical cancer patients: a phase I escalating-dose trial. J Virol. 2008, 82: 1968-1979. 10.1128/JVI.02343-07.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  129. Tseng CW, Trimble C, Zeng Q, Monie A, Alvarez RD, Huh WK, Hoory T, Wang MC, Hung CF, Wu TC: Low-dose radiation enhances therapeutic HPV DNA vaccination in tumor-bearing hosts. Cancer Immunol Immunother. 2009, 58: 737-748. 10.1007/s00262-008-0596-0.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  130. Kang TH, Lee JH, Song CK, Han HD, Shin BC, Pai SI, Hung CF, Trimble C, Lim JS, Kim TW, Wu TC: Epigallocatechin-3-gallate enhances CD8 + T cell-mediated antitumor immunity induced by DNA vaccination. Cancer Res. 2007, 67: 802-811. 10.1158/0008-5472.CAN-06-2638.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  131. Tseng CW, Hung CF, Alvarez RD, Trimble C, Huh WK, Kim D, Chuang CM, Lin CT, Tsai YC, He L, Monie A, Wu TC: Pretreatment with cisplatin enhances E7-specific CD8+ T-Cell-mediated antitumor immunity induced by DNA vaccination. Clin Cancer Res. 2008, 14: 3185-3192. 10.1158/1078-0432.CCR-08-0037.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  132. Tseng CW, Monie A, Wu CY, Huang B, Wang MC, Hung CF, Wu TC: Treatment with proteasome inhibitor Bortezomib enhances antigen-specific CD8+ T-cell-mediated antitumor immunity induced by DNA vaccination. J Mol Med. 2008, 86: 899-908. 10.1007/s00109-008-0370-y.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  133. Zeng Q, Peng S, Monie A, Yang M, Pang X, Hung CF, Wu TC: Control of cervicovaginal HPV-16 E7-expressing tumors by the combination of therapeutic HPV vaccination and vascular disrupting agents. Hum Gene Ther. 2011, 22: 809-819. 10.1089/hum.2010.071.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  134. Peng S, Monie A, Pang X, Hung CF, Wu TC: Vascular disrupting agent DMXAA enhances the antitumor effects generated by therapeutic HPV DNA vaccines. J Biomed Sci. 2011, 18: 21-10.1186/1423-0127-18-21.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  135. Cao Y, Zhao J, Yang Z, Cai Z, Zhang B, Zhou Y, Shen GX, Chen X, Li S, Huang B: CD4+FOXP3+ regulatory T cell depletion by low-dose cyclophosphamide prevents recurrence in patients with large condylomata acuminata after laser therapy. Clin Immunol. 2010, 136: 21-29. 10.1016/j.clim.2010.02.020.

    Article  CAS  PubMed  Google Scholar 

  136. Bae SH, Park YJ, Park JB, Choi YS, 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. 2007, 13: 341-349. 10.1158/1078-0432.CCR-06-1838.

    Article  CAS  PubMed  Google Scholar 

  137. Petit RG, Basu P: ADXS11-001 immunotherapy targeting HPV-E7: Preliminary survival data from a P2 study in Indian women with recurrent/refractory cervical cancer [abstract]. J Clin Oncol. 2013, 31: 5529-

    Google Scholar 

  138. Haas AR, Sun J, Vachani A, Wallace AF, Silverberg M, Kapoor V, Albelda SM: Cycloxygenase-2 inhibition augments the efficacy of a cancer vaccine. Clin Cancer Res. 2006, 12: 214-222. 10.1158/1078-0432.CCR-05-1178.

    Article  CAS  PubMed  Google Scholar 

  139. Mocellin S, Marincola FM, Young HA: Interleukin-10 and the immune response against cancer: a counterpoint. J Leukoc Biol. 2005, 78: 1043-1051. 10.1189/jlb.0705358.

    Article  CAS  PubMed  Google Scholar 

  140. Van Seters M, Van Beurden M, Ten Kate FJW, Beckmann I, Ewin PC, Eijkemans MJC, Kagie MJ, Meijer CJM, Aaronson NK, KleinJan A, Heijmans-Antonissen C, Zijlstra FJ, Burger MPM, Helmerhors TJ: Treatment of vulvar intraepithelial neoplasia with topical imiquimod. N Engl J Med. 2008, 358: 1465-1473. 10.1056/NEJMoa072685.

    Article  CAS  PubMed  Google Scholar 

  141. Ahn WS, Bae SM, Kim TY, Kim TG, Lee JM, Namkoong SE, Kim CK, Sin JI: 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. 2003, 14: 1389-1399. 10.1089/104303403769211619.

    Article  CAS  PubMed  Google Scholar 

  142. Guzmán-Olea E, Bermúdez-Morales VH, Peralta-Zaragoza O, Torres-Poveda K, Madrid-Marina V: Molecular mechanism and potential targets for blocking HPV-induced lesion development. J Oncol. 2012, 2012: 278312-

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  143. Mkrtichyan M, Najjar YG, Raulfs EC, Abdalla MY, Samara R, Rotem-Yehudar R, Cook L, Khleif SN: Anti-PD-1 synergizes with cyclophosphamide to induce potent anti-tumor vaccine effects through novel mechanisms. Eur J Immunol. 2011, 41: 2977-2986. 10.1002/eji.201141639.

    Article  CAS  PubMed  Google Scholar 

  144. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF: Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Eng J Med. 2012, 366: 2443-2454.

    Article  CAS  Google Scholar 

  145. Brahmer JR, Tykodi SS, Chow LQM, Hwu WJ, Topalian SL, Hwu P: Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Eng J Med. 2012, 366: 2455-2465.

    Article  CAS  Google Scholar 

  146. 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: 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. 2013, 73: 1733-1741. 10.1158/0008-5472.CAN-12-2384.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  147. Graziani G, Tentori L, Navarra P: Ipilimumab: a novel immunostimulatory monoclonal antibody for the treatment of cancer. Pharmacol Res. 2012, 65: 9-22. 10.1016/j.phrs.2011.09.002.

    Article  CAS  PubMed  Google Scholar 

  148. Venuti A, Badaracco G, Rizzo C, Mafera B, Rahimi S, Vigili M: Presence of HPV in head and neck tumours: high prevalence in tonsillar localization. J Exp Clin Cancer Res. 2004, 23: 561-566.

    CAS  PubMed  Google Scholar 

  149. Paolini F, Carbone A, Benevolo M, Silipo V, Rollo F, Covello R, Piemonte P, Frascione P, Capizzi R, Catricalà C, Venuti A: Human Papillomaviruses, p16INK4a and Akt expression in basal cell carcinoma. J Exp Clin Cancer Res. 2011, 30: 108-10.1186/1756-9966-30-108.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This review is not intended to be an encyclopedic one; we thank to all the researchers who have done so powerful work and apologize to the researchers those works were not cited.

We thank Anna Maria Edlisca for editing assistance and Tania Merlino for English editing.

The work was partially supported by AIRC IG 12916 and by Project of Mutual Collaboration of the Planning Department of the National Health Service of Italian Ministry of Health and Regina Elena National Cancer Institute for Implementation of HPV-UNIT.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Aldo Venuti.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

The outline was conceived by AV. All authors contributed to initial drafts, edited version, and the final version. All authors read and approved the final manuscript.

Rights and permissions

Open Access  This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.

The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

To view a copy of this licence, visit https://creativecommons.org/licenses/by/4.0/.

The Creative Commons Public Domain Dedication waiver (https://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Cite this article

Vici, P., Mariani, L., Pizzuti, L. et al. Immunologic treatments for precancerous lesions and uterine cervical cancer. J Exp Clin Cancer Res 33, 29 (2014). https://doi.org/10.1186/1756-9966-33-29

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1756-9966-33-29

Keywords