2,971
Views
61
CrossRef citations to date
0
Altmetric
Reviews

Current therapeutic vaccination and immunotherapy strategies for HPV-related diseases

, , , &
Pages 1418-1429 | Received 04 Nov 2015, Accepted 20 Dec 2015, Published online: 06 Apr 2016

ABSTRACT

Carcinomas of the anogenital tract, in particular cervical cancer, remains one of the most common cancers in women, and represent the most frequent gynecological malignancies and the fourth leading cause of cancer death in women worldwide. Human papillomavirus (HPV)-induced lesions are immunologically distinct in that they express viral antigens, which are necessary to maintain the cancerous phenotype. The causal relationship between HPV infection and anogenital cancer has prompted substantial interest in the development of therapeutic vaccines against high-risk HPV types targeting the viral oncoproteins E6 and E7. This review will focus on the most recent clinical trials for immunotherapies for mucosal HPV-induced lesions as well as emerging therapeutic strategies that have been tested in pre-clinical models for HPV-induced diseases. Progress in peptide- and protein-based vaccines, DNA-based vaccines, viral/bacterial vector-based vaccines, immune checkpoint inhibition, immune response modifiers, and adoptive cell therapy for HPV will be discussed.

Introduction

Human papillomavirus (HPV) is currently the number one sexually transmitted virus.Citation1 The lifetime risk of acquiring one or more anogenital HPV-types is >80%, and it is estimated that two-thirds of women worldwide acquire an HPV infection within 2 years after becoming sexually active.Citation2 The World Health Organization estimates that there are approximately 14 million new HPV infections each year.Citation3 While it has been estimated that 90% of HPV infections are cleared naturally by the immune system, infections by cancer-causing high-risk HPVs (hrHPVs) can persist, remaining largely asymptomatic.Citation4-6 Persistent infection with a hrHPV genotype is associated with an increased risk of developing anogenital cancers. Specifically, hrHPV DNA can be detected in over 99% of cervical cancers with the most common hrHPV types 16 and 18 accounting for 50% and 20% of cervical cancers, respectively.Citation7,8 In addition to HPV16 and HPV18, there are currently 13 other identified hrHPVs including types 31, 33, 35, 39, 45, 51, 52, 56, 58, 59, 68, 73, and 82.Citation9 In 2012, 85% of the 528,000 newly diagnosed cervical cancer cases and 87% of the 266,000 cervical cancer deaths occurred in less developed regions, highlighting socio-economic disparities that exist for HPV-associated diseases.Citation10,11 Even in the US, where screening programs have reduced the overall rate of cervical cancer, inequality exists in the incidence of cervical cancer between white, black, Hispanic, and economically disadvantaged women.Citation12,13 Beyond cervical cancer, hrHPV infections have also been linked to approximately 70% of head and neck squamous cell carcinomas (HNSCC) with HPV16 being detected in over 90% of these cases.Citation14,15 Moreover, it is estimated that the number of HPV-positive HNSCC will surpass the annual number of cervical cancers diagnosed within the US by the year 2020.Citation16

HPVs are non-enveloped, double-stranded DNA viruses of the Papillomaviridae family that consists of over 170 identified members.Citation9 The 8 kb genome encodes for 6 ‘early’ proteins (E1, E2, E4, E5, E6 and E7) and 2 ‘late’ proteins (L1 and L2).Citation17 The early proteins interact with cellular gene products and facilitate viral DNA replication, while the late proteins provide the structural components of the viral capsid and are involved in the packaging of DNA into progeny virions.Citation18 Though HPV DNA usually replicates in episomal form, it has the potential to integrate into host DNA. This event often coincides with the loss of several early (E2, E4 and E5) and late (L1 and L2) genes due to integration, leaving E6 and E7 as the primary protein products expressed within the infected cell.Citation19 Moreover, E2 expression acts as a transcriptional repressor of E6 and E7, and thus the loss of E2 results in upregulation of the E6 and E7 genes, and hence increases downstream protein production. E6 and E7 interact with p53 and retinoblastoma (Rb) proteins respectively, which are important cell cycle regulatory proteins. These interactions result in the disruption of cell cycle regulation and genomic integrity, and are the primary contributors in the transformation of HPV-infected cells into cancer.Citation20

Prospective well-controlled phase 3 trials of prophylactic HPV vaccines have demonstrated effective prevention of high-grade cervical lesions associated with HPV16 and HPV18.Citation21,22 More recently, the nonavalent Gardasil vaccine, 9vHPV, has been FDA approved to cover 7 high-risk genotypes associated with cervical, vulvar, vaginal, and anal cancers (16, 18, 31, 33, 45, 52, and 58), and 2 low-risk genotypes that cause genital warts (6 and 11).Citation23,24 Despite the effectiveness of these vaccines, widespread general adoption and administration of HPV vaccinations is poor in most countries.Citation25 The percentage of girls aged 13-17 receiving at least one dose has improved from 25.1% to 60.0% from 2007 to 2014 in the US, however the percentage receiving the recommended 3 doses is still only 39.7% as of 2014, and this figure remains low at 21.6% in boys aged 13-17 for whom the vaccine has been routinely recommended since 2011.Citation26

Diagnostic tests that detect HPV infections have only been carried out when women present with abnormal cytology on a Papanicolaou (Pap) smear or when women have been in the age range of 30-65.Citation27 However, in April 2014 the FDA approved the use of the Cobas HPV DNA test (Roche) as a primary screening method for detecting HPV in females 25 years of age and older.Citation28 This approval was largely due to results from the Addressing THE Need for Advanced HPV Diagnostics (ATHENA) trial of over 47,000 women in the US, where it was found that 1 in 10 women aged 30 and older who tested positive for HPV16 or 18 had cervical precancer despite exhibiting normal cytology on Pap smears.Citation29,30 Importantly, as HPV DNA testing becomes widely implemented in conjunction with cytological review, there will be a larger number of women who test positive for infection with hrHPV genotypes, potentially with no abnormal cytology or with low-grade cervical intraepithelial neoplastic (CIN) lesions. Furthermore, there are currently no screening recommendations or standardized testing procedures for detecting HPV infection in males, a problem that limits assessing rates of detectable HPV infection to half of the population.

Given that the lifetime risk of HPV infection is high, and that populations severely underrepresented in vaccine coverage will likely continue to develop HPV-related cancers, it is clear that there is a need to develop new therapeutic strategies for HPV. When properly engaged, the human immune system has the ability to eradicate virus-infected cells that express non-self viral antigens. Immunotherapies are a promising strategy against HPV-induced cancers. Therefore, this review will begin with a brief comparison of prophylactic vaccinations and immunotherapeutic approaches, and then will summ-arize current information on HPV therapeutic vaccinations and other immunotherapeutic strategies for HPV-related diseases, followed by a summary of challenges in HPV immunotherapies.

Comparison of prophylactic versus immunotherapeutic strategies for HPV-driven cancers

The primary goal of prophylactic vaccination is to induce a humoral response such that high-titers of HPV-neutralizing antibodies are produced that are capable of preventing initial infections, making HPV antigen-specific B cells the target cell type for these vaccines. HPV vaccines are composed of HPV L1 containing virus-like particles (VLP), mainly of high-risk genotypes, and adjuvants to enhance immune responses.Citation31 When overexpressed in cells, the L1 major capsid protein is capable of spontaneous formation of the icosahedral capsid of the HPV virion,Citation32 mimicking the shape of naturally occurring virions and able to illicit high-titers of HPV-neutralizing antibodies in vaccinated individuals.Citation33 Routine vaccination of girls and young women aged 13-26 against HPV was recommended by The Advisory Committee on Immunization Practices in 2006, followed by boys aged 11-26 in 2011.Citation34 To enhance the immune responses Gardasil has been formulated with the adjuvant amorphous aluminum hydroxyphosphate sulfate,Citation35,36 whereas Cervarix is formulated in the AS04 adjuvant (3-O-desacyl-40-monophosphoryl lipid A and aluminum hydroxide).Citation37 While HPV prophylactic vaccines may induce some degree of cell-mediated immune responses, they are considered irrelevant to cancer prevention given that infected basal epithelial cells that would form tumor cells do not to express significant levels of L1 protein,Citation38 which is only expressed much later in the viral life cycle when infected cells have advanced into the upper epithelial stratum and are dying naturally.Citation39 Due to this, it is no surprise that prophylactic vaccination platforms have failed to report any therapeutic activity in individuals presenting with HPV-positive cancers.

In contrast to prophylactics, immunotherapies for HPV-driven cancers focus on the generation of cellular immune responses against antigens associated with cellular transformation, where antigen specific cytotoxic CD8+ T cells, also known as cytotoxic T-lymphocytes (CTLs), are the primary cell type involved. Given that the HPV16 E6 and E7 proteins are both drivers of unchecked cell proliferation in the most common oncogenic HPV types and are constitutively expressed,Citation40,41 they are the most frequently targeted antigens in HPV therapy development.Citation41 While HPV16 E2 and E5 have recently been investigated as therapeutic targets,Citation42 this review will focus on therapies against E6 and E7 of HPV16 unless otherwise noted. HPV immunotherapies can be broken down into therapeutic vaccines, cell based approaches, and immunomodulators, each of which will be discussed in more detail below.

Therapeutic vaccines

While prophylactic vaccines aim to prevent HPV infections, the benefits of therapeutic vaccine strategies against HPV-driven cancers lie in their potential to generate robust CTL responses against the viral proteins E6 and E7 to clear infected cells. CTL responses against HPV are generated by the presentation of foreign viral antigens on the surface of antigen presenting cells (APC) through MHC class I molecules in the presence of costimulatory molecules to antigen-specific T cells expressing the corresponding T cell receptor (TCR). For example, E6 and E7 contain immunogenic epitopes that are processed and presented in association with the HLA*0201 allele.Citation43,44 Functionally, therapeutic vaccines need to deliver adequate amounts of the viral antigens along with other immune stimulatory components to generate the appropriate immune cells in order to achieve the desired therapeutic outcome, though stimulation against the viral antigens themselves can take on many forms depending on the vaccine platform as discussed below.

Protein and peptide vaccines

The use of whole HPV proteins, like E6 and E7, or HPV fusion proteins as the antigenic source in early HPV therapeutic vaccines has been widely employed.Citation45-48 An advantage of these vaccines is that they do not require the identification of particular HLA-specific epitopes, and the use of whole proteins theoretically covers all available epitopes. However, non-immunogenic peptides may be dominant during antigen processing and presentation, limiting responses to immunogenic peptide sequences. Despite this, the trend of utilizing whole protein is due to the desire to allow presentation of more CTL epitopes and T helper epitopes.Citation49 Many protein-based vaccines have induced HPV-specific CTLs and have induced tumor regression in mice, including those utilizing an E7-Bordetella pertussis CyaA fusion,Citation50 and an E7-HBcAg-Hsp65 fusion.Citation51 Nevertheless, only a handful of protein-based vaccines have reached clinical trials such as a Mycobacterium bovis Hsp65 fused to HPV16 E7 (for review see ref. Citation52).

HPV peptide vaccines utilize only a portion of E6 or E7,Citation53-56 and have several advantages over other platforms including ease of production and safety.Citation52 These vaccines have included a wide range of both short and long E6 and E7 peptides. Short, defined, HLA class I-restricted HPV peptides have thus been used to elicit the desired CTL response.Citation57-59 However, where it is an advantage of whole proteins, a limitation of short peptide-based vaccines includes the necessity to define epitopes for specific HLA alleles. More specifically, at least one epitope for each HLA class I allele are required for wide clinical application of peptide vaccines as patients who have different HLA genetic backgrounds inherently recognize different HPV epitopes.Citation54 To overcome this limitation, a synthetic overlapping long peptide approach has been used since they contain all potential CD4+ and CD8+ epitopes. For example, a mix of long synthetic peptides of E6 and E7 in Montanide ISA-51 (incomplete Freund's adjuvant) were used to vaccinate vulvar intraepithelial neoplasia (VIN) patients, and durable complete regression was observed in nearly half of those vaccinated.Citation60 Recent improvements in the peptide vaccine platform have focused on enhanced peptide-fusion proteins; for example, data from an HPV16 E7 fusion-peptide (TheVax Genetic Vaccine Company) coupled with a GP100 adjuvant (Hawaii Biotech) demonstrated a strong HPV16 E7 CTL response and protected against a tumor challenge in mice. The fusion protein itself contains a full length HPV16 E7 peptide sequence, flanked by an exotoxin from P. aeruginosa and an endoplasmic reticulum retention signal.Citation61 However, even with these promising studies in animal models it still remains that while HPV protein/peptide-based vaccine trials in humans are able to demonstrate HPV-specific CTL response in some patients, often these CTL responses do not correlate with clinical outcome, and tumor regression in most cases has been minimal.Citation49

Bacterial-based and viral vectors

Attenuated bacteria (e.g. Salmonella, Shigella, Yersinia, Saccharomyces cerevisiae, Escherichia coli, and Listeria monocytogenes) can serve as vectors to deliver proteins of interest or plasmids encoding genes in vaccines.Citation62,63 The rationale for the use of microbes as delivery vehicles is based on the ability of these agents to interact with multiple pattern-recognition receptors, such as Toll-like receptors (TLRs), and stimulate robust responses in APCs to induce CTLs against target antigens.Citation64 TLR signaling events play a central role in the stimulation and activation of both innate and adaptive immunity.Citation65 Additionally, viral vectors including adenovirus, vaccinia virus, and a modified vaccinia Ankara (MVA) have been evaluated and tested as platforms for HPV vaccination. Given that adenovirus and vaccinia virus infect dendritic cells (DCs), upregulation of co-stimulatory molecules along with increased cytokine and chemokine production accompanying the infection can contribute to effective CTL induction. For example, an adenoviral vaccine against HPV16 E2 is able to reduce papilloma-forming sites in rabbits.Citation66 Original clinical trials utilizing the vaccinia platform to generate a CTLs against HPV16 and 18 E6 and E7 showed no clinical response for patients with late stage cervical cancer,Citation67 however an MVA vaccine inducing expression of HPV16 E6 and E7 along with the immunostimulant IL-2 (TG4001 from Transgene) showed a clinical response in 7 out of 10 patients, specifically cytologic clearance of CIN2/3 lesions and elimination of HPV16 mRNA and DNA in previously infected tissue.Citation68 Another study utilized a calreticulin (CRT)-E7 fusion protein delivered by a replication-deficient adenovirus vector (Ad-CRT-E7), which had a therapeutic effect against established tumors in mice, and a later tumor re-challenge showed that the generated CTLs had long-term immunological memory against E7-expressing cells.Citation69

The use of replication-deficient forms of viruses such as alphavirus replicon particles, HPV pseudovirions, and adeno-associated viruses have been used to generate potent immunogenic responses with minimal toxicity.Citation70 Alphaviruses such as Semliki Forest virus (SFV) or Venezuelan equine encephalitis virus (VEE) as vaccine platforms have several advantages. Alphaviruses that are engineered to express the RNA of the E6 and E7 oncogenes do so only in the cytosol, eliminating the potential for integration of oncogenes into host cell DNA. Furthermore, VEE replicons have a DC tropism, and there is no preexisting immunity to these viruses in a vast majority of the population.Citation71 SFV- and VEE-based vectors expressing an E6-E7 fusion protein were shown to induce HPV-specific CTLs, prevent tumor establishment of an HPV16-E6/E7 transformed cell line, and eliminate tumors in mice.Citation72,73 Another study showed that the VEE platform with mutated-fused E6 and E7 proteins was able to generate 100% protection from tumor challenge in vaccinated mice and eliminate 90% of established tumors in HLA-A*0201 transgenic mice,Citation74 which is important given that HLA-A*02 is the most prevalent MHC I allele family in humans.Citation75 As for both bacterial and viral based HPV vaccines, immune recognition of components inhibits repeat vaccination with the same delivery system, therefore a heterologous prime-boost strategy may be best for these platforms to maximize the immune response to target HPV antigens.

DNA vaccines

DNA vaccines are yet another potentially powerful and economical vaccine platform. The ability of DNA plasmids to generate full-length peptide sequences and allow processing and presentation of multiple epitopes on both MHC I and II, in addition to their efficient production and ease of storage, could lead to vaccines that provide exceptional immune-stimulating function and that are cost effective.Citation76 Typically, DNA vaccines code for antigens of interest (i.e. E6 and/or E7) that have been cloned into a bacterial vector, which is then optimized for expression in mammalian cells. Importantly, all completed trials have shown that DNA vaccines are safe and well tolerated. However, DNA vaccines designed to induce immune responses against full-length E6 and E7 hold the risk of causing cellular transformation. This hazard can be partially circumvented by mutating the P53 and Rb binding sites in E6 and E7 or more completely through shuffling the gene sequence in a way that allows for expression of putative HLA epitopes but disables the resultant protein.Citation77,78 Although DNA vaccines in animal models showed induction of E6 and E7-specific CTLs and the elimination of established tumors in mice,Citation69,79 clinical trials using DNA vaccines in humans showed no significant responses over spontaneous clearance of CIN lesions.Citation80,81

Recent improvements in HPV DNA vaccine delivery including the use of intramuscular (i.m.) injection coupled with electroporation (EP) have shown promising results.Citation82,83 Briefly, EP consists of applying a series of short electrical pulses at the site of the DNA vaccine injection. These pulses increase plasmid uptake and jumpstart the immune response to the vaccine. In a phase I study, it was shown that the DNA vaccine VGX-3100 combined with EP was able to generate CTLs that could target cells expressing E6 or E7.Citation82 Findings from a phase 2 trial that examined patients with HPV16 or 18 positive CIN2-3 showed histological regression in 53 out of the 107 patients enrolled (49.5%) compared to the 11 out of 36 (30%) in the placebo group. It is important to note that this is the first randomized control trial of a therapeutic vaccine that has shown significant regression in CIN2+ patients.Citation84 Additionally, treatment with GX-188E, a pGX27 vector with shuffled overlapping N- and C-termini of HPV16 and 18 E6 and E7 flanked by the secretory signal sequence of tissue plasminogen activator and the extracellular domain of Fms-related tyrosine kinase 3 ligand, resulted in enhanced poly-functional CTL responses as shown by an increase in cytolytic activity, proliferative capacity, and secretion of effector molecules when exposed to E6 and E7 peptides. Notably, 7 out of 9 patients exhibited complete regression of their lesions and had viral clearance within 36 weeks of follow up.Citation83 Most recently a plasmid encoding HPV-16 E7 antigen linked with calreticulin (CRT), CRT/E7, was co-administered with DNA encoding for bovine papillomavirus (BPV) L1 or L2 resulting in enhanced CD4+ T cell help for improved HPV CTL responses in mice.Citation85

Cell based therapies

Cell-based cancer immunotherapies include DC vaccines and adoptive cell transfer (ACT). These therapies entail the isolation of target cells (e.g., DCs or T cells) from the patient, manipulation of the cells ex vivo (e.g. activation, genetic manipulation, or expansion), and delivery of the cells back into the patient to elicit effector functions.

DC vaccines

DCs are potent APCs, and with proper stimulation they efficiently present antigens with significant levels of costimulation to induce strong CTL responses. As such, DCs have been investigated for usage as cell-based vaccines, in which autologous DCs are either loaded with peptide/protein antigens or transduced to express antigens and then returned to the patient (for review see ref. Citation86). A small number of HPV-targeting DC vaccines have already been investigated in cervical cancer.Citation87-89 For example, autologous DCs pulsed with recombinant HPV16 or HPV18 E7 protein were utilized in a pilot study conducted in stage IV cervical cancer patients. Although, HPV-specific CTLs were measured in some patients, no clinical responses were observed.Citation87 A similar study with DC pulsed with HPV16 and HPV18 E7 delivered with daily low-does IL-2 for several days post vaccination in late-stage cervical cancer patients detected E7-specific CTLs in all patients, though again no clinical responses were observed.Citation88 Another strategy utilized has been DCs transduced with a CD40-targeted adenoviral vector with a mutated HPV16 E7, which resulted in protection against E7-expressing tumors in mice.Citation90 Most recently, a recombinant adenovirus expressing codon-optimized HPV16 E6 and E7 fusion protein (Ad-ofE6E7) was used to transduce DCs, which resulted in robust CTL induction and protective immunity against tumor challenge in mice.Citation91 Limitations of DC vaccines include the requirement of sufficient autologous DCs from each patient, low efficiency of transduction, the inability of terminally differentiated DC to expand ex vivo, and limited DC lifespan .

Adoptive cell transfer

Adoptive cell transfer (ACT) involves the expansion of autologous antigen specific CTLs ex vivo prior to delivery back into a patient. ACT offers the following advantages: antigen-specific CTLs that can be expanded to large numbers in vitro; CTLs can be genetically engineered and/or activated ex vivo to acquire anti-antigenic functions; and the host can be manipulated before cell transfer to eliminate suppressor cells such as regulatory T cells (Treg). The transferred CTLs in ACT act as “living drugs” and can induce long-term protection.Citation92 In a recent pilot study, complete regression was observed in 2 out of 9 metastatic cervical cancer patients after a single infusion of HPV16 E6 and E7 reactive CTLs.Citation93 Additionally, CTLs were transduced to express a TCR against an HPV E7 epitope, and these CTLs showed functional responses toward the target antigen in vitro.Citation94 Most recently, this approach was used to introduce another TCR against E6 into CTLs, which were then able to kill HPV+ cells from cervical and head and neck cancer cell lines,Citation95 and this is now being used in an ongoing clinical trial (NCT02280811).

Immunomodulators

It is important to note that successful therapeutic vaccines developed from the aforementioned platforms likely result in the generation of HPV-type specific responses to target proteins. While there have been a handful of studies that suggest histological regression regardless of HPV-type after treatment, including one study that found significant regression of late-stage CIN over spontaneous clearance,Citation96-98 there has yet to be an identified T-cell epitope that shows cross-reactivity across multiple HPV-types. Because of the limitations imposed by type-specificity in therapeutic vaccines, immunomodulators that can stimulate positive or negative immune system responses regardless of HPV-type are an attractive option for therapy. Here we will discuss a form of negative modulation known as checkpoint inhibition, as well as positive modulation with non-specific immunostimulants, and their use in HPV immunotherapies.

Checkpoint inhibitors

Though promising, multiple inhibitory mechanisms limit endogenous CTLs and modified T cells used in ACT therapies from effective tumor eradication. Beyond Tregs and myeloid-derived suppressor cells (MDSC) that populate the suppressive tumor microenvironment,Citation99 T cells have intrinsic regulatory mechanisms that abolish their effector functions. For example, activated CTLs upregulate inhibitory receptors including CTL-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1),Citation100,101 of which ligand engagement transduces inhibitory signals to attenuate CTL activation.Citation102 Briefly, CTLA-4 competes with CTL surface CD28 for CD86 binding on APC, and the interaction of CTLA-4 with CD86 results in CTL attenuation, decreased proliferation, and an increased threshold for activation.Citation103-105 Moreover, HPV-positive tumor cells often present with upregulated levels of the PD-1 ligand, programmed death ligand-1 (PD-L1), on their surface,Citation106,107 leading to an inverse correlation between PD-L1 expression and the number of CTLs within tumors.Citation108-110 As such, these receptors act as checkpoints that protect against unbridled CTL responses. This has led to new approaches in CTL-targeted immunotherapies focused on disrupting these receptor-ligand interactions, and compounds that accomplish this are known as checkpoint inhibitors.Citation111 Utilizing antibodies targeted against CTLA-4, PD-1, PDL-1, or more importantly a combination of these receptors results in a prevention of suppressive signaling events from taking place, resulting in enhanced CTL activity.Citation112,113 Though agents against CTLA-4 and PD-1 were the first checkpoint inhibitors tested, combinations of PD-1 with LAG-3,Citation114 TIM-3,Citation115,116 TIGIT,Citation117 or BTLA Citation118 have been shown to bolster anti-tumor CTL function.

One benefit of checkpoint inhibitors compared to T cell therapies is the ease of generalizability to a spectrum of malignancies. Unlike ACT that relies on the presence of cancer-specific antigens and ex vivo manipulation, checkpoint inhibitors can be used as off-the-shelf treatments that can be applied across tumor types. There are currently several clinical trials examining the effects of checkpoint inhibitors in patients with advanced stage HPV-associated diseases. Ipilimumab (Yervoy by Bristol-Myers Squibb), an anti-CTLA-4 monoclonal antibody, is being tested by our lab in a phase I study of chemoradiation followed by ipilimumab for advanced cervical cancer patients (NCT01711515) as well as in a phase II study for HPV+ cervical cancer that is either metastatic or recurrent (NCT01693783). Dual checkpoint inhibition is being examined with an anti-PD-L1 antibody combined with tremelimumab, another anti-CTLA-4 antibody, in 2 phase I trials for patients with solid tumors (NCT02261220) and patients with cervical cancer (NCT01975831). Furthermore, combination approaches that are examining a “brakes off, gas on” approach with the immunomodulator OX40 (MedImmune/AstraZeneca) and either anti-PD1 or anti-CTLA4 and are underway in phase I and II trials for patients with advanced stage solid tumors, including cervical cancers (NCT02205333).

Non-specific immunostimulants

Non-specific immunostimulants are agents that can stimulate the immune system through increasing the activity of certain immune cells, and that can act irrespective of antigen specificity. Toll-like receptors (TLRs) are a family of receptors expressed by APC and recognize pathogen associated molecular patterns (PAMPs), which are general patterns associated with different classes of pathogens. These PAMPs provide danger signals to APC to induce potent activation. For instance, TLR3 is found primarily in the endosomes of APC, including Langerhans cells (LC), the resident APC of the epithelial and mucosal layers where HPV infects, and recognizes viral dsRNA.Citation119,120 Along these lines, it was shown that Poly-I:C, a dsRNA viral mimic TLR3 agonist, used as an adjuvant for an intravaginal HPV-vaccine increased mucosal E7-specific CTL responses in mice.Citation121 A poly-lysine stabilized form of Poly-I:C, Poly-ICLC, was also shown to potently activate LC, which were subsequently able to induce robust E7-specific CTL responses in vitro.Citation122,123 Similar results were seen via LC treatment with TLR8 or TLR7/8 agonists (3M-002 and resiquimod, respectively).Citation124 Importantly, these studies demonstrated that TLR agonists could activate LC that had naturally processed and presented E7 antigens, suggesting that such compounds could activate APC such as LC at the site of HPV infection where they take up antigens in vivo, and subsequently initiate HPV-specific CTL responses without the need for exogenous administration of HPV antigens. Alternative approaches have included using a mix of naturally produced cytokines from stimulated PBMC to activate HPV-exposed LC.Citation125

Immune modulating mechanisms caused by HPV-driven disease is a challenge for immunotherapies

HPV utilizes a variety of immune escape mechanisms to evade immune responses during initial and recurrent infections.Citation126 For example, HPV infection is associated with the suppression of MHC class I antigen presentation by infected cells via HPV16 E5, which functionally inhibits the transport of class I molecules through the golgi apparatus.Citation127 Additional evidence suggests that the interaction of α and β HPVs with LC (epithelial APC) may delay or prevent adaptive immune responses.Citation128,129 Specifically, it has been shown that HPV16 entry via the annexin A2/S100A10 heterotetramer causes suppression of LC maturation resulting in failure of CTL induction.Citation130 These mechanisms may act as barriers for treatments of early stage disease (CIN I). In the case of progressive changes, while many of the aforementioned immunotherapies are able to overcome barriers associated with inducing HPV-specific CTLs, they have all fallen short in effectiveness. Several underlying immunosuppressive mechanisms caused by HPV-driven disease are hypothesized to prevent CTLs from eliminating transformed cells or even homing into tumors. For example, the ability of CTLs to migrate into CIN lesions can be a predictor of disease regression, however it has been found that as patients develop higher grade CIN2/3 there are fewer CTL found within lesions due to multiple mechanisms including the dysregulation of addressin cell adhesion molecule-1.Citation131 In addition to preventing CTL infiltration, as CIN progresses into high grade, the expression PD-1 and PDL-1 on cervical T-cells and DC is enhanced, respectively.Citation132 These changes synergize with the increasingly immunosuppressive microenvironment created through the distorted equilibrium of type-1 T-helper (Th1) cells and Th2 cells.Citation133 Specifically, as disease advances from low grade CIN toward cancer it has been shown that Th1 cytokines (IL-2, IL-12, and TNFα) are reduced while Th2 cytokines are increased.Citation132,134,135 As a result there is a parallel increase between CIN progression and the number of immunosuppressive cells found within the microenvironment of the CIN lesion. This cell population include TregsCitation136,137 and MDSC,Citation138,139 both of which generate suppressive cytokines such as IL-10,Citation140,141 IL-4, and TGF-β.142 and are involved in the inhibition of CTL responses.

Because these changes show that the microenvironment of lesions becomes more immunosuppressive as HPV-disease progresses it is reasonable to assume that patients with more advanced disease will be more difficult to treat with immunotherapy as they develop higher grades of CIN and eventually cancer. Work has shown that appearance of naturally occurring HPV-specific CTLs detected in peripheral blood capable of responding to the patient's HPV-type are not indicative of disease clearance.Citation143,144 Paradoxically, some patients treated with a single dose of ACT comprised of a large number of ex vivo genetically engineered HPV-specific CTL have clinical responses involving regression of metastatic cancer lesions (2 complete and 1 partial response out of 9 patients).Citation145 These latter findings suggest that cervical cancer is amenable to T cell immunotherapy even in later stages of disease and CTL infiltration into tumors can occur, however it remains that single-pronged approaches as treatments in HPV-associated disease will most likely fail to overcome the multiple immune barriers generated within transforming/transformed tissue.

Summary and perspective

Promising clinical trials utilizing most of the aforementioned therapies that have recently been completed as of 2012 or are ongoing have been summarized in , for a comprehensive list see Kallhouf et al. 2014.Citation146 However, it still remains that initial attempts at generating effector T cell responses via therapeutic vaccinations have fallen short of expectations. Therefore, there is still a significant need to develop new treatments for HPV-associated diseases. Given our current understanding of HPV-cellular interactions, the interplay of the virus with the host immune system, and the hallmark changes that occur in the tumor microenvironment, the field may now be on the forefront of identifying clinically effective immunotherapies against HPV-transformed cells. One lesson learned from the previous immunotherapy studies to date is that the in vivo induction or ex vivo generation of HPV-specific CTLs alone may not be sufficient for eliminating cancer cells in individuals with advanced-staged diseases. This is because HPV-driven cancers utilize multiple mechanisms to evade the immune system, some of which may still be unknown. Due to this, effective immunotherapeutic regimens in the future will likely require a multi-pronged approach. Therefore, in addition to the process of generating antigen-specific CTLs, the focus should shift to modulation of the tumor microenvironment, elimination of Tregs, inhibition of immune attenuation via checkpoint inhibitors, and stimulation of immune responses with immunostimulants, in a synergistic manner in order to most effectively battle HPV-associated diseases.

Table 1. RECENT AND ONGOING CLINICAL TRIALS FOR IMMUNOTHERAPY OF HPV-INDUCED CANCERS

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

Funding

This study was supported by NIH grants R01 CA074397 and RC2 CA148298 to W. M. K. who holds the Walter A. Richter Cancer Research Chair. Additional support was received from NIH grant P30 CA014089 (Norris Comprehensive Cancer Center Support Grant), the Keck School of Medicine/USC Graduate School PhD Fellowship (to J. G. S.), the ARCS Foundation John and Edith Leonis Award (to A. W. W.) and SC-CTSI (NIH/NCRR/NCATS) grant TL1TR000132 (to A. W. W.). Support from The Netherlands American Foundation, Ella Selders, Yvonne Bogdanovich, Johannes Van Tilburg, Christine Ofiesh, and Sammie's Circle is gratefully acknowledged.

References

  • Dunne EF, Unger ER, Sternberg M, McQuillan G, Swan DC, Patel SS, Markowitz LE. Prevalence of HPV infection among females in the United States. Jama-J Am Med Assoc 2007; 297:813-9; http://dx.doi.org/10.1001/jama.297.8.813
  • Koutsky L. Epidemiology of genital human papillomavirus infection. Am J Med 1997; 102:3-8; PMID:9217656; http://dx.doi.org/10.1016/S0002-9343(97)00177-0
  • Satterwhite CL, Torrone E, Meites E, Dunne EF, Mahajan R, Ocfemia MC, Su J, Xu F, Weinstock H. Sexually transmitted infections among US women and men: prevalence and incidence estimates, 2008. Sex Transm Dis 2013; 40:187-93; PMID:23403598; http://dx.doi.org/10.1097/OLQ.0b013e318286bb53
  • Franco EL, Villa LL, Sobrinho JP, Prado JM, Rousseau MC, Desy M, Rohan TE. Epidemiology of acquisition and clearance of cervical human papillomavirus infection in women from a high-risk area for cervical cancer. J Infect Dis 1999; 180:1415-23; PMID:10515798; http://dx.doi.org/10.1086/315086
  • Ho GY, Burk RD, Klein S, Kadish AS, Chang CJ, Palan P, Basu J, Tachezy R, Lewis R, Romney S. Persistent genital human papillomavirus infection as a risk factor for persistent cervical dysplasia. J Natl Cancer Inst 1995; 87:1365-71; PMID:7658497; http://dx.doi.org/10.1093/jnci/87.18.1365
  • Moscicki AB. Impact of HPV infection in adolescent populations. J Adolesc Health 2005; 37:S3-9; PMID:16310138; http://dx.doi.org/10.1016/j.jadohealth.2005.09.011
  • Walboomers JM, Jacobs MV, Manos MM, Bosch FX, Kummer JA, Shah KV, Snijders PJ, Peto J, Meijer CJ, Munoz N. Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol 1999; 189:12-9; PMID:10451482; http://dx.doi.org/10.1002/(SICI)1096-9896(199909)189:1%3c12::AID-PATH431%3e3.0.CO;2-F
  • Bosch FX, Manos MM, Munoz N, Sherman M, Jansen AM, Peto J, Schiffman MH, Moreno V, Kurman R, Shah KV. Prevalence of human papillomavirus in cervical cancer: a worldwide perspective. International biological study on cervical cancer (IBSCC) Study Group. J Natl Cancer Inst 1995; 87:796-802; PMID:7791229; http://dx.doi.org/10.1093/jnci/87.11.796
  • Ghittoni R, Accardi R, Chiocca S, Tommasino M. Role of human papillomaviruses in carcinogenesis. Ecancermedicalscience 2015; 9:526; PMID:25987895; http://dx.doi.org/10.3332/ecancer.2015.526
  • Forman D, de Martel C, Lacey CJ, Soerjomataram I, Lortet-Tieulent J, Bruni L, Vignat J, Ferlay J, Bray F, Plummer M, et al. Global burden of human papillomavirus and related diseases. Vaccine 2012; 30 Suppl 5:F12-23; PMID:23199955; http://dx.doi.org/10.1016/j.vaccine.2012.07.055
  • Ferlay J SI, Ervik M, Dikshit R, Esser 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]. Lyon, France: International Agency for Research on Cancer; 2013, 2013.
  • Watson M, Saraiya M, Benard V, Coughlin SS, Flowers L, Cokkinides V, Schwenn M, Huang Y, Giuliano A. Burden of cervical cancer in the United States, 1998-2003. Cancer 2008; 113:2855-64; PMID:18980204; http://dx.doi.org/10.1002/cncr.23756
  • Smith JS. Ethnic disparities in cervical cancer illness burden and subsequent care: a prospective view in managed care. The American journal of managed care 2008; 14:S193-9; PMID:18611087
  • Mehanna H, Beech T, Nicholson T, El-Hariry I, McConkey C, Paleri V, Roberts S. Prevalence of human papillomavirus in oropharyngeal and nonoropharyngeal head and neck cancer–systematic review and meta-analysis of trends by time and region. Head Neck 2013; 35:747-55; PMID:22267298; http://dx.doi.org/10.1002/hed.22015
  • Ragin CC, Taioli E. Survival of squamous cell carcinoma of the head and neck in relation to human papillomavirus infection: review and meta-analysis. Int J Cancer 2007; 121:1813-20; PMID:17546592; http://dx.doi.org/10.1002/ijc.22851
  • Chaturvedi AK, Engels EA, Pfeiffer RM, Hernandez BY, Xiao W, Kim E, Jiang B, Goodman MT, Sibug-Saber M, Cozen W, et al. Human papillomavirus and rising oropharyngeal cancer incidence in the United States. J Clin Oncol 2011; 29:4294-301; PMID:21969503; http://dx.doi.org/10.1200/JCO.2011.36.4596
  • Ganguly N, Parihar SP. Human papillomavirus E6 and E7 oncoproteins as risk factors for tumorigenesis. J Biosci 2009; 34:113-23; PMID:19430123; http://dx.doi.org/10.1007/s12038-009-0013-7
  • Buck CB, Thompson CD, Pang YY, Lowy DR, Schiller JT. Maturation of papillomavirus capsids. J Virol 2005; 79:2839-46; PMID:15709003; http://dx.doi.org/10.1128/JVI.79.5.2839-2846.2005
  • Hafner N, Driesch C, Gajda M, Jansen L, Kirchmayr R, Runnebaum IB, Durst M. Integration of the HPV16 genome does not invariably result in high levels of viral oncogene transcripts. Oncogene 2008; 27:1610-7; PMID:17828299; http://dx.doi.org/10.1038/sj.onc.1210791
  • zur Hausen H. Papillomaviruses and cancer: from basic studies to clinical application. Nat Rev Cancer 2002; 2:342-50; PMID:12044010; http://dx.doi.org/10.1038/nrc798
  • Group TFIS. Quadrivalent vaccine against human papillomavirus to prevent high-grade cervical lesions. N Engl J Med 2007; 356:1915-27; PMID:17494925; http://dx.doi.org/10.1056/NEJMoa061741
  • FDA licensure of quadrivalent human papillomavirus vaccine (HPV4, Gardasil) for use in males and guidance from the Advisory Committee on Immunization Practices (ACIP). MMWR Morbidity and mortality weekly report 2010; 59:630-2; PMID:20508594
  • Kirby T. FDA approves new upgraded Gardasil 9. The Lancet Oncology 2015; 16:e56; PMID:25532625; http://dx.doi.org/10.1016/S1470-2045(14)71191-X
  • Joura EA, Giuliano AR, Iversen OE, Bouchard C, Mao C, Mehlsen J, Moreira ED, Jr., Ngan Y, Petersen LK, Lazcano-Ponce E, et al. A 9-valent HPV vaccine against infection and intraepithelial neoplasia in women. N Engl J Med 2015; 372:711-23; PMID:25693011; http://dx.doi.org/10.1056/NEJMoa1405044
  • Stokley S, Jeyarajah J, Yankey D, Cano M, Gee J, Roark J, Curtis RC, Markowitz L. Human papillomavirus vaccination coverage among adolescents, 2007-2013, and postlicensure vaccine safety monitoring, 2006-2014–United States. MMWR Morbidity and mortality weekly report 2014; 63:620-4; PMID:25055185
  • Reagan-Steiner S, Yankey D, Jeyarajah J, Elam-Evans LD, Singleton JA, Curtis CR, MacNeil J, Markowitz LE, Stokley S. National, Regional, State, and Selected Local Area Vaccination Coverage Among Adolescents Aged 13-17 Years–United States, 2014. MMWR Morb Mortal Wkly Rep 2015; 64:784-92; PMID:26225476; http://dx.doi.org/10.15585/mmwr.mm6429a3
  • Saslow D, Solomon D, Lawson HW, Killackey M, Kulasingam SL, Cain J, Garcia FA, Moriarty AT, Waxman AG, Wilbur DC, et al. American Cancer Society, American Society for Colposcopy and Cervical Pathology, and American Society for Clinical Pathology screening guidelines for the prevention and early detection of cervical cancer. Am J Clin Pathol 2012; 137:516-42; PMID:22431528; http://dx.doi.org/10.1309/AJCPTGD94EVRSJCG
  • Administration USFaD. FDA approves first human papillomavirus test for primary cervical cancer screening. 2014.
  • Stoler MH, Wright TC, Jr., Sharma A, Apple R, Gutekunst K, Wright TL, Group AHS. High-risk human papillomavirus testing in women with ASC-US cytology: results from the ATHENA HPV study. Am J Clin Pathol 2011; 135:468-75; PMID:21350104; http://dx.doi.org/10.1309/AJCPZ5JY6FCVNMOT
  • Wright TC, Jr., Stoler MH, Sharma A, Zhang G, Behrens C, Wright TL. Evaluation of HPV-16 and HPV-18 genotyping for the triage of women with high-risk HPV+ cytology-negative results. American journal of clinical pathology 2011; 136:578-86; PMID:21917680; http://dx.doi.org/10.1309/AJCPTUS5EXAS6DKZ
  • Schmiedeskamp MR, Kockler DR. Human papillomavirus vaccines. Ann Pharmacother 2006; 40:1344-52; PMID:16849621; http://dx.doi.org/10.1345/aph.1G723
  • Kirnbauer R, Booy F, Cheng N, Lowy DR, Schiller JT. Papillomavirus L1 major capsid protein self-assembles into virus-like particles that are highly immunogenic. Proc Natl Acad Sci U S A 1992; 89:12180-4; PMID:1334560; http://dx.doi.org/10.1073/pnas.89.24.12180
  • Stanley M. Immune responses to human papillomavirus. Vaccine 2006; 24 Suppl 1:S16-22; PMID:16219398; http://dx.doi.org/10.1016/j.vaccine.2005.09.002
  • Centers for Disease C, Prevention. Recommendations on the use of quadrivalent human papillomavirus vaccine in males–Advisory Committee on Immunization Practices (ACIP), 2011. MMWR Morbidity and mortality weekly report 2011; 60:1705-8; PMID:22189893
  • Markowitz LE, Dunne EF, Saraiya M, Lawson HW, Chesson H, Unger ER, Centers for Disease C, Prevention, Advisory Committee on Immunization P. Quadrivalent Human Papillomavirus Vaccine: Recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep 2007; 56:1-24; PMID:17380109
  • Barr E, Tamms G. Quadrivalent human papillomavirus vaccine. Clin Infect Dis 2007; 45:609-7; PMID:17682997; http://dx.doi.org/10.1086/520654
  • Harper DM, Franco EL, Wheeler CM, Moscicki AB, Romanowski B, Roteli-Martins CM, Jenkins D, Schuind A, Costa Clemens SA, Dubin G, et al. Sustained efficacy up to 4.5 years of a bivalent L1 virus-like particle vaccine against human papillomavirus types 16 and 18: follow-up from a randomised control trial. Lancet 2006; 367:1247-55; PMID:16631880; http://dx.doi.org/10.1016/S0140-6736(06)68439-0
  • Ault KA, Group FIS. Effect of prophylactic human papillomavirus L1 virus-like-particle vaccine on risk of cervical intraepithelial neoplasia grade 2, grade 3, and adenocarcinoma in situ: a combined analysis of four randomised clinical trials. Lancet 2007; 369:1861-8; PMID:17544766; http://dx.doi.org/10.1016/S0140-6736(07)60852-6
  • Kajitani N, Satsuka A, Kawate A, Sakai H. Productive Lifecycle of Human Papillomaviruses that Depends Upon Squamous Epithelial Differentiation. Front Microbiol 2012; 3:152; PMID:22536200; http://dx.doi.org/10.3389/fmicb.2012.00152
  • Hudson JB, Bedell MA, McCance DJ, Laiminis LA. Immortalization and altered differentiation of human keratinocytes in vitro by the E6 and E7 open reading frames of human papillomavirus type 18. J Virol 1990; 64:519-26; PMID:2153221
  • Tan S, de Vries EG, van der Zee AG, de Jong S. Anticancer drugs aimed at E6 and E7 activity in HPV-positive cervical cancer. Curr Cancer Drug Targets 2012; 12:170-84; PMID:22165971; http://dx.doi.org/10.2174/156800912799095135
  • Ganguly N. Human papillomavirus-16 E5 protein: oncogenic role and therapeutic value. Cell Oncol (Dordr) 2012; 35:67-76; PMID:22262402; http://dx.doi.org/10.1007/s13402-011-0069-x
  • Steller MA, Gurski KJ, Murakami M, Daniel RW, Shah KV, Celis E, Sette A, Trimble EL, Park RC, Marincola FM. 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; 4:2103-9; PMID:9748126
  • Kast WM, Brandt RM, Sidney J, Drijfhout JW, Kubo RT, Grey HM, Melief CJ, Sette A. Role of HLA-A motifs in identification of potential CTL epitopes in human papillomavirus type 16 E6 and E7 proteins. J Immunol 1994; 152:3904-12; PMID:7511661
  • Davidson EJ, Faulkner RL, Sehr P, Pawlita M, Smyth LJ, Burt DJ, Tomlinson AE, Hickling J, Kitchener HC, Stern PL. Effect of TA-CIN (HPV 16 L2E6E7) booster immunisation in vulval intraepithelial neoplasia patients previously vaccinated with TA-HPV (vaccinia virus encoding HPV 16/18 E6E7). Vaccine 2004; 22:2722-9; PMID:15246603; http://dx.doi.org/10.1016/j.vaccine.2004.01.049
  • Frazer IH, Quinn M, Nicklin JL, Tan J, Perrin LC, Ng P, O'Connor VM, White O, Wendt N, Martin J, et al. Phase 1 study of HPV16-specific immunotherapy with E6E7 fusion protein and ISCOMATRIX adjuvant in women with cervical intraepithelial neoplasia. Vaccine 2004; 23:172-81; PMID:15531034; http://dx.doi.org/10.1016/j.vaccine.2004.05.013
  • Einstein MH, Kadish AS, Burk RD, Kim MY, Wadler S, Streicher H, Goldberg GL, Runowicz CD. Heat shock fusion protein-based immunotherapy for treatment of cervical intraepithelial neoplasia III. Gynecol Oncol 2007; 106:453-60; PMID:17586030; http://dx.doi.org/10.1016/j.ygyno.2007.04.038
  • de Jong A, O'Neill T, Khan AY, Kwappenberg KM, Chisholm SE, Whittle NR, Dobson JA, Jack LC, St Clair Roberts JA, Offringa R, et al. Enhancement of human papillomavirus (HPV) type 16 E6 and E7-specific T-cell immunity in healthy volunteers through vaccination with TA-CIN, an HPV16 L2E7E6 fusion protein vaccine. Vaccine 2002; 20:3456-64; PMID:12297390; http://dx.doi.org/10.1016/S0264-410X(02)00350-X
  • Brinkman JA, Hughes SH, Stone P, Caffrey AS, Muderspach LI, Roman LD, Weber JS, Kast WM. Therapeutic vaccination for HPV induced cervical cancers. Dis Markers 2007; 23:337-52; PMID:17627067; http://dx.doi.org/10.1155/2007/245146
  • Berraondo P, Nouze C, Preville X, Ladant D, Leclerc C. Eradication of large tumors in mice by a tritherapy targeting the innate, adaptive, and regulatory components of the immune system. Cancer Research 2007; 67:8847-55; PMID:17875726; http://dx.doi.org/10.1158/0008-5472.CAN-07-0321
  • Zhou CM, Zhang GX, Ma XX. Characterization and evaluation of the immune responses elicited by a novel human papillomavirus (HPV) therapeutic vaccine: HPV 16E7-HBcAg-Hsp65 fusion protein. J Virol Methods 2014; 197:1-6; PMID:24291739; http://dx.doi.org/10.1016/j.jviromet.2013.10.033
  • Khallouf H, Grabowska AK, Riemer AB. Therapeutic Vaccine Strategies against Human Papillomavirus. Vaccines (Basel) 2014; 2:422-62; PMID:26344626; http://dx.doi.org/10.3390/vaccines2020422
  • Jang S, Kim YT, Chung HW, Lee KR, Lim JB, Lee K. Identification of novel immunogenic human leukocyte antigen-A 2402-binding epitopes of human papillomavirus type 16 E7 for immunotherapy against human cervical cancer. Cancer 2012; 118:2173-83; PMID:21918960; http://dx.doi.org/10.1002/cncr.26468
  • Kim S, Chung HW, Lee KR, Lim JB. Identification of novel epitopes from human papillomavirus type 18 E7 that can sensitize PBMCs of multiple HLA class I against human cervical cancer. J Transl Med 2014; 12:229; PMID:25141788; http://dx.doi.org/10.1186/s12967-014-0229-7
  • Kenter GG, Welters MJ, Valentijn AR, Lowik MJ, Berends-van der Meer DM, Vloon AP, Drijfhout JW, Wafelman AR, Oostendorp J, Fleuren GJ, et al. Phase I immunotherapeutic trial with long peptides spanning the E6 and E7 sequences of high-risk human papillomavirus 16 in end-stage cervical cancer patients shows low toxicity and robust immunogenicity. Clin Cancer Res 2008; 14:169-77; PMID:18172268; http://dx.doi.org/10.1158/1078-0432.CCR-07-1881
  • Rudolf MP, Man S, Melief CJ, Sette A, Kast WM. Human T-cell responses to HLA-A-restricted high binding affinity peptides of human papillomavirus type 18 proteins E6 and E7. Clin Cancer Res 2001; 7:788s-95s; PMID:11300474
  • Solares AM, Baladron I, Ramos T, Valenzuela C, Borbon Z, Fanjull S, Gonzalez L, Castillo D, Esmir J, Granadillo M, et al. Safety and Immunogenicity of a Human Papillomavirus Peptide Vaccine (CIGB-228) in Women with High-Grade Cervical Intraepithelial Neoplasia: First-in-Human, Proof-of-Concept Trial. ISRN Obstet Gynecol 2011; 2011:292951; PMID:21748025; http://dx.doi.org/10.5402/2011/292951
  • 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; 6:3406-16; PMID:10999722
  • van Driel WJ, Ressing ME, Kenter GG, Brandt RM, Krul EJ, van Rossum AB, Schuuring E, Offringa R, Bauknecht T, Tamm-Hermelink A, et al. Vaccination with HPV16 peptides of patients with advanced cervical carcinoma: clinical evaluation of a phase I-II trial. Eur J Cancer 1999; 35:946-52; PMID:10533477; http://dx.doi.org/10.1016/S0959-8049(99)00048-9
  • Kenter GG, Welters MJ, Valentijn AR, Lowik MJ, Berends-van der Meer DM, Vloon AP, Essahsah F, Fathers LM, Offringa R, Drijfhout JW, et al. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med 2009; 361:1838-47; PMID:19890126; http://dx.doi.org/10.1056/NEJMoa0810097
  • Wu CW, J; Hwang, K. Preclinical Efficacy and Safety Studies of PEK Fusion Protein in GPI-0100 Adjuvant for HPV-infectious Disease Immnotherapy. HPV 2015 - Lisbon 2015.
  • Paterson Y, Maciag PC. Listeria-based vaccines for cancer treatment. Curr Opin Mol Ther 2005; 7:454-60; PMID:16248280
  • Carleton HA. Pathogenic bacteria as vaccine vectors: teaching old bugs new tricks. Yale J Biol Med 2010; 83:217-22; PMID:21165341
  • van Duin D, Medzhitov R, Shaw AC. Triggering TLR signaling in vaccination. Trends Immunol 2006; 27:49-55; PMID:16310411; http://dx.doi.org/10.1016/j.it.2005.11.005
  • Trinchieri G, Sher A. Cooperation of Toll-like receptor signals in innate immune defence. Nat Rev Immunol 2007; 7:179-90; PMID:17318230; http://dx.doi.org/10.1038/nri2038
  • Brandsma JL, Shlyankevich M, Zhang L, Slade MD, Goodwin EC, Peh W, Deisseroth AB. Vaccination of rabbits with an adenovirus vector expressing the papillomavirus E2 protein leads to clearance of papillomas and infection. J Virol 2004; 78:116-23; PMID:14671093; http://dx.doi.org/10.1128/JVI.78.1.116-123.2004
  • Borysiewicz LK, Fiander A, Nimako M, Man S, Wilkinson GW, Westmoreland D, Evans AS, Adams M, Stacey SN, Boursnell ME, et al. A recombinant vaccinia virus encoding human papillomavirus types 16 and 18, E6 and E7 proteins as immunotherapy for cervical cancer. Lancet 1996; 347:1523-7; PMID:8684105; http://dx.doi.org/10.1016/S0140-6736(96)90674-1
  • Brun JL, Dalstein V, Leveque J, Mathevet P, Raulic P, Baldauf JJ, Scholl S, Huynh B, Douvier S, Riethmuller D, et al. Regression of high-grade cervical intraepithelial neoplasia with TG4001 targeted immunotherapy. Am J Obstet Gynecol 2011; 204:169 e1-8; PMID:21284968; http://dx.doi.org/10.1016/j.ajog.2010.09.020
  • 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; PMID:17146630; http://dx.doi.org/10.1007/s00262-006-0247-2
  • Roden RB, Ling M, Wu TC. Vaccination to prevent and treat cervical cancer. Hum Pathol 2004; 35:971-82; PMID:15297964; http://dx.doi.org/10.1016/j.humpath.2004.04.007
  • Nishimoto KP, Laust AK, Wang K, Kamrud KI, Hubby B, Smith JF, Nelson EL. Restricted and selective tropism of a Venezuelan equine encephalitis virus-derived replicon vector for human dendritic cells. Viral Immunol 2007; 20:88-104; PMID:17425424; http://dx.doi.org/10.1089/vim.2006.0090
  • Daemen T, Riezebos-Brilman A, Regts J, Dontje B, van der Zee A, Wilschut J. Superior therapeutic efficacy of alphavirus-mediated immunization against human papilloma virus type 16 antigens in a murine tumour model: effects of the route of immunization. Antivir Ther 2004; 9:733-42; PMID:15535411
  • Riezebos-Brilman A, Regts J, Freyschmidt EJ, Dontje B, Wilschut J, Daemen T. Induction of human papilloma virus E6/E7-specific cytotoxic T-lymphocyte activity in immune-tolerant, E6/E7-transgenic mice. Gene therapy 2005; 12:1410-4; PMID:15843807; http://dx.doi.org/10.1038/sj.gt.3302536
  • 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; 22:520-7; PMID:14670335; http://dx.doi.org/10.1016/j.vaccine.2003.07.003
  • Song S, Han M, Zhang H, Wang Y, Jiang H. Full screening and accurate subtyping of HLA-A*02 alleles through group-specific amplification and mono-allelic sequencing. Cell Mol Immunol 2013; 10:490-6; PMID:23954948; http://dx.doi.org/10.1038/cmi.2013.33
  • Moniz M, Ling M, Hung CF, Wu TC. HPV DNA vaccines. Frontiers in bioscience : a journal and virtual library 2003; 8:d55-68; PMID:12456324; http://dx.doi.org/10.2741/936
  • Ohlschlager P, Pes M, Osen W, Durst M, Schneider A, Gissmann L, Kaufmann AM. An improved rearranged Human Papillomavirus Type 16 E7 DNA vaccine candidate (HPV-16 E7SH) induces an E7 wildtype-specific T cell response. Vaccine 2006; 24:2880-93; PMID:16472545; http://dx.doi.org/10.1016/j.vaccine.2005.12.061
  • Brinkman JA, Xu X, Kast WM. The efficacy of a DNA vaccine containing inserted and replicated regions of the E7 gene for treatment of HPV-16 induced tumors. Vaccine 2007; 25:3437-44; PMID:17241713; http://dx.doi.org/10.1016/j.vaccine.2006.12.045
  • Peng S, Ji H, Trimble C, He L, Tsai YC, Yeatermeyer J, Boyd DA, Hung CF, Wu TC. Development of a DNA vaccine targeting human papillomavirus type 16 oncoprotein E6. J Virol 2004; 78:8468-76; PMID:15280455; http://dx.doi.org/10.1128/JVI.78.16.8468-8476.2004
  • 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; 15:361-7; PMID:19118066; http://dx.doi.org/10.1158/1078-0432.CCR-08-1725
  • Garcia F, Petry KU, Muderspach L, Gold MA, Braly P, Crum CP, Magill M, Silverman M, Urban RG, Hedley ML, et al. ZYC101a for treatment of high-grade cervical intraepithelial neoplasia: a randomized controlled trial. Obstet Gynecol 2004; 103:317-26; PMID:14754702; http://dx.doi.org/10.1097/01.AOG.0000110246.93627.17
  • Bagarazzi ML, Yan J, Morrow MP, Shen X, Parker RL, Lee JC, Giffear M, Pankhong P, Khan AS, Broderick KE, et al. Immunotherapy against HPV16/18 generates potent TH1 and cytotoxic cellular immune responses. Sci Transl Med 2012; 4:155ra38; http://dx.doi.org/10.1126/scitranslmed.3004414
  • Kim TJ, Jin HT, Hur SY, Yang HG, Seo YB, Hong SR, Lee CW, Kim S, Woo JW, Park KS, et al. Clearance of persistent HPV infection and cervical lesion by therapeutic DNA vaccine in CIN3 patients. Nat Commun 2014; 5:5317; PMID:25354725; http://dx.doi.org/10.1038/ncomms6317
  • Trimble CL, Morrow MP, Kraynyak KA, Shen X, Dallas M, Yan J, Edwards L, Parker RL, Denny L, Giffear M, et al. Safety, efficacy, and immunogenicity of VGX-3100, a therapeutic synthetic DNA vaccine targeting human papillomavirus 16 and 18 E6 and E7 proteins for cervical intraepithelial neoplasia 2/3: a randomised, double-blind, placebo-controlled phase 2b trial. Lancet 2015; 386:2078-88; PMID:26386540; http://dx.doi.org/10.1016/S0140-6736(15)00239-1
  • Yang B, Yang A, Peng S, Pang X, Roden RB, Wu TC, Hung CF. Co-administration with DNA encoding papillomavirus capsid proteins enhances the antitumor effects generated by therapeutic HPV DNA vaccination. Cell Biosci 2015; 5:35; PMID:26113972; http://dx.doi.org/10.1186/s13578-015-0025-y
  • Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer 2012; 12:265-77; PMID:22437871; http://dx.doi.org/10.1038/nrc3258
  • 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 pilot study in 15 individual patients. J Cancer Res Clin Oncol 2003; 129:521-30; PMID:12898233; http://dx.doi.org/10.1007/s00432-003-0463-5
  • Santin AD, Bellone S, Palmieri M, Ravaggi A, Romani C, Tassi R, Roman JJ, Burnett A, Pecorelli S, Cannon MJ. HPV16/18 E7-pulsed dendritic cell vaccination in cervical cancer patients with recurrent disease refractory to standard treatment modalities. Gynecol Oncol 2006; 100:469-78; PMID:16249018; http://dx.doi.org/10.1016/j.ygyno.2005.09.040
  • Kang TH, Lee JH, Bae HC, Noh KH, Kim JH, Song CK, Shin BC, Hung CF, Wu TC, Park JS, et al. Enhancement of dendritic cell-based vaccine potency by targeting antigen to endosomal/lysosomal compartments. Immunol Lett 2006; 106:126-34; PMID:16844231; http://dx.doi.org/10.1016/j.imlet.2006.05.004
  • Tillman BW, Hayes TL, DeGruijl TD, Douglas JT, Curiel DT. Adenoviral vectors targeted to CD40 enhance the efficacy of dendritic cell-based vaccination against human papillomavirus 16-induced tumor cells in a murine model. Cancer Research 2000; 60:5456-63; PMID:11034088
  • Zhou ZX, Li D, Guan SS, Zhao C, Li ZL, Zeng Y. Immunotherapeutic Effects of Dendritic Cells Pulsed with a Coden-optimized HPV 16 E6 and E7 Fusion Gene in Vivo and in Vitro. Asian Pac J Cancer Prev 2015; 16:3843-7; PMID:25987047; http://dx.doi.org/10.7314/APJCP.2015.16.9.3843
  • Houot R, Schultz LM, Marabelle A, Kohrt H. T-cell-based Immunotherapy: Adoptive Cell Transfer and Checkpoint Inhibition. Cancer Immunol Res 2015; 3:1115-22; PMID:26438444; http://dx.doi.org/10.1158/2326-6066.CIR-15-0190
  • Stevanovic S, Draper LM, Langhan MM, Campbell TE, Kwong ML, Wunderlich JR, Dudley ME, Yang JC, Sherry RM, Kammula US, et al. Complete regression of metastatic cervical cancer after treatment with human papillomavirus-targeted tumor-infiltrating T cells. J Clin Oncol 2015; 33:1543-50; PMID:25823737; http://dx.doi.org/10.1200/JCO.2014.58.9093
  • Scholten KB, Schreurs MW, Ruizendaal JJ, Kueter EW, Kramer D, Veenbergen S, Meijer CJ, Hooijberg E. Preservation and redirection of HPV16E7-specific T cell receptors for immunotherapy of cervical cancer. Clinical Immunology 2005; 114:119-29; PMID:15639645; http://dx.doi.org/10.1016/j.clim.2004.11.005
  • Draper LM, Kwong ML, Gros A, Stevanovic S, Tran E, Kerkar S, Raffeld M, Rosenberg SA, Hinrichs CS. Targeting of HPV-16+ Epithelial Cancer Cells by TCR Gene Engineered T Cells Directed against E6. Clin Cancer Res 2015; 21:4431-9; PMID:26429982; http://dx.doi.org/10.1158/1078-0432.CCR-14-3341
  • Luxton JC, Rowe AJ, Cridland JC, Coletart T, Wilson P, Shepherd PS. Proliferative T cell responses to the human papillomavirus type 16 E7 protein in women with cervical dysplasia and cervical carcinoma and in healthy individuals. J Gen Virol 1996; 77 (Pt 7):1585-93; PMID:8758003; http://dx.doi.org/10.1099/0022-1317-77-7-1585
  • Einstein MH, Kadish AS, Burk RD, Kim MY, Wadler S, Streicher H, Goldberg GL, Runowicz CD. Heat shock fusion protein-based immunotherapy for treatment of cervical intraepithelial neoplasia III. Gynecol Oncol 2007; 106:453-60; PMID:17586030; http://dx.doi.org/10.1016/j.ygyno.2007.04.038
  • Petit RG, Mehta A, Jain M, Gupta S, Nagarkar R, Kumar V, Premkumar S, Neve R, John S, Basu P. ADXS11-001 immunotherapy targeting HPV-E7: final results from a Phase II study in Indian women with recurrent cervical cancer. Journal for ImmunoTherapy of Cancer 2014; 2 (Suppl 3):92; http://dx.doi.org/10.1186/2051-1426-2-S3-P92
  • Berezhnaya NM. Interaction between tumor and immune system: the role of tumor cell biology. Exp Oncol 2010; 32:159-66; PMID:21403611
  • Sharpe AH, Freeman GJ. The B7-CD28 superfamily. Nat Rev Immunol 2002; 2:116-26; PMID:11910893; http://dx.doi.org/10.1038/nri727
  • Agata Y, Kawasaki A, Nishimura H, Ishida Y, Tsubata T, Yagita H, Honjo T. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol 1996; 8:765-72; PMID:8671665; http://dx.doi.org/10.1093/intimm/8.5.765
  • Callahan MK, Postow MA, Wolchok JD. CTLA-4 and PD-1 Pathway Blockade: Combinations in the Clinic. Front Oncol 2014; 4:385; PMID:25642417
  • Sansom DM, Walker LS. The role of CD28 and cytotoxic T-lymphocyte antigen-4 (CTLA-4) in regulatory T-cell biology. Immunol Rev 2006; 212:131-48; PMID:16903911; http://dx.doi.org/10.1111/j.0105-2896.2006.00419.x
  • Teft WA, Kirchhof MG, Madrenas J. A molecular perspective of CTLA-4 function. Annu Rev Immunol 2006; 24:65-97; PMID:16551244; http://dx.doi.org/10.1146/annurev.immunol.24.021605.090535
  • Krummel MF, Allison JP. CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J Exp Med 1995; 182:459-65; PMID:7543139; http://dx.doi.org/10.1084/jem.182.2.459
  • Ukpo OC, Thorstad WL, Lewis JS, Jr. B7-H1 expression model for immune evasion in human papillomavirus-related oropharyngeal squamous cell carcinoma. Head Neck Pathol 2013; 7:113-21; PMID:23179191; http://dx.doi.org/10.1007/s12105-012-0406-z
  • Lyford-Pike S, Peng S, Young GD, Taube JM, Westra WH, Akpeng B, Bruno TC, Richmon JD, Wang H, Bishop JA, et al. Evidence for a role of the PD-1:PD-L1 pathway in immune resistance of HPV-associated head and neck squamous cell carcinoma. Cancer research 2013; 73:1733-41; PMID:23288508; http://dx.doi.org/10.1158/0008-5472.CAN-12-2384
  • Lu B, Finn OJ. T-cell death and cancer immune tolerance. Cell Death Differ 2008; 15:70-9; PMID:18007660; http://dx.doi.org/10.1038/sj.cdd.4402274
  • Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, Fitz LJ, Malenkovich N, Okazaki T, Byrne MC, et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med 2000; 192:1027-34; PMID:11015443; http://dx.doi.org/10.1084/jem.192.7.1027
  • Eppihimer MJ, Gunn J, Freeman GJ, Greenfield EA, Chernova T, Erickson J, Leonard JP. Expression and regulation of the PD-L1 immunoinhibitory molecule on microvascular endothelial cells. Microcirculation 2002; 9:133-45; PMID:11932780; http://dx.doi.org/10.1080/713774061
  • La-Beck NM, Jean GW, Huynh C, Alzghari SK, Lowe DB. Immune Checkpoint Inhibitors: New Insights and Current Place in Cancer Therapy. Pharmacotherapy 2015; 35:963-76; PMID:26497482; http://dx.doi.org/10.1002/phar.1643
  • Peggs KS, Quezada SA, Korman AJ, Allison JP. Principles and use of anti-CTLA4 antibody in human cancer immunotherapy. Curr Opin Immunol 2006; 18:206-13; PMID:16464564; http://dx.doi.org/10.1016/j.coi.2006.01.011
  • Blank C, Mackensen A. Contribution of the PD-L1/PD-1 pathway to T-cell exhaustion: an update on implications for chronic infections and tumor evasion. Cancer Immunol Immunother 2007; 56:739-45; PMID:17195077; http://dx.doi.org/10.1007/s00262-006-0272-1
  • Woo SR, Turnis ME, Goldberg MV, Bankoti J, Selby M, Nirschl CJ, Bettini ML, Gravano DM, Vogel P, Liu CL, et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer research 2012; 72:917-27; PMID:22186141; http://dx.doi.org/10.1158/0008-5472.CAN-11-1620
  • Sakuishi K, Apetoh L, Sullivan JM, Blazar BR, Kuchroo VK, Anderson AC. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med 2010; 207:2187-94; PMID:20819927; http://dx.doi.org/10.1084/jem.20100643
  • Fourcade J, Sun Z, Benallaoua M, Guillaume P, Luescher IF, Sander C, Kirkwood JM, Kuchroo V, Zarour HM. Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J Exp Med 2010; 207:2175-86; PMID:20819923; http://dx.doi.org/10.1084/jem.20100637
  • Johnston RJ, Comps-Agrar L, Hackney J, Yu X, Huseni M, Yang Y, Park S, Javinal V, Chiu H, Irving B, et al. The immunoreceptor TIGIT regulates antitumor and antiviral CD8(+) T cell effector function. Cancer Cell 2014; 26:923-37; PMID:25465800; http://dx.doi.org/10.1016/j.ccell.2014.10.018
  • Derre L, Rivals JP, Jandus C, Pastor S, Rimoldi D, Romero P, Michielin O, Olive D, Speiser DE. BTLA mediates inhibition of human tumor-specific CD8+ T cells that can be partially reversed by vaccination. J Clin Invest 2010; 120:157-67; PMID:20038811; http://dx.doi.org/10.1172/JCI40070
  • Flacher V, Bouschbacher M, Verronese E, Massacrier C, Sisirak V, Berthier-Vergnes O, de Saint-Vis B, Caux C, Dezutter-Dambuyant C, Lebecque S, et al. Human Langerhans cells express a specific TLR profile and differentially respond to viruses and Gram-positive bacteria. J Immunol 2006; 177:7959-67; PMID:17114468; http://dx.doi.org/10.4049/jimmunol.177.11.7959
  • Merad M, Ginhoux F, Collin M. Origin, homeostasis and function of Langerhans cells and other langerin-expressing dendritic cells. Nat Rev Immunol 2008; 8:935-47; PMID:19029989; http://dx.doi.org/10.1038/nri2455
  • Domingos-Pereira S, Decrausaz L, Derre L, Bobst M, Romero P, Schiller JT, Jichlinski P, Nardelli-Haefliger D. Intravaginal TLR agonists increase local vaccine-specific CD8 T cells and human papillomavirus-associated genital-tumor regression in mice. Mucosal Immunol 2013; 6:393-404; PMID:22968420; http://dx.doi.org/10.1038/mi.2012.83
  • Da Silva DM, Woodham AW, Skeate JG, Rijkee LK, Taylor JR, Brand HE, Muderspach LI, Roman LD, Yessaian AA, Pham HQ, et al. Langerhans cells from women with cervical precancerous lesions become functionally responsive against human papillomavirus after activation with stabilized Poly-I:C. Clinical Immunology 2015; 161:197-208; PMID:26360252; http://dx.doi.org/10.1016/j.clim.2015.09.003
  • Da Silva DM, Woodham AW, Rijkee LK, Skeate JG, Taylor JR, Koopman ME, Brand HE, Wong MK, McKee GM, Salazar AM, et al. Human papillomavirus-exposed Langerhans cells are activated by stabilized Poly-I:C. Papillomavirus Research 2015; 1:12-21; PMID:26665182
  • Fahey LM, Raff AB, Da Silva DM, Kast WM. Reversal of human papillomavirus-specific T cell immune suppression through TLR agonist treatment of Langerhans cells exposed to human papillomavirus type 16. J Immunol 2009; 182:2919-28; PMID:19234187; http://dx.doi.org/10.4049/jimmunol.0803645
  • Da Silva DM, Woodham AW, Naylor PH, Egan JE, L. BN, Kast WM. Immunostimulatory activity of the cytokine-based biologic IRX-2 on human papillomavirus-exposed Langerhans cells Journal of Interferon and Cytokine Research In Press.
  • Kanodia S, Fahey LM, Kast WM. Mechanisms used by human papillomaviruses to escape the host immune response. Curr Cancer Drug Targets 2007; 7:79-89; PMID:17305480; http://dx.doi.org/10.2174/156800907780006869
  • Ashrafi GH, Haghshenas M, Marchetti B, Campo MS. E5 protein of human papillomavirus 16 downregulates HLA class I and interacts with the heavy chain via its first hydrophobic domain. Int J Cancer 2006; 119:2105-12; PMID:16823848; http://dx.doi.org/10.1002/ijc.22089
  • Fahey LM, Raff AB, Da Silva DM, Kast WM. A major role for the minor capsid protein of human papillomavirus type 16 in immune escape. J Immunol 2009; 183:6151-6; PMID:19864613; http://dx.doi.org/10.4049/jimmunol.0902145
  • Da Silva DM, Movius CA, Raff AB, Brand HE, Skeate JG, Wong MK, Kast WM. Suppression of Langerhans cell activation is conserved amongst human papillomavirus alpha and beta genotypes, but not a micro genotype. Virology 2014; 452-453:279-86; PMID:24606705; http://dx.doi.org/10.1016/j.virol.2014.01.031
  • Woodham AW, Raff AB, Raff LM, Da Silva DM, Yan L, Skeate JG, Wong MK, Lin YG, Kast WM. Inhibition of Langerhans Cell Maturation by Human Papillomavirus Type 16: A Novel Role for the Annexin A2 Heterotetramer in Immune Suppression. J Immunol 2014; 192(10):4748-57; PMID:24719459
  • Trimble CL, Clark RA, Thoburn C, Hanson NC, Tassello J, Frosina D, Kos F, Teague J, Jiang Y, Barat NC, et al. Human papillomavirus 16-associated cervical intraepithelial neoplasia in humans excludes CD8 T cells from dysplastic epithelium. J Immunol 2010; 185:7107-14; PMID:21037100; http://dx.doi.org/10.4049/jimmunol.1002756
  • Yang W, Song Y, Lu YL, Sun JZ, Wang HW. Increased expression of programmed death (PD)-1 and its ligand PD-L1 correlates with impaired cell-mediated immunity in high-risk human papillomavirus-related cervical intraepithelial neoplasia. Immunology 2013; 139:513-22; PMID:23521696; http://dx.doi.org/10.1111/imm.12101
  • Bais AG, Beckmann I, Lindemans J, Ewing PC, Meijer CJ, Snijders PJ, Helmerhorst TJ. A shift to a peripheral Th2-type cytokine pattern during the carcinogenesis of cervical cancer becomes manifest in CIN III lesions. J Clin Pathol 2005; 58:1096-100; PMID:16189158; http://dx.doi.org/10.1136/jcp.2004.025072
  • Peghini BC, Abdalla DR, Barcelos AC, Teodoro L, Murta EF, Michelin MA. Local cytokine profiles of patients with cervical intraepithelial and invasive neoplasia. Hum Immunol 2012; 73:920-6; PMID:22749886; http://dx.doi.org/10.1016/j.humimm.2012.06.003
  • Scott ME, Shvetsov YB, Thompson PJ, Hernandez BY, Zhu X, Wilkens LR, Killeen J, Vo DD, Moscicki AB, Goodman MT. Cervical cytokines and clearance of incident human papillomavirus infection: Hawaii HPV cohort study. Int J Cancer 2013; 133:1187-96; PMID:23436563; http://dx.doi.org/10.1002/ijc.28119
  • Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, Evdemon-Hogan M, Conejo-Garcia JR, Zhang L, Burow M, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med 2004; 10:942-9; PMID:15322536; http://dx.doi.org/10.1038/nm1093
  • Lukesova E, Boucek J, Rotnaglova E, Salakova M, Koslabova E, Grega M, Eckschlager T, Rihova B, Prochazka B, Klozar J, et al. High level of Tregs is a positive prognostic marker in patients with HPV-positive oral and oropharyngeal squamous cell carcinomas. Biomed Res Int 2014; 2014:303929; PMID:24864233; http://dx.doi.org/10.1155/2014/303929
  • Nagaraj S, Gupta K, Pisarev V, Kinarsky L, Sherman S, Kang L, Herber DL, Schneck J, Gabrilovich DI. Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer. Nat Med 2007; 13:828-35; PMID:17603493; http://dx.doi.org/10.1038/nm1609
  • Stone SC, Rossetti RA, Lima AM, Lepique AP. HPV associated tumor cells control tumor microenvironment and leukocytosis in experimental models. Immun Inflamm Dis 2014; 2:63-75; PMID:25400927; http://dx.doi.org/10.1002/iid3.21
  • Yue FY, Dummer R, Geertsen R, Hofbauer G, Laine E, Manolio S, Burg G. Interleukin-10 is a growth factor for human melanoma cells and down-regulates HLA class-I, HLA class-II and ICAM-1 molecules. Int J Cancer 1997; 71:630-7; PMID:9178819; http://dx.doi.org/10.1002/(SICI)1097-0215(19970516)71:4%3c630::AID-IJC20%3e3.0.CO;2-E
  • Bolpetti A, Silva JS, Villa LL, Lepique AP. Interleukin-10 production by tumor infiltrating macrophages plays a role in Human Papillomavirus 16 tumor growth. BMC Immunol 2010; 11:27; PMID:20525400; http://dx.doi.org/10.1186/1471-2172-11-27
  • Alcocer-Gonzalez JM, Berumen J, Tamez-Guerra R, Bermudez-Morales V, Peralta-Zaragoza O, Hernandez-Pando R, Moreno J, Gariglio P, Madrid-Marina V. In vivo expression of immunosuppressive cytokines in human papillomavirus-transformed cervical cancer cells. Viral Immunol 2006; 19:481-91; PMID:16987066; http://dx.doi.org/10.1089/vim.2006.19.481
  • Steele JC, Mann CH, Rookes S, Rollason T, Murphy D, Freeth MG, Gallimore PH, Roberts S. T-cell responses to human papillomavirus type 16 among women with different grades of cervical neoplasia. Br J Cancer 2005; 93:248-59; PMID:15986031; http://dx.doi.org/10.1038/sj.bjc.6602679
  • Trimble CL, Peng S, Thoburn C, Kos F, Wu TC. Naturally occurring systemic immune responses to HPV antigens do not predict regression of CIN2/3. Cancer Immunol Immunother 2010; 59:799-803; PMID:20012604; http://dx.doi.org/10.1007/s00262-009-0806-4
  • Stevanovic S, Draper LM, Langhan MM, Campbell TE, Kwong ML, Wunderlich JR, Dudley ME, Yang JC, Sherry RM, Kammula US, et al. Complete regression of metastatic cervical cancer after treatment with human papillomavirus-targeted tumor-infiltrating T cells. J Clin Oncol 2015; 33:1543-50; PMID:25823737; http://dx.doi.org/10.1200/JCO.2014.58.9093
  • Khallouf H, Grabowska AK, Riemer AB. Therapeutic Vaccine Strategies against Human Papillomavirus. Vaccines (Basel) 2014; 2:422-62; PMID:26344626; http://dx.doi.org/10.3390/vaccines2020422
  • Corona Gutierrez CM, Tinoco A, Navarro T, Contreras ML, Cortes RR, Calzado P, Reyes L, Posternak R, Morosoli G, Verde ML, et al. 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; 15:421-31; PMID:15144573; http://dx.doi.org/10.1089/10430340460745757

Reprints and Corporate Permissions

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

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

Academic Permissions

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

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

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