You are on page 1of 15

Cancer Letters 471 (2020) 88–102

Contents lists available at ScienceDirect

Cancer Letters
journal homepage: www.elsevier.com/locate/canlet

Mini-review

Human papillomavirus vaccine against cervical cancer: Opportunity and T


challenge
Renjie Wanga,1, Wei Panb,1, Lei Jina,1, Weiming Huanga, Yuehan Lia, Di Wua, Chun Gaoa,
Ding Maa, Shujie Liaoa,∗
a
Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College of HUST, Wuhan, Hubei, 430030, PR China
b
School of Economics and Management, Wuhan University, Wuhan, Hubei, 430072, PR China

A R T I C LE I N FO A B S T R A C T

Keywords: Cervical cancer is one of the most common cancers threatening women's health, and the persistent infection of
Human papillomavirus high-risk human papillomavirus (HPV) is closely related to the pathogenesis of cervical cancer and many other
Cervical cancer cancers. The carcinogenesis is a complex process from precancerous lesion to cancer, which provides an ex-
Prophylactic vaccine cellent window for clinical prevention, diagnosis, and treatment. However, despite the various preventions and
Therapeutic vaccine
treatments such as HPV screening, prophylactic HPV vaccines, surgery, radiotherapy, and chemotherapy, the
disease burden remains heavy worldwide. Currently, three types of prophylactic vaccines, quadrivalent HPV
vaccine, bivalent HPV vaccine, and a new nonavalent HPV vaccine, are commercially available. Although these
vaccines are effective in protecting against 90% of HPV infection, they provide limited benefits to eliminate pre-
existing infections. Therefore, new progress has been made in the development of therapeutic vaccines.
Therapeutic vaccines differ from prophylactic vaccines in that they aim to stimulate cell-mediated immunity and
kill the infected cells rather than neutralizing antibodies. This review aims at systematically covering the pro-
gress, current status and future prospects of various vaccines in development for the prevention and treatment of
HPV-associated lesions and cancers and laying foundations for the development of the new original vaccine.

1. Introduction infection is limited to the basal cells of the epithelium. HPV lesions arise
from the unchecked cell proliferation and mutations and lead to cancer
Cervical cancer is one of the most common cancers threatening ultimately [5,6]. Due to the rapid immune clearance, most HPV infec-
women's health and is the fourth most common cancer in women tions do not cause symptoms and resolve spontaneously within 1 or 2
worldwide. According to the World Health Organization (WHO), cer- years; while those women who develop cervical cancer can test positive
vical cancer is the second most common cancer in women living in less for a high-risk HPV genotype 3–5 years prior to cancer [6]. Persistent
developed regions with an estimated 570000 cases and 311000 deaths high-risk HPV infection has been shown to be involved in the increased
in 2018, and more than 85% of these deaths occurring in less developed risk of high-grade cervical intraepithelial neoplasia (CIN) and cancer
regions [1]. Currently, the study clarifies that cervical cancer is caused [7]. The two HPV types, HPV16 and HPV18, have been identified as the
by sexually transmitted infection with human papillomavirus (HPV), most prevalent types associated with cervical cancer and are accoun-
and 84% of HPV-related cancer lesions are cervical cancer [2]. HPV table for 70% of cervical cancers and precancerous cervical lesions [1].
infection is the most prevalent sexually transmitted disease, which re- Moreover, high-risk HPV types are responsible for anogenital and or-
sults in over 14000000 individuals annually and 80% of sexually active opharyngeal cancer [8]. Immunosuppression is also a high-risk factor
individuals in their lifetime to be infected from HPV [3]. for persistent infections with HPV and HPV-associated diseases [9].
HPV is a double-stranded circular DNA virus with a genome of ap- However, studies have found that some patients with cervical cancer
proximately 8000 base pairs. The genome encodes six early regulatory are tested HPV-negative regardless of the detection methods. The rea-
proteins(E1, E2, E4, E5, E6, and E7) and two late structural proteins(L1 sons lie in that testing results include the true negative and false ne-
and L2) [4]. There are more than 100 types of HPV, of which at least 14 gative [10]. Patients with HPV-negative cervical cancer are easily
high-risk HPV types have been defined as carcinogenic [1]. HPV missed diagnosis in routine screening, thus missing the opportunity for


Corresponding author.
E-mail address: sjliao@tjh.tjmu.edu.cn (S. Liao).
1
Renjie Wang, Wei Pan and Lei Jin contributed equally to this work.

https://doi.org/10.1016/j.canlet.2019.11.039
Received 29 September 2019; Received in revised form 28 November 2019; Accepted 30 November 2019
0304-3835/ © 2019 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/BY-NC-ND/4.0/).
R. Wang, et al. Cancer Letters 471 (2020) 88–102

early intervention and treatment. Though the proportion of these pa- family Papillomaviridae, among which 5 genera (alphapapilloma-
tients is small, more attention needs to be paid. viruses, betapapillomaviruses, gammapapillomaviruses, mupapilloma-
The comprehensive cervical cancer control is composed of primary viruses, and nupapillomaviruses) are associated with HPVs. The phy-
prevention (vaccination against HPV), secondary prevention (screening logenetic classification of HPVs is characterized by genotype and based
and treatment of precancerous lesions), tertiary prevention (diagnosis on the L1 ORF sequence homology, which is the most conserved ORF. A
and treatment of invasive cervical cancer) and palliative care. novel HPV type is defined as differences over 10% on L1 ORF sequence
Screening of precancerous and cancerous lesions and the primary pre- from other known genotypes, while a subtype is defined as differences
vention through vaccination allow reducing the incidence of HPV-re- between 2 and 10%. The HPV types are numbered based on the date of
lated disease and death effectively [3]. Three types of prophylactic discovery [16,17]. Currently, more than 200 HPV types have been
vaccines, quadrivalent HPV vaccine, bivalent HPV vaccine, and the identified by the International HPV Reference Center [18], and the
nonavalent HPV vaccine have been approved for use in many countries. number has been a rapid increase with the application of new mole-
These three vaccines prevent 70%–90% of the HPV-related cancers and cular tools such as next-generation sequencing during the past decade
reveal safety and effectiveness in randomized trials and post-marketing [19].
surveillance. The quadrivalent and nonavalent HPV vaccines also in- HPVs induces different lesions and they can result in overlapping
clude the protection against HPV6 and HPV11, which cause anogenital outcomes. The betapapillomaviruses and gammapapillomaviruses are
warts. mainly cutaneous HPV types [20]. Based on epidemiological data, the
Currently, at least 118 million women have received one dose of International Agency for Research on Cancer (IARC) designates a sub-
HPV vaccine worldwide [11]. These vaccines are effective at protecting group of alphapapillomaviruses (HPV16, HPV18, HPV31, HPV33,
against HPV infection and neoplastic diseases; however, they are pro- HPV35, HPV39, HPV45, HPV51, HPV52, HPV56, HPV58, and HPV59)
phylactic vaccines and provide no therapeutic benefit and limited as group 1 (carcinogenic), which is considered as high-risk HPV types.
benefits to eliminating pre-existing infections. Moreover, the impact of The IARC group 2A (probably carcinogenic) and group 2B (possibly
vaccination may not reduce the cancer incidence apparently because of carcinogenic) include several HPV types (such as HPV26, HPV53,
the long-time required for the development of precancerous lesions HPV66, HPV67, HPV68, HPV70, HPV73, and HPV82). The high-risk
[12]. Therefore, the ability to stimulate cell-mediated immune response HPV types are the etiological agents of anogenital and oropharyngeal
and kill the infected cells rather than neutralizing antibodies (nAbs) has cancer, such as cervix, vagina, vulva, penis, anus, and head and neck
led to increasing interest in therapeutic vaccines. These vaccines could cancers [21]. Of new cancer cases in 2012, 15.4% of cancers worldwide
have a significant impact on the morbidity and mortality related to are caused by carcinogenic infections [22]. 96% of cervical cancers are
HPV. Different classes of therapeutic vaccines have been developed. attributed to the 13 HPV types in groups 1 and 2A (HPV68) [23].
The safety and efficacy of therapeutic vaccines are the main concerns However, little studies are indicating that HPV68 is a high-risk HPV.
on their development and utilization. Additional alphapapillomaviruses (HPV26, HPV30, HPV34, HPV53,
The extensive studies of virology and epidemiology have provided HPV66, HPV67, HPV69, HPV70, HPV73, HPV82, HPV85, and HPV97)
an understanding of HPV and carcinogenesis and revealed opportu- are associated with infrequent incidences of cervical cancer and are
nities to prevent and treat HPV-related diseases. The immunotherapy referred to as group 2B, which is hard to assess the carcinogenicity
for cervical cancer and other diseases related to HPV infection has [10]. Moreover, the alphapapillomaviruses also includes low-risk HPV
become a research hotspot. In this review, we summarized the recent types that have been linked to benign lesions, such as HPV 6 and HPV
advances in HPV-related cancer biology and the viral action in carci- 11, which are responsible for approximately 90% of anogenital wart
nogenesis. Then we discussed the progress, current status, and pro- cases [24].
spects of various vaccines in development for the prevention and
treatment. This review focused on the development and safety of cur- 2.3. HPV pathogenesis and cervical carcinogenesis
rent HPV prophylactic vaccines and the prospects for the landscape of
therapeutic vaccines. Located between the vagina and the uterus, the cervix is char-
acterized by the simple columnar secretory epithelium. In contrast, the
2. HPV and cervical cancer vaginal cavity is the stratified non-keratinizing squamous epithelium.
The squamocolumnar junction is particularly vulnerable to transfor-
2.1. HPV genome and structure mation by high-risk HPV and is the area in which over 90% of lower
genital tract malignancies initiate [25,26]. As there are two kinds of
The HPV belongs to the Papillomaviridae family of small, none- epithelial cells (squamous and glandular cells) in the transition zone,
nveloped, double-stranded DNA viruses, able to target epithelial cells of cervical cancers can occur in two different forms. Mucosa and skin are
skin, oral and anogenital mucosa [13]. The viral genome consists of the most common areas of infection for HPV [27]. Women, especially
three regions:(1) early (E) region, containing several open reading newly sexually active adolescent and young adult women, may acquire
frames (ORFs), which encode the replication proteins (E1, E2, and E4) HPV via sexual exposures with an infected partner. HPV infects the
and the oncoproteins (E5, E6, and E7); (2) late (L) region, containing basal epithelial cells through the epithelial abrasion [28] and subse-
the late genes (L1 and L2), which encode major (L1) and minor (L2) quently induces cervical dysplasia and CIN, which typically develops
virus capsid proteins. The capsid of HPV is 50–60 nm in diameter and a into cervical cancer due to the persistent infection of high-risk HPV. The
T = 7 icosahedral formation consisting of 72 L1 protein pentamers carcinogenic progression to cervical cancer is a slow process and can be
(360 copies). The L2 protein is located in the center of the pentamers in divided into several stages. In the first stage, HPV infection can be
an L2:L1 ratio of 1:5–1:10 [14]; (3) upstream regulatory region (URR), detected in clinical, but the majority is usually transient and becomes
also known as non-coding region (NCR) or long control region (LCR), undetectable without intervention within 1–2 years due to the viral
which is located between the L1 and E6 ORFs and contains the reg- clearance. After infection, the early HPV genes (E1, E2, E4, E5, E6, and
ulatory elements associated with the viral DNA replication and tran- E7) are expressed in the productive lifecycle. In the upper layers of
scription [13]. epithelium, the viral genome is replicated, the late HPV genes (L1 and
L2) and E4 are expressed, and progeny viral particles are assembled.
2.2. HPV classification and oncogenic genotypes The shed virions repeat the virus life cycle. Some HPV infections persist
beyond 1 year and thus increasing the risk of progression to high-grade
According to the PV study group within the International squamous intraepithelial lesion (CIN2/3) or potentially invasive carci-
Committee on Taxonomy of Viruses [15], there are 53 genera in the noma if untreated [29]. The development of cervical cancer is

89
R. Wang, et al. Cancer Letters 471 (2020) 88–102

dependent not only on the negative regulation of cell-cycle control and virus uptake and internalization via endosomal vesicles into the target
the accumulation of genetic damage by viral oncoproteins, but also the cells [45]. Once HPV has reached the cell, L2 protein is necessary for
immune evasion [30]. infection as the endosomal virus engages the retromer and travels to-
wards the nucleus [46,47]. The L2 protein chaperones the viral genome
2.3.1. HPV infection and immune response to promyelocytic leukemia nuclear bodies for efficient viral transcrip-
Most natural HPV infections are restricted to the intraepithelial tion [48,49].
layer of the mucosa and do not develop into cancer. Approximately As the HPV does not express polymerases and is unable to self-re-
90% of patients with HPV infection present an innate and humoral plicate, early viral proteins E1 and E2 are expressed first and regulate
immune-mediated viral clearance within a few months after viral in- the host cell life cycle into the S phase, which supplies DNA poly-
fection [31,32]. 10% of the patients have a persistent infection and an merases for viral replication [27]. Use of the host replication machinery
increased risk of cancer in about 1% of all patients [32]. The persistent to perform DNA replication generates 50–100 copies of the genome per
HPV infection is an essential event for the development of cervical cell [48]. As the cells undergo cell division, viral genomes segregate
cancer. The competition between persistence and clearance is central in between the daughter cells. The late proteins L1 and L2 are regulated in
the HPV carcinogenesis. During the progression process of precancerous a cell differentiation-dependent manner and are expressed in mature
lesion, the host immune system reveals infiltration of CD4+, CD8+ squamous cells. As the epithelial differentiation of daughter cells
lymphocytes and macrophages, an increase of proinflammatory cyto- leaving the BM and moving up, the cells perform the high copy number
kines, and induction of neutralizing antibodies [33]. However, the HPV replication (> 103 per cell) and the expression of L1 and L2 pro-
immune response against infection is slow and induces low antibody tein in the outermost layer of the epithelium, which is recognized as the
titers. The nAbs are triggered after viral infection and target only the terminally differentiated layers of the epithelium. The new virions
viral particles instead of virus-infected cells, which thus cannot cure the which are composed of the newly synthesized viral DNA are deposited
infection. Moreover, the role of macrophages and natural killer (NK) in squames and are shed [50]. Consequently, the virus life cycle is re-
cells involved in the immune response is also unclear. The main an- peated. As the immunogenic virions mainly stay in the outer sites of the
tigen-presenting cell (APC) in the epithelium, Langerhans cells, are epithelium, HPV can escape from the host immune system. Currently,
essential in the recognition of HPV infection and the induction of cel- the prophylactic vaccines targeting the capsid antigens have no ther-
lular immune response [34]. apeutic effect because the basal epithelial cells carrying HPV express
The combination of innate and adaptive immunity prevents HPV only the early genes and the L1 protein is expressed in the epithelium
infection [34]. The effector T cells targeting early viral proteins can before viral shedding. Clearance of infected cells depends on inducing
eliminate virus-infected cells. The helper T cells identifying L1 protein cell-mediated immune response [51].
can induce the nAbs, which can prevent virus transmission and re- Through the expression of the three viral oncoproteins E5, E6, and
infection of the host [35]. However, the immune response able to E7, HPV obtains the host replication factors and drives the host kera-
protect from reinfection by the same or even other HPV types is dis- tinocyte into S phase [4]. The E6 protein promotes the ubiquitin-de-
puted [36]. In a phase of HPV persistence during which the host im- pendent proteasomal degradation of apoptosis regulator protein p53
mune system fails to eliminate the virus, the expression of E6 and E7 [52] and activates MYC expression and telomerase, which lead to
protein can promote lesion progression, which typically results from the promoting cell survival and extended cell life. The E7 protein mainly
methylation of the E2 promoter and viral integration, contributing to binds to the hypophosphorylated form of tumor suppressor retino-
immune deviation [4,37]. Despite there are abundant HPV-specific T blastoma protein (pRb) and promotes the pRb phosphorylation, leading
cells in neoplastic tissue, the immune system cannot eradicate the to the movement of the cell life cycle into the S phase and subsequent
tumor, suggesting the existence of an immunosuppressive tumor mi- DNA synthesis and cell proliferation. The E6 and E7 oncoprotein modify
croenvironment [38]. The mechanisms of HPV to evade the immune the control of the cell cycle and regulate apoptosis, resulting in genomic
system include: (1) the downregulation of antigen presentation ma- instability and eventually cancer [4]. The E5 protein has been identified
chinery such as MHC class I; (2) resistance to the cytotoxic T lympho- as a channel protein that can modulate ion homeostasis, vesicle traf-
cyte (CTL)-mediated cytotoxicity; (3) aberrant expression of immune ficking, virion production, and viral genome entry [53]. It also stimu-
factors such as the downregulation of interferon, upregulation of in- lates the epidermal growth factor (EGF)-mediated cell proliferation,
terleukin (IL)-10 and transforming growth factor (TGF)-β1; (4) the at- inhibits the apoptosis induced by tumor necrosis factor ligand and
traction of immune cells that inhibit the immune response such as the CD95 ligand [54], and modulates several cellular pathways involved in
immature dendritic cells (DCs), tolerogenic DCs, T regulatory cells cell adhesion, cell motility and mitogenic signaling [55]. In addition,
(Treg), tumor-associated macrophages (TAMs), and myeloid derived the viral genome is able to integrate into the host's chromosomal DNA
suppressor cells (MDSCs) [38,39]. In addition, the overexpression of E6 at random positions, which is associated with the transition into in-
and E7 protein compromises cellular DNA repair, leading to genomic vasive carcinoma [56]. The integration may disrupt the viral genes
instability and immune escape [40]. during the linearization of the genome. For example, loss of E2, the
transcriptional repressor of E6 and E7 protein, could cause dysregulated
2.3.2. The life cycle of HPV expression of these oncoproteins and further lead to aberrant pro-
Epithelial abrasion leads to the denudation of the basement mem- liferation and malignant progression.
brane (BM) and guarantees the HPV access to the basal keratinocytes.
During the HPV infects the epithelium, the virus sequentially engages 2.4. HPV-negative cervical cancer
the host proteins and delivers the genetic information into target cells.
This multistep process starts with binding of the major capsid protein Invasive cervical cancer (ICC) can be divided into three groups:
L1 to heparin sulfate proteoglycan (HSPGs) on the BM, inducing con- squamous cervical cancers (SCC) which account for 75–90% of ICCs,
formational changes in the viral capsid [41]. Recent studies also reveal adenocarcinomas (ADC), and adenosquamous cell carcinomas (ASC)
that the L1 protein can also bind to other integrins, such as laminin 5 [57]. Studies have found that almost 100% of the SCC and 86% of the
[42]. The conformational change in the capsid exposes a conserved site ADC are HPV positive [58,59]. The prevalence of HPV among ADC
on the amino terminus of minor capsid protein L2, which is then varies between the subtypes. The usual type, intestinal, villoglandular,
cleaved by extracellular furin [43]. The cleavage reveals several con- signet-ring cell and the endometrioid ADCs have a high prevalence of
served protective epitopes of L2 protein and is critical to promote a HPV positive. While the gastric type, the mesonephric type, and the
second conformational change in viral capsid that allows the virus endometrioid ADC from the upper part of the endocervix and lower
binding to cell receptors, such as α6β4 integrin [44]. This is followed by uterine segment are mainly HPV negative [60]. In clinical, high-risk

90
R. Wang, et al. Cancer Letters 471 (2020) 88–102

HPV-negative cervical cancer is extremely rare. The reasons for HPV vaccine that targets HPV16 and HPV 18; and Gardasil 9®, a nonavalent
negative include multiple aspects: the detection methods of HPV, cer- HPV (9vHPV) vaccine that targets HPV6, HPV11, HPV16, HPV18,
vical cancers independent of high-risk HPV, cervical cancers that lose HPV31, HPV33, HPV45, HPV52, and HPV58 (Fig. 1). The 4vHPV and
HPV expression, cervical cancers that associated with non-high risk 9vHPV vaccines were licensed in 2006 and 2014, respectively, and are
HPV, and cancers that are misclassified such as uterine endometrioid produced in the Saccharomyces cerevisiae (S. cerevisiae) expression
adenocarcinoma or metastasis from other primary tumors. The HPV system with the amorphous aluminum hydroxyphosphate sulfate
testing methods and the misclassification of cancers are the common (AAHS) adjuvant. The 4vHPV vaccine is indicated for the prevention of
reasons for false-negative HPV tumors. There are three main options for genital warts, precancerous or dysplastic lesions, and cervical cancer in
cervical cancer screening: cytology, HPV testing, and cytology-HPV co- females and males aged 9–26 years [71]. The 9vHPV vaccine covers 5
testing [61]. Cytology-based screening is the most widely used with a additional HPV types and is approved for the prevention of genital
lower sensitivity that ranges between 50 and 70% compared to HPV warts, precancerous or dysplastic lesions, and cervical, vulvar, vaginal,
testing with a sensitivity of more than 90% [62]. Screening with HPV and anal cancer in females and males aged 9–45 years [72]. The 2vHPV
testing (preferably using triage markers) is still superior to cytology received first approval in 2007 is produced in the baculovirus expres-
even though the HPV-negative cancers may be missed diagnosis, and sion vector system with the AS04 adjuvant containing alumi-
the HPV genotyping cannot definitively differentiate the different steps niumhydroxide and 3-deacylated monophosphoryl lipid A (MPL) [73].
in the natural history of cancer such as a transient infection, an early HPV vaccination has the potential to prevent cervical cancer, CIN and
persistent infection, and a prevalent precancer [60,63]. Moreover, only adenocarcinoma in situ (AIS) [74,75].
a very small proportion of cervical cancers are HPV true negative [40]. These three prophylactic vaccines are the application of VLPs as-
HPV vaccination and HPV screening cannot prevent or detect these sembled from the recombinant expression of L1. It has been shown that
cancers. Therefore, the colposcopy and biopsy should be performed in the recombinant L1 protein can self-assemble into VLPs that are lack of
time for suspected clinical cases, so as to early diagnose and reduce the oncogenic viral genome and morphologically similar to the native
missed diagnosis. Whether the HPV-negative cervical cancer patients virions [41,76]. Vaccination with HPV vaccines can protect against
need an individualized treatment protocol requires further research, HPV infections and are immune to subsequent viral challenge by the
and the primary HPV screening should be continued. induction of high and durable titers of nAbs that bind to the native
virion and neutralize the virus. The immune responses induced by the
3. HPV vaccine against cervical cancer HPV vaccination are significantly stronger than those seen in natural
HPV infections. However, it has been found that these vaccines provide
3.1. The requirement for vaccine limited benefits to eliminate pre-existing infections and elicit type-re-
stricted protection. HPV vaccines need to contain a wider range of VLP
Inducing nAbs is the main basis for vaccine-induced protection. types for broad protection.
When encountering with HPV, the vaccine binds to the virus and pre-
vents it from infecting the epithelium. However, the level of antibodies
produced in natural infections is usually insufficient to prevent sub- 3.2.1. Vaccine immunogenicity
sequent reinfection. The infected cervical site lacks secondary lymphoid Evaluation of the type-specific immune response to HPV following
tissue where contains a large number of memory B cells, subsequently vaccination includes cellular and humoral assays. The cellular assays
producing antibody and neutralizing the virus before uptake [64]. Be- include the evaluation of T cell and memory B cell responses. Currently,
sides, to ensure a protective immune response throughout sexually much more emphasis has been put on humoral responses as the nAbs
active life, high and sustained levels of nAbs induced by vaccination are play a critical role in the protection against infection and subsequent
required. Therefore, an ideal HPV vaccine should provide an improved disease [77]. Serology assays include detecting antibodies (neutralizing
protective immune response, protection against all high-risk HPV types assays and binding assays) and measuring antibody binding strength
and other probably and possibly carcinogenic types. Currently, the HPV (avidity) [78]. Three main types of serological assays have been used to
vaccines are based on virus-like particles (VLPs), which are assembled evaluate the immune responses to HPV vaccines: neutralization assays
from recombinant HPV capsid proteins and are non-infectious due to [79], competitive immunoassays [80], and VLP enzyme-linked im-
the lack of viral DNA. Vaccination with VLPs can induce different types munosorbent assay (ELISA) [81]. The spectrum and potential relevance
of type-specific nAbs, which can bind to the native virus particles and of the detected antibodies among these approaches are different. Neu-
neutralize the virus by preventing uptake by the target cell. tralization assays are considered the most relevant for measuring the
The cellular immune responses also play a crucial role in the re- biological activity of the antibodies. Competitive immunoassays, such
gression of precancerous lesions and the clearance of persistent infec- as the competitive Luminex immunoassay (cLIA), estimate neutralizing
tion. It has been reported that the increased CD4: CD8 ratios in the activity by measuring competition of test serum with neutralizing
stroma, the CD4+T cell helper responses to E2, and the infiltration of monoclonal antibodies. In contrast, ELISAs detect all antibodies, re-
CD8+T cells specific for the E6 and E7 protein have been detected in gardless of neutralization ability.
the spontaneous regression of CIN [65–68]. Induction of effective cel- Ongoing research reveals that the antibody titers elicited by vacci-
lular immunity to E6, E7, and other viral antigens, may be critical to the nation with Gardasil®, Cervarix®, and Gardasil 9® can persist at least
efficacy of therapeutic HPV vaccines. The ideal HPV therapeutic vac- 9.9, 10.0, and 5.0 years, respectively, when women aged 9–15 years
cines should target these proteins to induce a robust tumor-specific T- receive a 3-dose schedule [82]. The seropositivity rates following vac-
cell type 1 and cytotoxic lymphocyte responses where CD4+ T-cells cination with three doses of 4vHPV vaccine for the total im-
secrete large amounts of cytokines such as IFN-γ and IL2 labeling in- munoglobulin (IG)G assay were 94.3, 89.4, 99.5, and 88.8%, respec-
fected and malignant cells and cytotoxic CD8+ T-cells eliminate in- tively [83]. Recent studies have shown that HPV16 and HPV18
fected cells by secreting granzyme B and perforin which lead to cell antibody levels after vaccination with the 2vHPV vaccine for at least
death [69,70]. 9.4 years are significantly higher than natural infection levels [84]. The
4vHPV vaccine elicits stable HPV16 antibody levels for at least 9 years
3.2. Prophylactic HPV vaccines [85]. Besides, the HPV16 and HPV18 antibody responses induced by
the 9vHPV vaccine are comparable to the Gardasil®. Follow vaccination
Currently, three licensed prophylactic HPV vaccines are available with 3 doses of Gardasil 9®, the HPV antibodies persisted through 5
(Table 1): Gardasil®, a quadrivalent HPV (4vHPV) vaccine that targets years with seropositivity rates ranging from 77.5% to 100% [86].
HPV6, HPV11, HPV16, and HPV18; Cervarix®, a bivalent HPV (2vHPV)

91
R. Wang, et al. Cancer Letters 471 (2020) 88–102

Table 1
Characteristics of three commercially available HPV vaccines.
Cervarix Gardasil Gardasil 9

Antigen L1 VLP of HPV-16 and 18 L1 VLP of HPV-6, 11, 16 and 18 L1 VLP of HPV-6, 11, 16, 18, 31, 33, 45, 52 and 58
Expression system Baculovirus expression vector system S. cerevisiae S. cerevisiae
Adjuvant (μg) AS04: AAHS (225) AAHS (500)
MPL (50); Aluminum hydroxide salt (500)
Indications: Females, age between 9 and 25 years Females and males, age between 9 and 26 years Females and males, age between 9 and 45 years
Manufacturer GlaxoSmithKline Biologicals Merck & Co., Inc Merck & Co., Inc

VLP: virus-like particle; S. cerevisiae: Saccharomyces cerevisiae;AS04: Adjuvant System 04; AAHS: amorphous aluminumhydroxyphosphate sulfate.

Fig. 1. A brief history of HPV and HPV


vaccine research
In the 1930s, the carcinogenic potential of
rabbit papillomaviruses in cottontail and
domestic rabbits were demonstrated for the
first time [194,195]. However, research on
the human wart virus remained low. Be-
tween 1974 and 1976, researchers began to
analyze the possible role of HPV in cervical
cancer [196,197]. In 1976, Meisels and
Fortin [198]found the appearance of Koilo-
cytes in cervical smears, which indicated the
presence of papillomavirus infection. In
1983 and 1984, HPV16 and HPV18, the
cervical-cancer-linked HPV type, were iso-
lated from cancer biopsies of the cervix, re-
spectively [199,200]. Between 1985 and
1992, the activities of HPV oncogenes in cervical cancer etiology have been determined [201–205]. In 1987, de Villiers et al. [206] conducted the first epide-
miological study of HPV infections. In 1991, the first VLPs were produced [207]. In 1999, HPV was first proposed as a necessary cause of cervical cancer development
[208]. In 2001, Harro et al. reported the safety and immunogenicity of VLP vaccines in humans [209]. In 2006, the first prophylactic HPV vaccine (Gardasil®) was
licensed. Subsequently, the bivalent HPV vaccine (Cervarix®) and nonavalent HPV vaccine (Gardasil 9®) received approval in 2007 and 2014, respectively.

3.2.2. Vaccine efficacy against cervical cancer and cross-protection subsequent disease [97]. The vaccine-mediated protection was not
Prophylactic HPV vaccines have been licensed in many countries for type-specific because of the cross-protection, such as HPV16 with
over 10 years. The introduction of HPV vaccines has shown the re- HPV31 and HPV33, and HPV18 with HPV45. These related types share
duction of HPV prevalence and HPV-related diseases, such as genital considerable amino acid sequence in the major capsid protein L1 [98].
warts, CIN and cervical cancer [3,87–89]. The implementation of HPV Therefore, 4vHPV and 2vHPV vaccines induced cross-protection against
vaccination program will result in a dramatical decrease in cervical HPV types which were not included in the vaccines, such as HPV31,
cancer rate. Recently, a meta-analysis of 4vHPV vaccines showed that HPV33, HPV45, and HPV51 [91,99]. The 2vHPV vaccine exerted
up to approximately 90% for HPV-6/11/16/18 infection and genital higher cross-protection for HPV31, HPV33, and HPV45 than the 4vHPV
warts reduced compared to unvaccinated populations. The incidence of vaccine. The 2vHPV vaccine showed 93% efficacy against CIN3 irre-
low-grade cytological cervical abnormalities (approximately 45%) and spective of the HPV type, while only 52% of CIN3is related to HPV16
high-grade histologically proven cervical abnormalities (approximately and HPV18 [92,100].
85%) continued to decline [88]. The 2vHPV vaccine was evaluated in
phase III randomized, double-blind, controlled Papilloma TRIal against 3.2.3. Vaccination schedules and strategies
Cancer In young Adults (PATRICIA). The vaccine showed 92.9% ef- The prophylactic HPV vaccines were initially licensed using a 3-
fective against CIN 2 + associated with HPV-16/18, 54.0% effective dose vaccination schedule. Recent studies have shown that 2 doses or
against CIN2+ associated with 12 non-vaccine oncogenic types, and even 1 dose of 2vHPV vaccine were highly efficacious compared to
consistent cross-protective efficacy against HPV-31/33/45/51 in the the3-dose regimen [101]. An observer-blind study confirmed that su-
woman aged 15–25years [90,91]. The trial also showed high vaccine perior HPV-16/18 antibody responses were induced in girls aged 9–14
efficacy against CIN3+ and AIS irrespective of HPV type in the lesion years who received 2 doses of 2vHPV vaccine at months 0 and 6
[92]. In another phase 3, double-blind, randomized controlled VIVIANE compared with those who received 2 doses of 4vHPV vaccine at months
study, the 2vHPV vaccine showed sustained protection against infec- 0 and 6 or 3 doses of 4vHPV vaccine at months 0, 2, and 6 [102]. The
tions, cytological abnormalities, and lesions in the woman older than 25 cost-effectiveness analysis showed that if the vaccines could provide
years [93,94]. Efficacy of the 4vHPV and 9vHPV vaccine was evaluated protection for more than 20 years, the 2-dose vaccination schedule
in a randomized, international, double-blind, phase 2b-3 study in seemed to be the most cost-effective option, while it was unknown
14215 women aged 16–26 years. Compared to the 4vHPV vaccine, the whether the duration of the 2-dose schedule can last for 20 years [103].
9vHPV vaccine represented 97.4%efficacy against high-grade cervical, Based on these studies, the World Health Organization (WHO) has now
vulvar and vaginal disease associated with HPV-31/33/45/52/58 (0.5 recommended that people aged between 9 and 15 years should take 2
and 19·0 cases per 10 000 person-years in the 9vHPV and 4vHPV doses of HPV vaccines at least six months after the first dose (Time
groups, respectively) and non-inferior antibody response to HPV-6/11/ interval between the first and second dose should be 6–15 months).
16/18 in the 9vHPV and 4vHPV vaccines. The 9vHPV vaccine provided Individuals who receive their two doses less than five months at in-
sustained efficacy up to 6 years and broader coverage and prevention tervals and are older than 14 years old or immuno-compromised re-
for approximately 90%of cervical cancers and HPV-related diseases gardless of age require a third-dose schedule [104]. Additionally, sev-
[95,96]. Overall, all three vaccines have demonstrated sufficiently high eral randomized studies of 4vHPV, 2vHPV, and 9vHPV vaccines
and durable levels of antibody response to protect against infection and demonstrated that women receiving the 2-dose regimen showed a non-

92
R. Wang, et al. Cancer Letters 471 (2020) 88–102

inferior or uncertain immune response compared with the 3-dose re- limited cross-protection and HPV genotypes in prophylactic HPV vac-
gimen [105–107]. Recent studies have shown that a single dose of cines. Currently, the 2vHPV and 4vHPV vaccines provide approxi-
4vHPV vaccine [108] or 2vHPV vaccine [109] provide protection mately 100% protection against infection with HPV16 and HPV18
against persistent HPV infection similar to the 3-dose regimen. [125]. However, the non-vaccine high-risk HPV types account for ap-
proximately 30% of cervical cancers [126]. Therefore, there is an ur-
3.2.4. Safety and limitations of prophylactic HPV vaccines gent need for vaccines that broadly target HPV types.
To date, more than 200 million doses of prophylactic HPV vaccines
have been administered worldwide [88]. Increased evidence confirms 3.2.5. Future prospects of prophylactic HPV vaccines
the vaccine safety. However, a series of safety issues have emerged, 3.2.5.1. L2-based HPV vaccines. A key feature of L1 VLPs is their high
such as pain, redness, or swelling in the arm where the shot was given, immunogenicity, even in the absence of adjuvant. Vaccination with
fever, headache or feeling tired, nausea, and muscle or joint pain. The HPV L1 VLPs provides HPV type-specific protection and cross-
Centre of Disease Control and Prevention (CDC) and FDA use three protection against certain HPV types. In contrast, the L2 protein
systems to monitor the vaccine safety: The Vaccine Adverse Event Re- cannot form VLP alone and is poorly immunogenic. The L2 protein is
porting System (VAERS), the Vaccine Safety Datalink (VSD), and the located inside the HPV capsid and therefore cannot enter the immune
Clinical Immunization Safety Assessment (CISA) Project. In 2014, the system [127]. Compared with 360 L1 in the capsid, there are only
safety of 9vHPV vaccine was evaluated in seven studies prior to ob- 12–72 L2copies per virion, and thus the interval between L2 is further,
taining the license by the FDA. These pre-licensure studies have in- which may result in poor immunogenicity of L2 in the capsid
dicated that the 9vHPV vaccine has similar safety to 4vHPV vaccine. In [128,129]. Despite these differences, vaccination with L2 using
addition, approximately 92% of the 4vHPV vaccine reports were clas- adjuvants such as alum provides long-lasting immunity in animal
sified as non-serious [110]. Serious adverse events are very rare after models, and passive-transfer studies have shown that low titers of
vaccination with HPV vaccines, and the reasons are unclear. Based on nAbs with moderate affinity were sufficient for protection [130,131].
the VAERS data [111,112] and a series of studies in Southern Italy Moreover, a variety of strategies have been applied to elicit robust
[113], Denmark [114], and Indonesia [115], the overall results of these immune responses, including displaying L2 epitopes on a carrier
studies demonstrated that no serious adverse events following im- protein, the use of peptide multimers, and binding to the fusion
munization were medically confirmed to be related to HPV vaccines. partner [14,132]. Therefore, the L2-based vaccines have the potential
The adverse events related to HPV vaccination, such as lymphadeno- for providing protection against various HPVs at low vaccination prices
pathy in 2 clinical cases [116], myasthenia gravis in a 23-year-old due to their simple form [133,134].
woman [117], were reported on individual cases. Recent evidence has
suggested that HPV infection may affect reproductive health and fer- 3.2.5.2. The second-generation prophylactic HPV vaccines. The three
tility [118]. The fetal adverse events following HPV vaccination mainly currently licensed prophylactic HPV vaccines use insect or yeast cell
relate to spontaneous miscarriage, while the vaccines do not seem to expression systems and their manufacturing processes are therefore
escalate the occurrence of miscarriage. The HPV vaccination during complex, which is the main reason for the high vaccine price. Numerous
pregnancy, peripregnancy, or immediately post-conception is not as- workers have attempted to enhance the production processes for VLP-
sociated with risks of adverse pregnancy outcomes [119–121]. The HPV based vaccines and have achieved considerable success. HPV L1 VLP-
vaccination is not recommended for pregnant women, but breast- based vaccines produced in S. cerevisiae, Escherichia coli (E. coli) and
feeding women may get the vaccine and receiving HPV vaccines during methylotrophic yeast species, such as Hansenulapolymorpha (H.
pregnancy should not cause panic [110]. Moreover, the nonavalent polymorpha) and Pichia pastoris (P. pastoris) are currently being
HPV vaccine was evaluated in Sprague-Dawley rats and the researchers studied in preclinical or clinical trials (Table 2).
found that there were no effects on fertility or reproductive perfor-
mance and no evidence of developmental toxicity [122], supporting the 3.3. Therapeutic HPV vaccines
safety profile of 9vHPV vaccine. In a word, the HPV vaccine is recently
commercially available, and post-market surveillance must be main- Prophylactic HPV vaccines aim to induce humoral immunity against
tained to detect any adverse events following HPV vaccination. Addi- target late proteins L1 or L2, eventually leading to antibody induction
tional studies are needed to imply the possible cause-effect relationship and neutralization of antigens. In contrast to the prophylactic vaccines,
between the HPV vaccination and serious advents and to validate the the main aim of therapeutic vaccines is to eliminate the precancerous
safety of HPV vaccines. lesions and the persistent infection caused by HPV. The early protein E6
Currently, these three vaccines have been widely used and effec- and E7 are expressed constantly in HPV infection and most cervical
tively prevent HPV infections caused by the targeted types. However, cancer and precancerous lesions, but not in normal tissues. More im-
there is still a considerable population suffering from high-risk HPV portantly, the sustained expression E6 and E7 are essential for the in-
infections and associated diseases. There are some restrictions on this. duction and maintenance of the malignant phenotype of cancer cells,
Firstly, the main factor limiting vaccination is inadequate population and therefore E6 and E7 are the ideal target proteins for the develop-
coverage and global vaccine uptake. Developing countries suffer the ment of antigen-specific immunotherapy of HPV infections and related
greatest burden of HPV infection and malignancy due to the lack of diseases. In the past two decades, various therapeutic HPV vaccines
resources to implement efficient vaccination and screening programs targeting E6, E7, and other viral antigens have been widely studied,
[123]. Therefore, many women only detect infections when they have including live vector-based vaccines, nucleic acid/protein/peptide-
exceeded CIN 1 or have developed cancer. Additionally, the high prices based vaccines, and cell-based vaccines. Therapeutic vaccines are di-
of vaccines make them unable to meet the needs of low-income popu- verse and the following table describes the characteristics of vaccine
lations. By mid-2016, only 8% of low-to middle-income countries had platform technologies (Table 3) and the progress of current therapeutic
introduced the HPV vaccine, compared with71% in high-income HPV vaccines.
countries [11]. The refrigeration of the vaccine such as the cold chain
also restricts large-scale deployments in developing countries. This may 3.3.1. Live vector-based vaccines
be solved by lyophilized formulations or heat-stable capsomer pre- Live vector-based vaccines can be subdivided into recombinant viral
parations [124]. Secondly, all three vaccines have little benefit for and bacterial vector-based vaccines that can carry HPV antigens, re-
people already infected with HPV. The therapeutic vaccines that are plicate inside the host cells, and induce immune responses against HPV.
capable of inducing a cell-mediated immune response againstE6 and E7 In addition, they can drive antigen presentation through both major
proteins and clearing the infected cells are required. Thirdly, the histocompatibility complex (MHC) class I and class II pathways, thus

93
R. Wang, et al. Cancer Letters 471 (2020) 88–102

providing high levels of immunogenicity. However, the generation of


Clinical trial number
nAbs against pre-existing virus and bacteria may limit the use of these

NCT03935204

NCT01735006

NCT02710851

NCT01548118

NCT03813940

NCT04083196
vectors and restrict repeated therapy. The potential immunodominance
to the vectors instead of the HPV antigen remains another obstacle in
the use of live vector vaccines [135,136].

3.3.1.1. Bacterial vector vaccines. An ideal vaccine vector should trigger

National Vaccine and Serum


both potent innate and adaptive immune responses in the inoculated
Xiamen Innovax Biotech

Xiamen Innovax Biotech

Xiamen Innovax Biotech


host. Attenuated bacteria can stimulate both cellular and humoral

Biotechnology Co.,Ltd
immune responses to protect the host against challenge of pathogens.

Xiamen University
Shanghai Zerun
Conserved molecular patterns, such as lipoteichoic acid in gram-

Institute, China
Manufacturer

positive bacteria, lipopolysaccharides in gram-negative bacteria,


CO., LTD

CO., LTD

Co., Ltd peptidoglycans, and flagellin, are recognized by pattern recognition


receptors (PRRs) and induce innate immune responses [137]. Using a
bacterial vector to deliver heterologous antigens is an effective method
Phase III

for developing new vaccines. Although safety and efficacy issues have
Phase Ⅱ

Phase Ⅱ

Phase I

Phase I

Phase I
Status

always limited the development of bacterial vector vaccines, several


bacteria vectors have been tested in clinical trials, such as Listeria
monocytogenes (Lm), Lactobacillus casei (L. casei), Lactobacillus lactis and
Salmonella. Compared to other vaccines, bacterial vector shows several
Aluminum

Aluminum

advantages: the induction of innate and adaptive immune response to


hydroxide

hydroxide
Adjuvant

pathogens could enhance attenuated bacteria to enable specific immune


N/A

N/A

N/A

N/A

response against target antigens [138]; Using bacterial vector to deliver


heterologous antigens could reduce the difficulties of target antigen
purification and thus relatively simple and inexpensive to large-scale
Hanson's Yeast

manufacture and inoculation.


Expression

Lm is a gram-positive intracellular facultative anaerobe and is


system

E. coli

E. coli

E. coli

E. coli
Yeast

commonly used as a bacterial vector for vaccine development. By ex-


pressing membrane-active virulence factors listeriolysin O (LLO) and
phospholipase C, Lm can degrade the phagolysosomal membrane and
Acuminata

• Condylomata Acuminata

Acuminata
Intraepithelial

• Human Papillomavirus
Intraepithelial

Cervical
Intraepithelial

entry into the cytoplasm of the host [139]. Therefore, Lm could access
Infection

the cytoplasm of antigen-presenting cells (APCs), lead to the en-


HPV-related disease

Cancer

Cancer

Cancer

dogenous antigen-processing pathway and elicit both CD4+ and CD8+


Infection
• Condylomata

• Condylomata
• HPV-Related
Carcinoma

T cell-mediated immune responses through MHC class II and MHC I


Neoplasia

Neoplasia

Neoplasia
• Persistent
• Cervical
• Cervical
• Cervical

• Cervical
• Vaginal
• Vulvar

class pathways [140,141]. Currently, attenuated Lm has been used for


• HPV

developing as vaccine vector and treatment and prevention of HPV


infection and related diseases. ADXS11-001(ADXS-HPV) is a live atte-
nuated Lm-based immunotherapy that secretes antigen-adjuvant fusion
HPV-6, 11, 16, 18, 31, 33, 45, 52, and

HPV-6, 11, 16, 18, 31, 33, 45, 52, 58,

protein Lm-LLO-E7 targeting HPV-associated tumors [142]. In 2009,


HPV-6,11,16,18,31,33,45,52, and 58

Lm-LLO-E7 vaccine was tested in a phase I trial and was administrated


in 15 patients with advanced cervical cancer. The vaccine was shown to
be safe with severe (grade 3) adverse events in 6 patients, no grade 4
L1 VLP of HPV-16 and 18

L1 VLP of HPV-6 and 11

adverse events, and a reduction in total tumor size in 4 patients [143].


In 2014, ADXS11-001 was evaluated in 110recurrent cervical cancer
patients with and without cisplatin in a randomized phase II study
HPV-16 and 18

[144]. Patients were randomized to either three doses or four doses of


59 and 68

vaccines with cisplatin chemotherapy. It was encouraging that the final


Antigen

12-month survival was 36%, the 18-month survival was 28%, and the
58

response rate was 11%. The incidence of grade 3 adverse events was 2%
Recombinant human papillomavirus bivalent (types 16 and

and most non-serious adverse events were infusion-related, resolved on


11-valent Recombinant Human Papillomavirus Vaccine

their own, or responded to symptomatic treatment. GLBL101c, an oral


vaccination of HPV16 E7-expressing L. casei bacterial vector vaccine,
was tested in 17 patients with HPV16-related CIN3. The authors found
(6,11,16,18,31,33,45,52,58 Type) (E.Coli)

(6,11,16,18,31,33,45,52,58 Type)(E.Coli)
Recombinant Human Papillomavirus Vaccine

Recombinant Human Papillomavirus Vaccine

that the vaccine could elicit E7-specific mucosal immunity in the


HPV vaccines that are in clinical trial.

uterine cervical lesions, with a pathological down-grade to CIN2 in 70%


of patients who took the optimized doses. No patient was found an
adverse event [145]. Moreover, Komatsu A et al. [146] has optimized a
recombinant HPV16 E7-expressing L. casei (IGMKK16E7) to generate
optimal ratio for the amount of E7 and lactobacilli. The IGMKK16E7
18) vaccine (Yeast)

E. coli: Escherichia coli.

showed a higher induction of E7-specific IFN γ-producing cells com-


pared with the GLBL101c.
Product name

3.3.1.2. Viral vector vaccines. Because of the high infection efficiency


Gecolin
Cecolin

and high antigens expression of viral vector-based vaccines and the


Table 2

natural propensity to transduce their genetic information into the host


for replication of the viruses, viral vectors have been extensively tested,

94
R. Wang, et al. Cancer Letters 471 (2020) 88–102

Table 3
Characteristics of vaccine platform technologies.
Vaccine Platform Advantages Disadvantages Reference

Bacterial vector • Highly immunogenic • Potentially toxic/harmful to patients [135,136,138]


vaccines • Inducing both potent innate and adaptive immune • Potential immunodominance to the vectors
responses • Generation of neutralizing antibodies restrict the
• Low difficulties of target antigen purification efficacy of repeated therapy
• Simple and inexpensive to large-scale manufacture and
inoculation.
Viral vector vaccines • Highly immunogenic • Potentially toxic/harmful to patients [12,135,136,147–149,154,155]
• Wide range of selection for available vectors • Potential immunodominance to the vectors
• Can be engineered to include and express multiple genes
such as costimulatory molecules/cytokines
• Generation of neutralizing antibodies restrict the
efficacy of repeated therapy
• Inducing
responses
both potent innate and adaptive immune

DNA-based vaccines • Stable, safe, easy to manufacture and purify • Low immunogenicity [158,159]
• Capable of repeated vaccination for long-term protection • Potential risk of chromosomal integration
• Can be engineered to include and express multiple genes
such as costimulatory molecules/cytokines
• Lack intrinsic ability to self-amplify and spread to
surrounding cells
• Multiple delivery methods available
RNA-based vaccines • Easy to produce • Poor stability [167–169]
• Wide range of host cells • Inability to spread intercellularly
• Safety with no risk of chromosomal integration or • Preparation/production is labor intensive
cellular transformation • Difficult to prepare in large quantity
• Efficient delivery and high expression levels of
recombinant antigens due to high capacity of
autonomous RNA replication
Peptide-based • Stable, safe, easy to produce and store • Low immunogenicity [161,170]
vaccines • Can include multiple epitopes • MHC specific that need to match the patient's
• Can be modified for better MHC binding human leukocyte antigen (HLA) haplotypes
Protein-based • Stable, safe, easy to produce • Low immunogenicity [175,178]
vaccines • No limitation of MHC restriction • Inducing more humoral responses than cell-
mediated responses
Dendritic cell-based • Highly immunogenic • Difficult for large-scale production [182,183]
vaccines • Serve as natural adjuvants • Lack of a consensus on the optimal methods of DCs
• Multiple methods of antigen loading preparation
• Inducing
responses
both potent innate and adaptive immune • Different culture techniques may result in
inconsistent vaccine quality
• Limited
apoptosis
lifespan due to the T cell-mediated

MHC: major histocompatibility complex; DCs: dendritic cells.

such as adenoviruses (Ad), adeno-associated viruses, alphaviruses, and the immunogenicity of this vaccine in prime-boost immunization regi-
vaccinia viruses. Foreign genes that code immunogenic proteins from mens via heterologous methods combining intramuscular and in-
pathogens are used to replace non-essential viral genes, and thus the travaginal immunization in mice. They found that systemic prime fol-
recombinant viral vectors could transduce the target cell and express lowing local intravaginal boost represented the most promising
the encoded antigens. combination to efficiently induce considerable CD8+ T cell responses.
Adenovirus, an80-100 nm, non-enveloped, double-stranded DNA Vaccinia virus is a double-stranded DNA virus, which belongs to the
virus, represents one of the most promising vaccine platforms used for Poxviridae family. The poxvirus has a large and stable genome that can
gene therapy and vaccine vectors. Ad vector vaccines have many ad- express large amounts of foreign antigen. Modified vaccinia virus
vantages that make them attractive and widely used [147–149]: (1) Ankara (MVA) is licensed as the third-generation vaccine against
Deletion of the E1 region which is critical for the initiation of virus smallpox and serves as an effective vector platform for the development
replication and replacement with the foreign gene results in replication- of new vaccines. The MVA has several features that make the re-
defective virions that have low risk of insertional oncogenesis for combinant MVA platform highly suitable as a heterologous viral vector:
human use; (2) Deletion of theE2, E3 and E4 regions increase the ca- (1)large capacity for transgene insertion; (2) ease of inexpensive man-
pacity for transgene insertion, and multiple transgenes such as the ufacture; (3)safe administration by the intradermal, intranasal, in-
target antigen and other genes that enhance the response to vaccination travaginal and intrarectal routes; (4) thermostability for application in
can be inserted into the recombinant viral vectors; (3) Inducing robust resource-poor settings without a cold chain [12,154,155]. Recently,
immune responses that are associated with pro-inflammatory cytokines Rosales R et al. evaluated an MVA viral vector targeting HPV16 E2
and both the innate and adaptive immunity. However, Ad is ubiquitous (MVA-E2)to treat intraepithelial lesions associated with the HPV in-
in humans and up to 60% of adults in Europe and the USA have high fection in a phase III trial of a total of 1176 female and 180 male pa-
titers of nAbs to adenovirus type 5 (Ad5). Therefore, animal or rare tients. MVA E2 virus particles were directly injected into the uterus,
human Ad serotypes such as Ad26 and Ad35 are currently evaluated to urethra, vulva or anus. After treatment with MVA E2, 89.3% of the
overcome type-specific anti-vector immunity [150,151]. In 2017, Khan female patients and all the male patients observed complete elimination
et al. [152]developed replication‐deficient Ad26 and Ad35-based of lesions, and no apparent side effects were generated. However, there
vector vaccines that consisted of fusion proteins of HPV E2, E6, and E7 is no control vaccine in the study, the true vaccine efficacy is uncertain
for HPV16 and HPV18 related disease. They found considerable ther- [156].
apeutic efficacy in the TC‐1 mouse model, indicating that the developed
vaccine vectors form a promising therapeutic vaccine candidate for
active immunotherapy. Subsequently, Çuburu Net al. [153]evaluated 3.3.2. Nucleic acid/protein/peptide-based vaccines
3.3.2.1. DNA-based vaccines. DNA-based vaccines have become a safe

95
R. Wang, et al. Cancer Letters 471 (2020) 88–102

alternative to standard live and inactivated vaccines for human and adjuvants are required to enhance the immune response. Peptide-
animal infections [157]. They exhibit several advantages over based vaccines can be divided into synthetic long peptides (SLPs) and
traditional strategies: (1) easy to manufacture and purify; (2) high specific epitope (short) peptides. The short peptides are restricted to the
purity and stability compared to protein-based vaccines; (3) safe patient's specific HLA type and need preliminary HLA typing before
compared to live vector-based vaccines; (4) capable of inducing both administration [160]. Short peptide-based therapeutic HPV vaccines
humoral and cell‐mediated immune responses; (5) unlike the live (CIGB-228) adjuvated with very small size proteoliposomes (VSSP) was
vector-based vaccine and protein-based vaccines, they do not produce tested in 7 women with HPV16-positive high-grade CIN. The vaccine-
nAbs to the vector, allowing for repeated vaccination for long-term induced IFN-γ-associated T-cell response and 5 patients had lesion
protection [158,159]. However, the main limitation of DNA-based regression and HPV clearance [173].
vaccines is poor immunogenicity. To increase immunogenicity, Compared to short peptides, SLPs do not require patient selection
strategies such as optimizing delivery approaches with the use of with specific MHC profile, but they need to be processed and presented
electroporation, encapsulation, gene gun or laser therapy, fusions of by APCs [174]. In the dose-escalation phase I clinical study, PepCan, a
antigens with T-cells activating molecules, the use of peptide-based vaccine consisting of 4 HPV16 E6 synthetic peptides and
immunomodulators such as potent adjuvants, priming with DNA a novel adjuvant Candin, was carried out in 24 patients with CIN2/3.
vectors followed by boosting with viral vectors have been The trial showed that no dose-limiting toxicity, histological regression
investigated, and modification of the properties of antigen-presenting of disease in 45%of patients, a significant decrease in viral load, and an
cells, especially dendritic cells (DCs) [160–162]. In 2014, Kim et al. increased immune response [175]. Additionally, the phase II trial
[163] developed the DNA vaccine GX-188E that expressed HPV16/18 evaluating the efficacy and safety of PepCan in cervical high-grade
E6 and E7 antigens and the Fms-like tyrosine kinase-3 ligand (Flt3L), squamous intraepithelial lesions is ongoing (NCT02481414). Kenter
which could activate DCs. They used electroporation to enhance et al. [176]tested the immunogenicity and the efficacy of a synthetic
immunization. The vaccine could elicitE6/E7-specific IFN-γ-secreting long-peptide vaccine in 20 women with high-grade HPV16 related
T-cell responses in 9 patients with HPV16/18 + CIN 3, with 7 patients vulvar intraepithelial neoplasia (VIN). At 12 months of follow-up,
displaying complete lesion regression within 36 weeks of follow up and complete responses were found in 47% of the patients and maintained
no serious vaccine-associated adverse events in all patients. In a at 24 months. Patients with a complete response had a stronger IFN-γ-
randomized, double-blind, placebo-controlled phase IIb study [164], associated CD4+ andCD8+ T-cell response. In 2013, the HPV16 syn-
the VGX-3100, synthetic plasmids targeting E6 and E7 proteins of thetic long-peptide vaccine consisting of 13 HPV16 E6 and HPV16 E7
HPV16 and HPV18, delivered by electroporation, was tested in women overlapping long peptides and combination with Montanide ISA-51
with CIN2/3. Histopathological regression was observed in 49.5% of adjuvant was evaluated in 20 women with advanced or recurrent cer-
vaccine recipients compared with 30.6% of placebo recipients. VGX- vical cancer. The HPV16-specific T-cell response associated with IFN-γ,
3100 is the first and the most successful therapeutic DNA vaccine to TNFα, IL-5 and/or IL-10 was found in 9 patients. However, neither
show efficacy against CIN2/3to date. Currently, the efficacy, safety, and regressions of the tumor nor prevention of the progressive disease were
tolerability of VGX-3100 are evaluated in women with histologically developed [177]. Moreover, the novel therapeutic synthetic long pep-
confirmed HPV16/18-positive CIN2/3 in a phase III trial tide vaccine targeting HPV16 ISA101/ISA101b is ongoing to assess the
(NCT03185013). In another phase II study, VGX-3100 alone or in safety, tolerability and the HPV-specific immune responses in women
combination with imiquimod is tested in women with HPV16/18- with advanced or recurrent cervical cancer (NCT02128126).
positive high-grade lesions of the vulva (NCT03180684).
3.3.2.4. Protein-based vaccines. Protein-based vaccines can avoid the
3.3.2.2. RNA-based vaccines. RNA-based vaccines are based on RNA limitation of MHC restriction due to the numerous CD4+ and CD8+T
replicon systems derived from single-stranded RNA viruses of positive epitopes compared to peptide-based vaccines. However, the major
or negative polarity and the virus vectors are non‐pathogenic [161]. limitations are that they may induce the antibody responses rather
Various RNA viruses such as retroviruses, lentiviruses, alphaviruses, than CTL responses because of the preferential presentation via MHC
flaviviruses rhabdoviruses, measles viruses, Newcastle disease viruses, class II. Therefore, adjuvants have been used for therapeutic
and picornaviruses have been engineered as vectors for expression of vaccination to enhance the APC presentation, target the antigen to
antigens and treatment of diseases [165]. Currently, numerous studies DCs, and improve immunogenicity [175,178]. Several protein-based
have demonstrated that self-replicating RNA viral vectors could provide vaccines have entered clinical trials. TA-CIN, a subunit vaccine
efficient delivery and high expression levels of recombinant comprising an HPV16 E6E7L2 fusion protein, has been proven safe
heterologous antigens due to high capacity of autonomous RNA and immunogenic in a number of clinical trials [179,180]. Daayana
replication, and administration of RNA viral vectors could generate et al. [181]treated 19 women with VIN grades 2 and 3 using a topical
strong immune responses and provide protection against challenges of immunomodulator (imiquimod) followed by TA-CIN. At week 52, it
infectious agents and tumor cells. By acting as agonists for TLR7 and was shown that 63% of patients observed the complete histological
TLR8, RNA viral vectors can also induce the innate immune responses regression of VIN and increased local infiltration of CD4+ and CD8+ T
[166]. Easy to produce virus, wide range of host cells, safety with no cells. Currently, TA-CIN vaccine is evaluating in 14 patients with
risk of chromosomal integration or cellular transformation as the viral HPV16-associated cervical cancer to determine the safety and
RNA degrades within 3–5 days, targeting DCs, rapid generation of RNA feasibility as well as the induction of HPV antigen-specific immune
copies and high expression levels in the cytoplasm, and the combination response (NCT02405221).
with either polymer- or liposome-based encapsulation strategies make
the RNA viral vectors an attractive approach [167,168]. However, the 3.3.3. Cell-based vaccines
limitations of RNA-based vaccines are poor stability and inability to 3.3.3.1. Dendritic cell-based vaccines. Dendritic cells (DCs) are ideal
spread intercellularly [169]. To date, there is few clinical testing on candidates for immunotherapy strategies as they have been recognized
RNA-based vaccines associated with HPV-related diseases. as the most potent APCs in vivo, capable of mediating and inducing
both innate immune response and adaptive immune response. DCs have
3.3.2.3. Peptide-based vaccines. Peptide-based vaccines have several a strong ability to acquire and process antigens for presentation to T
advantages, particularly concerning stability, safety, and ease cells and express high levels of costimulatory or coinhibitory molecules
production and storage [161,170]. However, limitations include low [182]. Moreover, DCs can serve as natural adjuvants to increase the
immunogenicity and MHC specific that need to match the patient's potency of antigen-specific immunotherapy against cancer [183]. DCs
human leukocyte antigen (HLA) haplotypes [171,172]. Therefore, can be generated in vitro through optimal methods. Then the DCs can

96
R. Wang, et al. Cancer Letters 471 (2020) 88–102

be loaded with specific peptide or antigens to make a vaccine. The DC- promising clinical responses, there was no clear relationship between
based vaccines can be adoptively transferred into patients and maintain induced immune responses and clinical responders. This should con-
their capacity to induce specific CTL responses and exert efficient tribute to the lack of knowledge about the mechanisms of immune re-
antiviral or antitumor responses. DC-based vaccines have several sponse that control and clear the HPV-infected cells in the host.
limitations, such as difficult for large-scale production, lack of a Meanwhile, research on the cancer microenvironment and immune
consensus on the optimal methods of DCs preparation, different evasion mechanism are still unclear. We believe that when more in-
culture techniques may result in inconsistent vaccine quality, and formation regarding the immune mechanisms against HPV infection is
limit lifespan due to the T cell-mediated apoptosis [183]. To prolong obtained, strategies to design better vaccines, including the use of ef-
DCs survival, short interfering RNA (siRNAs)against pro-apoptotic ficient adjuvants will lead to the development of effective in the future.
molecules have been developed [184,185].
DC-based vaccines can be divided into DCs pulsed with HPV-specific Funding
peptides/protein antigens or DCs transduced with DNA/viral vectors
encoding foreign antigens [186]. DCs can be prepared as mature or This work was supported by the National Natural Science
immature cells. Most trials utilized the mature DCs as the immature Foundation of China (81672085; 81372804; 71871169; U1933120;
types lack the full T cell co-stimulatory activity [178]. However, it is 71373188; U1333115; 30901586); the Chinese medical association of
unclear whether DCs maturation is necessary to stimulate an immune clinical medicine special funds for scientific research projects
response in vaccines. The immature DCs has the potential to reduce (17020400709), the Hubei Provincial Natural Science Foundation of
vaccine production cost compared to the mature DCs [187]. Therefore, China (2019CFA062).
the pre-immature DCs (PIDCs) have been tested as cancer vaccines.
Rahma et al. [187] used the PIDC pulsed with HPV16 E6 or E7 peptide Declaration of competing interest
to treat 32patients with advanced cervical cancers. The specific im-
mune response was 63% (E6) and 58% (E7) of the patients. Further The authors have no conflicts of interest to report.
investigations are required to evaluate PIDCs for peptide delivery in
vaccines. References

3.3.3.2. Adoptive T-cell therapy. Adoptive T-cell therapy (ACT) involves [1] Human Papillomavirus (HPV) and Cervical Cancer, https://www.who.int/en/
the identification and expansion of desired specificity and phenotype T news-room/fact-sheets/detail/human-papillomavirus-(hpv)-and-cervical-cancer.
[2] Human papillomavirus vaccines: WHO position paper, May 2017-
cells in vitro and the reinfusion of antigen-specific T cells. ACT allows Recommendations, Vaccine 35 (2017) 5753–5755.
for appropriate in vitro manipulation and selection of the T cells for [3] D.M. Harper, L.R. DeMars, HPV vaccines - a review of the first decade, Gynecol.
immunotherapy, which produces 10-fold greater frequencies of Oncol. 146 (2017) 196–204.
[4] H. zur Hausen, Papillomaviruses and cancer: from basic studies to clinical appli-
antigen-specific T cells than those by using current vaccine regimens cation, Nat. Rev. Cancer 2 (2002) 342–350.
alone [188]. Stevanovic et al. [189]evaluated ACT in 9 patients [5] J.T. Schiller, P.M. Day, R.C. Kines, Current understanding of the mechanism of
diagnosed with metastatic cervical cancer. HPV-targeted tumor- HPV infection, Gynecol. Oncol. 118 (2010) S12–S17.
[6] S. de Sanjose, W.G. Quint, L. Alemany, D.T. Geraets, J.E. Klaustermeier,
infiltrating T cells (HPV-TILs) were selected and reinfused into B. Lloveras, S. Tous, A. Felix, L.E. Bravo, H.R. Shin, C.S. Vallejos, P.A. de Ruiz,
patients. Objective tumor responses were observed in 3 patients and M.A. Lima, N. Guimera, O. Clavero, M. Alejo, A. Llombart-Bosch, C. Cheng-Yang,
2 complete responses were observed at 22 and 15 months after S.A. Tatti, E. Kasamatsu, E. Iljazovic, M. Odida, R. Prado, M. Seoud, M. Grce,
A. Usubutun, A. Jain, G.A. Suarez, L.E. Lombardi, A. Banjo, C. Menendez,
treatment. Recently, Doran et al. [190] developed autologous
E.J. Domingo, J. Velasco, A. Nessa, S.C. Chichareon, Y.L. Qiao, E. Lerma,
genetically engineered T cells expressing a T-cell receptor directed S.M. Garland, T. Sasagawa, A. Ferrera, D. Hammouda, L. Mariani, A. Pelayo,
against HPV16 E6 and treated 12 patients with metastatic HPV- I. Steiner, E. Oliva, C.J. Meijer, W.F. Al-Jassar, E. Cruz, T.C. Wright, A. Puras,
associated epithelial cancers in Phase I/II Study. Regression of C.L. Llave, M. Tzardi, T. Agorastos, V. Garcia-Barriola, C. Clavel, J. Ordi,
M. Andujar, X. Castellsague, G.I. Sanchez, A.M. Nowakowski, J. Bornstein,
metastatic epithelial cancer and objective responses were observed N. Munoz, F.X. Bosch, S. Retrospective InternationalH.P.V.T.T.S. Group, Human
and the subsequent trial of T cell receptor gene therapy for papillomavirus genotype attribution in invasive cervical cancer: a retrospective
HPV16 + cancers targeting E7 is conducting (NCT02858310). cross-sectional worldwide study, Lancet Oncol. 11 (2010) 1048–1056.
[7] S.K. Kjaer, K. Frederiksen, C. Munk, T. Iftner, Long-term absolute risk of cervical
However, it is thought that CTLs alone may not be sufficient to intraepithelial neoplasia grade 3 or worse following human papillomavirus in-
eliminate cancer cells in patients with advanced HPV cancers. The fection: role of persistence, J. Nat. Cancer. Inst. 102 (2010) 1478–1488.
evasion of the immune system and the treatment of solid tumors remain [8] A.K. Chaturvedi, Beyond cervical cancer: burden of other HPV-related cancers
among men and women, J. Adolesc. Health 46 (2010) S20–S26.
a challenge. ACT needs to address issues such as tumor heterogeneity, [9] L.A. Denny, S. Franceschi, S. de Sanjose, I. Heard, A.B. Moscicki, J. Palefsky,
antigen escape, immunosuppressive microenvironment, and fragile Human papillomavirus, human immunodeficiency virus and immunosuppression,
blood vessel status [191–193]. Vaccine 30 (Suppl 5) (2012) F168–F174.
[10] X. Wang, X. Huang, Y. Zhang, Involvement of human papillomaviruses in cervical
cancer, Front. Microbiol. 9 (2018) 2896.
4. Conclusion [11] L. Bruni, M. Diaz, L. Barrionuevo-Rosas, R. Herrero, F. Bray, F.X. Bosch, S. de
Sanjose, X. Castellsague, Global estimates of human papillomavirus vaccination
coverage by region and income level: a pooled analysis, Lancet Glob. Health 4
Ten years after the introduction of HPV vaccines, there has been a
(2016) e453–463.
great success in the development of prophylactic HPV vaccines against [12] G. Hancock, K. Hellner, L. Dorrell, Therapeutic HPV vaccines, Best practice &
certain types of HPV infections, showing efficacy and safety in pre- research, Clin. Obstet. Gynaecol. 47 (2018) 59–72.
venting persistent infections of precancerous lesions by approximately [13] M. Tommasino, The human papillomavirus family and its role in carcinogenesis,
Semin. Cancer Biol. 26 (2014) 13–21.
95% of the vaccinated individuals. A series of limitations such as lim- [14] C. Schellenbacher, R.B.S. Roden, R. Kirnbauer, Developments in L2-based human
ited HPV type for prevention and high cost of manufacture restrict the papillomavirus (HPV) vaccines, Virus Res. 231 (2017) 166–175.
widespread use. Various types of second-generation prophylactic HPV [15] K. Van Doorslaer, Z. Chen, H.U. Bernard, P.K.S. Chan, R. DeSalle, J. Dillner,
O. Forslund, T. Haga, A.A. McBride, L.L. Villa, R.D. Burk, C. Ictv Report, ICTV
vaccines and HPV L2-based vaccines are currently undergoing pre- virus Taxonomy profile: Papillomaviridae, J. Gen. Virol. 99 (2018) 989–990.
clinical or clinical trials. Moreover, therapeutic HPV vaccines are ur- [16] H.U. Bernard, R.D. Burk, Z. Chen, K. van Doorslaer, H. zur Hausen, E.M. de
gently required to reduce the burden of cervical cancer as the licensed Villiers, Classification of papillomaviruses (PVs) based on 189 PV types and pro-
posal of taxonomic amendments, Virology 401 (2010) 70–79.
HPV vaccines cannot help those people who are already infected with [17] E.M. de Villiers, Cross-roads in the classification of papillomaviruses, Virology 445
HPV and developing the associated tumors. In contrast to the prophy- (2013) 2–10.
lactic HPV vaccines, progress in the development of therapeutic stra- [18] L.S.A. Muhr, C. Eklund, J. Dillner, Towards quality and order in human papillo-
mavirus research, Virology 519 (2018) 74–76.
tegies has so far been very slow. Though several studies showed

97
R. Wang, et al. Cancer Letters 471 (2020) 88–102

[19] R.N. Brancaccio, A. Robitaille, S. Dutta, C. Cuenin, D. Santare, G. Skenders, papillomaviruses by binding virions and transferring them to adjacent cells, J.
M. Leja, N. Fischer, A.R. Giuliano, D.E. Rollison, A. Grundhoff, M. Tommasino, Virol. 80 (2006) 8940–8950.
T. Gheit, Generation of a novel next-generation sequencing-based method for the [43] R.M. Richards, D.R. Lowy, J.T. Schiller, P.M. Day, Cleavage of the papillomavirus
isolation of new human papillomavirus types, Virology 520 (2018) 1–10. minor capsid protein, L2, at a furin consensus site is necessary for infection, Proc.
[20] M. Tommasino, The biology of beta human papillomaviruses, Virus Res. 231 Natl. Acad. Sci. U.S.A. 103 (2006) 1522–1527.
(2017) 128–138. [44] P.M. Day, J.T. Schiller, The role of furin in papillomavirus infection, Future
[21] J. Haedicke, T. Iftner, Human papillomaviruses and cancer, Radiother. Oncol. 108 Microbiol. 4 (2009) 1255–1262.
(2013) 397–402. [45] S. DiGiuseppe, M. Bienkowska-Haba, L.G. Guion, M. Sapp, Cruising the cellular
[22] M. Plummer, C. de Martel, J. Vignat, J. Ferlay, F. Bray, S. Franceschi, Global highways: how human papillomavirus travels from the surface to the nucleus,
burden of cancers attributable to infections in 2012: a synthetic analysis, Lancet Virus Res. 231 (2017) 1–9.
Glob. Health 4 (2016) e609–616. [46] P.M. Day, C.D. Thompson, R.M. Schowalter, D.R. Lowy, J.T. Schiller,
[23] M. Arbyn, M. Tommasino, C. Depuydt, J. Dillner, Are 20 human papillomavirus Identification of a role for the trans-Golgi network in human papillomavirus 16
types causing cervical cancer? J. Pathol. 234 (2014) 431–435. pseudovirus infection, J. Virol. 87 (2013) 3862–3870.
[24] M.G. Hawkins, D.M. Winder, S.L. Ball, K. Vaughan, C. Sonnex, M.A. Stanley, [47] A. Lipovsky, A. Popa, G. Pimienta, M. Wyler, A. Bhan, L. Kuruvilla, M.A. Guie,
J.C. Sterling, P.K. Goon, Detection of specific HPV subtypes responsible for the A.C. Poffenberger, C.D. Nelson, W.J. Atwood, D. DiMaio, Genome-wide siRNA
pathogenesis of condylomata acuminata, Virol. J. 10 (2013) 137. screen identifies the retromer as a cellular entry factor for human papillomavirus,
[25] E.M. Burd, Human papillomavirus laboratory testing: the changing paradigm, Proc. Natl. Acad. Sci. U.S.A. 110 (2013) 7452–7457.
Clin. Microbiol. Rev. 29 (2016) 291–319. [48] P.M. Day, C.C. Baker, D.R. Lowy, J.T. Schiller, Establishment of papillomavirus
[26] J. Mirkovic, B.E. Howitt, P. Roncarati, S. Demoulin, M. Suarez-Carmona, infection is enhanced by promyelocytic leukemia protein (PML) expression, Proc.
P. Hubert, F.D. McKeon, W. Xian, A. Li, P. Delvenne, C.P. Crum, M. Herfs, Natl. Acad. Sci. U.S.A. 101 (2004) 14252–14257.
Carcinogenic HPV infection in the cervical squamo-columnar junction, J. Pathol. [49] M. Bienkowska-Haba, W. Luszczek, T.R. Keiffer, L.G.M. Guion, S. DiGiuseppe,
236 (2015) 265–271. R.S. Scott, M. Sapp, Incoming human papillomavirus 16 genome is lost in PML
[27] C.A. Moody, L.A. Laimins, Human papillomavirus oncoproteins: pathways to protein-deficient HaCaT keratinocytes, Cell Microbiol. 19 (2017).
transformation, Nat. Rev. Cancer 10 (2010) 550–560. [50] K. Munger, J.R. Basile, S. Duensing, A. Eichten, S.L. Gonzalez, M. Grace,
[28] Y. Liu, J.D. Baleja, Structure and function of the papillomavirus E6 protein and its V.L. Zacny, Biological activities and molecular targets of the human papilloma-
interacting proteins, Front. Biosci. 13 (2008) 121–134. virus E7 oncoprotein, Oncogene 20 (2001) 7888–7898.
[29] E.J. Crosbie, M.H. Einstein, S. Franceschi, H.C. Kitchener, Human papillomavirus [51] A. Hildesheim, R. Herrero, S. Wacholder, A.C. Rodriguez, D. Solomon, M.C. Bratti,
and cervical cancer, Lancet 382 (2013) 889–899. J.T. Schiller, P. Gonzalez, G. Dubin, C. Porras, S.E. Jimenez, D.R. Lowy, Effect of
[30] S.J. Piersma, Immunosuppressive tumor microenvironment in cervical cancer human papillomavirus 16/18 L1 viruslike particle vaccine among young women
patients, Cancer Microenviron. 4 (2011) 361–375. with preexisting infection: a randomized trial, J. Am. Med. Assoc. 298 (2007)
[31] P. Bonanni, S. Boccalini, A. Bechini, Efficacy, duration of immunity and cross 743–753.
protection after HPV vaccination: a review of the evidence, Vaccine 27 (Suppl 1) [52] M. Narisawa-Saito, T. Kiyono, Basic mechanisms of high-risk human papilloma-
(2009) A46–A53. virus-induced carcinogenesis: roles of E6 and E7 proteins, Cancer Sci. 98 (2007)
[32] A.B. Moscicki, M. Schiffman, A. Burchell, G. Albero, A.R. Giuliano, M.T. Goodman, 1505–1511.
S.K. Kjaer, J. Palefsky, Updating the natural history of human papillomavirus and [53] L.F. Wetherill, K.K. Holmes, M. Verow, M. Muller, G. Howell, M. Harris,
anogenital cancers, Vaccine 30 (Suppl 5) (2012) F24–F33. C. Fishwick, N. Stonehouse, R. Foster, G.E. Blair, S. Griffin, A. Macdonald, High-
[33] R. De Vincenzo, C. Ricci, C. Conte, G. Scambia, HPV vaccine cross-protection: risk human papillomavirus E5 oncoprotein displays channel-forming activity
highlights on additional clinical benefit, Gynecol. Oncol. 130 (2013) 642–651. sensitive to small-molecule inhibitors, J. Virol. 86 (2012) 5341–5351.
[34] C.L. Trimble, R.A. Clark, C. Thoburn, N.C. Hanson, J. Tassello, D. Frosina, F. Kos, [54] A.C. de Freitas, T.H.A. de Oliveira, M.R. Barros Jr., A. Venuti, hrHPV E5 onco-
J. Teague, Y. Jiang, N.C. Barat, A.A. Jungbluth, Human papillomavirus 16-asso- protein: immune evasion and related immunotherapies, J. Exp. Clin. Cancer Res.
ciated cervical intraepithelial neoplasia in humans excludes CD8 T cells from 36 (2017) 71.
dysplastic epithelium, J. Immunol. 185 (2010) 7107–7114. [55] N. Kivi, D. Greco, P. Auvinen, E. Auvinen, Genes involved in cell adhesion, cell
[35] A. de Jong, M.I. van Poelgeest, J.M. van der Hulst, J.W. Drijfhout, G.J. Fleuren, motility and mitogenic signaling are altered due to HPV 16 E5 protein expression,
C.J. Melief, G. Kenter, R. Offringa, S.H. van der Burg, Human papillomavirus type Oncogene 27 (2008) 2532–2541.
16-positive cervical cancer is associated with impaired CD4+ T-cell immunity [56] A.H. Hopman, F. Smedts, W. Dignef, M. Ummelen, G. Sonke, M. Mravunac,
against early antigens E2 and E6, Cancer Res. 64 (2004) 5449–5455. G.P. Vooijs, E.J. Speel, F.C. Ramaekers, Transition of high-grade cervical in-
[36] P.E. Gravitt, The known unknowns of HPV natural history, J. Clin. Investig. 121 traepithelial neoplasia to micro-invasive carcinoma is characterized by integration
(2011) 4593–4599. of HPV 16/18 and numerical chromosome abnormalities, J. Pathol. 202 (2004)
[37] S. Jeon, P.F. Lambert, Integration of human papillomavirus type 16 DNA into the 23–33.
human genome leads to increased stability of E6 and E7 mRNAs: implications for [57] M. Seoud, W.A. Tjalma, V. Ronsse, Cervical adenocarcinoma: moving towards
cervical carcinogenesis, Proc. Natl. Acad. Sci. U.S.A. 92 (1995) 1654–1658. better prevention, Vaccine 29 (2011) 9148–9158.
[38] S.J. Piersma, Immunosuppressive tumor microenvironment in cervical cancer [58] K. Holl, A.M. Nowakowski, N. Powell, W.G. McCluggage, E.C. Pirog, S. Collas De
patients, Cancer Microenviron. 4 (2011) 361–375. Souza, W.A. Tjalma, M. Rosenlund, A. Fiander, M. Castro Sanchez, V. Damaskou,
[39] K. Torres-Poveda, M. Bahena-Roman, C. Madrid-Gonzalez, A.I. Burguete-Garcia, E.A. Joura, B. Kirschner, R. Koiss, J. O'Leary, W. Quint, O. Reich, A. Torne,
V.H. Bermudez-Morales, O. Peralta-Zaragoza, V. Madrid-Marina, Role of IL-10 and M. Wells, L. Rob, L. Kolomiets, A. Molijn, A. Savicheva, E. Shipitsyna, D. Rosillon,
TGF-beta1 in local immunosuppression in HPV-associated cervical neoplasia, D. Jenkins, Human papillomavirus prevalence and type-distribution in cervical
World J. Clin. Oncol. 5 (2014) 753–763. glandular neoplasias: results from a European multinational epidemiological
[40] N. Cancer Genome Atlas Research, M. Albert Einstein College of, S. Analytical study, Int. J. Cancer 137 (2015) 2858–2868.
Biological, H. Barretos Cancer, M. Baylor College of, H. Beckman Research [59] E.C. Pirog, Cervical adenocarcinoma: diagnosis of human papillomavirus-positive
Institute of City of, A. Buck Institute for Research on, C. Canada’s Michael Smith and human papillomavirus-negative tumors, Arch. Pathol. Lab Med. 141 (2017)
Genome Sciences, S. Harvard Medical, F.G.C.C. Helen, S. Research Institute at 1653–1667.
Christiana Care Health, B. HudsonAlpha Institute for, L.L.C. Ilsbio, M. Indiana [60] W. Tjalma, HPV negative cervical cancers and primary HPV screening, Facts Views
University School of, V. Institute of Human, B. Institute for Systems, C. Vis. Obgyn. 10 (2018) 107–113.
International Genomics, B. Leidos, H. Massachusetts General, U. McDonnell [61] N. Wentzensen, M. Schiffman, Filling a gap in cervical cancer screening pro-
Genome Institute at Washington, W. Medical College of, C. Medical University of grammes, Lancet Oncol. 15 (2014) 249–251.
South, C. Memorial Sloan Kettering Cancer, C. Montefiore Medical, NantOmics, I. [62] G. Ronco, J. Dillner, K.M. Elfstrom, S. Tunesi, P.J. Snijders, M. Arbyn,
National Cancer, A.N. National Hospital, I. National Human Genome Research, S. H. Kitchener, N. Segnan, C. Gilham, P. Giorgi-Rossi, J. Berkhof, J. Peto,
National Institute of Environmental Health, D. National Institute on, D. Other C.J. Meijer, Efficacy of HPV-based screening for prevention of invasive cervical
Communication, L.H.S.C. Ontario Tumour Bank, O.I.f.C.R. Ontario Tumour Bank, cancer: follow-up of four European randomised controlled trials, Lancet (London,
T.O.H. Ontario Tumour Bank, H. Oregon, U. Science, C.-S.M.C. Samuel Oschin England) 383 (2014) 524–532.
Comprehensive Cancer Institute, S.R.A. International, S. St Joseph’s Candler [63] N. Wentzensen, M. Schiffman, T. Palmer, M. Arbyn, Triage of HPV positive women
Health, Eli, L.B.I.o.M.I.o.T. Edythe, U. Harvard, H. Research Institute at in cervical cancer screening, J. Clin. Virol. 76 (Suppl 1) (2016) S49–S55.
Nationwide Children’s, U. Sidney Kimmel Comprehensive Cancer Center at Johns [64] T.F. Schwarz, M. Kocken, T. Petaja, M.H. Einstein, M. Spaczynski, J.A. Louwers,
Hopkins, B. University of, M.D.A.C.C. University of Texas, H. University of Abuja C. Pedersen, M. Levin, T. Zahaf, S. Poncelet, K. Hardt, D. Descamps, G. Dubin,
Teaching, B. University of Alabama at, I. University of California, C. University of Correlation between levels of human papillomavirus (HPV)-16 and 18 antibodies
California Santa, C. University of Kansas Medical, L. University of, C. University of in serum and cervicovaginal secretions in girls and women vaccinated with the
New Mexico Health Sciences, H. University of North Carolina at Chapel, C. HPV-16/18 AS04-adjuvanted vaccine, Hum. Vaccine 6 (2010) 1054–1061.
University of Oklahoma Health Sciences, P. University of, R.a.P.M.S. University of [65] J.C. Steele, C.H. Mann, S. Rookes, T. Rollason, D. Murphy, M.G. Freeth,
Sao Paulo, C. University of Southern, W. University of, M. University of Wisconsin P.H. Gallimore, S. Roberts, T-cell responses to human papillomavirus type 16
School of, H. Public, I. Van Andel Research, L. Washington University in St, among women with different grades of cervical neoplasia, Br. J. Canc. 93 (2005)
Integrated genomic and molecular characterization of cervical cancer, Nature 543 248–259.
(2017) 378–384. [66] K.H. Kim, W.W. Greenfield, M.J. Cannon, H.N. Coleman, H.J. Spencer,
[41] C.B. Buck, P.M. Day, B.L. Trus, The papillomavirus major capsid protein L1, M. Nakagawa, CD4+ T-cell response against human papillomavirus type 16 E6
Virology 445 (2013) 169–174. protein is associated with a favorable clinical trend, Cancer Immunol.
[42] T.D. Culp, L.R. Budgeon, M.P. Marinkovich, G. Meneguzzi, N.D. Christensen, Immunother.(CII) 61 (2012) 63–70.
Keratinocyte-secreted laminin 5 can function as a transient receptor for human [67] Y.L. Woo, M. van den Hende, J.C. Sterling, N. Coleman, R.A. Crawford,

98
R. Wang, et al. Cancer Letters 471 (2020) 88–102

K.M. Kwappenberg, M.A. Stanley, S.H. van der Burg, A prospective study on the J. Salmeron, S.N. Chow, D. Apter, H. Kitchener, J.C. Teixeira, S.R. Skinner,
natural course of low-grade squamous intraepithelial lesions and the presence of U. Jaisamrarn, G. Limson, B. Romanowski, F.Y. Aoki, T.F. Schwarz, W.A. Poppe,
HPV16 E2-, E6- and E7-specific T-cell responses, Int. J. Cancer 126 (2010) F.X. Bosch, D.M. Harper, W. Huh, K. Hardt, T. Zahaf, D. Descamps, F. Struyf,
133–141. G. Dubin, M. Lehtinen, H.P.S. Group, Cross-protective efficacy of HPV-16/18
[68] S. Monnier-Benoit, F. Mauny, D. Riethmuller, J.S. Guerrini, M. Capilna, S. Felix, AS04-adjuvanted vaccine against cervical infection and precancer caused by non-
E. Seilles, C. Mougin, J.L. Pretet, Immunohistochemical analysis of CD4+ and vaccine oncogenic HPV types: 4-year end-of-study analysis of the randomised,
CD8+ T-cell subsets in high risk human papillomavirus-associated pre-malignant double-blind PATRICIA trial, Lancet Oncol. 13 (2012) 100–110.
and malignant lesions of the uterine cervix, Gynecol. Oncol. 102 (2006) 22–31. [92] M. Lehtinen, J. Paavonen, C.M. Wheeler, U. Jaisamrarn, S.M. Garland,
[69] S.H. van der Burg, C.J. Melief, Therapeutic vaccination against human papilloma X. Castellsague, S.R. Skinner, D. Apter, P. Naud, J. Salmeron, S.N. Chow,
virus induced malignancies, Curr. Opin. Immunol. 23 (2011) 252–257. H. Kitchener, J.C. Teixeira, J. Hedrick, G. Limson, A. Szarewski, B. Romanowski,
[70] M.P. Morrow, J. Yan, N.Y. Sardesai, Human papillomavirus therapeutic vaccines: F.Y. Aoki, T.F. Schwarz, W.A. Poppe, N.S. De Carvalho, M.J. Germar, K. Peters,
targeting viral antigens as immunotherapy for precancerous disease and cancer, A. Mindel, P. De Sutter, F.X. Bosch, M.P. David, D. Descamps, F. Struyf, G. Dubin,
Expert Rev. Vaccines 12 (2013) 271–283. H.P.S. Group, Overall efficacy of HPV-16/18 AS04-adjuvanted vaccine against
[71] Gardasil, https://www.fda.gov/vaccines-blood-biologics/vaccines/gardasil. grade 3 or greater cervical intraepithelial neoplasia: 4-year end-of-study analysis
[72] Gardasil, 9 https://www.fda.gov/vaccines-blood-biologics/vaccines/gardasil-9. of the randomised, double-blind PATRICIA trial, Lancet Oncol. 13 (2012) 89–99.
[73] E.J. Crosbie, H.C. Kitchener, Cervarix–a bivalent L1 virus-like particle vaccine for [93] C.M. Wheeler, S.R. Skinner, M.R. Del Rosario-Raymundo, S.M. Garland,
prevention of human papillomavirus type 16- and 18-associated cervical cancer, A. Chatterjee, E. Lazcano-Ponce, J. Salmeron, S. McNeil, J.T. Stapleton,
Expert Opin. Biol. Ther. 7 (2007) 391–396. C. Bouchard, M.G. Martens, D.M. Money, S.C. Quek, B. Romanowski, C.S. Vallejos,
[74] A. Szarewski, HPV vaccine: Cervarix, Expert Opin. Biol. Ther. 10 (2010) 477–487. B. Ter Harmsel, V. Prilepskaya, K.L. Fong, H. Kitchener, G. Minkina, Y.K.T. Lim,
[75] Cervarix, https://www.fda.gov/vaccines-blood-biologics/vaccines/cervarix. T. Stoney, N. Chakhtoura, M.E. Cruickshank, A. Savicheva, D.P. da Silva,
[76] R. Kirnbauer, F. Booy, N. Cheng, D.R. Lowy, J.T. Schiller, Papillomavirus L1 major M. Ferguson, A.C. Molijn, W.G.V. Quint, K. Hardt, D. Descamps, P.V. Suryakiran,
capsid protein self-assembles into virus-like particles that are highly im- N. Karkada, B. Geeraerts, G. Dubin, F. Struyf, V.S. Group, Efficacy, safety, and
munogenic, Proc. Natl. Acad. Sci. U.S.A. 89 (1992) 12180–12184. immunogenicity of the human papillomavirus 16/18 AS04-adjuvanted vaccine in
[77] J.T. Schiller, D.R. Lowy, Immunogenicity testing in human papillomavirus virus- women older than 25 years: 7-year follow-up of the phase 3, double-blind, ran-
like-particle vaccine trials, J. Infect. Dis. 200 (2009) 166–171. domised controlled VIVIANE study, Lancet Infect. Dis. 16 (2016) 1154–1168.
[78] L.A. Pinto, J. Dillner, S. Beddows, E.R. Unger, Immunogenicity of HPV prophy- [94] S.R. Skinner, A. Szarewski, B. Romanowski, S.M. Garland, E. Lazcano-Ponce,
lactic vaccines: serology assays and their use in HPV vaccine evaluation and de- J. Salmeron, M.R. Del Rosario-Raymundo, R.H. Verheijen, S.C. Quek, D.P. da
velopment, Vaccine 36 (2018) 4792–4799. Silva, H. Kitchener, K.L. Fong, C. Bouchard, D.M. Money, A. Ilancheran,
[79] D.V. Pastrana, C.B. Buck, Y.Y. Pang, C.D. Thompson, P.E. Castle, P.C. FitzGerald, M.E. Cruickshank, M.J. Levin, A. Chatterjee, J.T. Stapleton, M. Martens, W. Quint,
S. Kruger Kjaer, D.R. Lowy, J.T. Schiller, Reactivity of human sera in a sensitive, M.P. David, D. Meric, K. Hardt, D. Descamps, B. Geeraerts, F. Struyf, G. Dubin,
high-throughput pseudovirus-based papillomavirus neutralization assay for V.S. Group, Efficacy, safety, and immunogenicity of the human papillomavirus 16/
HPV16 and HPV18, Virology 321 (2004) 205–216. 18 AS04-adjuvanted vaccine in women older than 25 years: 4-year interim follow-
[80] D. Opalka, C.E. Lachman, S.A. MacMullen, K.U. Jansen, J.F. Smith, N. Chirmule, up of the phase 3, double-blind, randomised controlled VIVIANE study, Lancet 384
M.T. Esser, Simultaneous quantitation of antibodies to neutralizing epitopes on (2014) 2213–2227.
virus-like particles for human papillomavirus types 6, 11, 16, and 18 by a multi- [95] E.A. Joura, A.R. Giuliano, O.E. Iversen, C. Bouchard, C. Mao, J. Mehlsen,
plexed luminex assay, Clin. Diagn. Lab. Immunol. 10 (2003) 108–115. E.D. Moreira Jr., Y. Ngan, L.K. Petersen, E. Lazcano-Ponce, P. Pitisuttithum,
[81] R. Kirnbauer, N.L. Hubbert, C.M. Wheeler, T.M. Becker, D.R. Lowy, J.T. Schiller, A J.A. Restrepo, G. Stuart, L. Woelber, Y.C. Yang, J. Cuzick, S.M. Garland, W. Huh,
virus-like particle enzyme-linked immunosorbent assay detects serum antibodies S.K. Kjaer, O.M. Bautista, I.S. Chan, J. Chen, R. Gesser, E. Moeller, M. Ritter,
in a majority of women infected with human papillomavirus type 16, J. Nat. S. Vuocolo, A. Luxembourg, H.P.V.V.S. Broad Spectrum, A 9-valent HPV vaccine
Cancer. Inst. 86 (1994) 494–499. against infection and intraepithelial neoplasia in women, N. Engl. J. Med. 372
[82] Human papillomavirus vaccines: WHO position paper, May 2017, Releve (2015) 711–723.
Epidemiologique Hebd. 92 (2017) 241–268. [96] W.K. Huh, E.A. Joura, A.R. Giuliano, O.E. Iversen, R.P. de Andrade, K.A. Ault,
[83] D. Ferris, R. Samakoses, S.L. Block, E. Lazcano-Ponce, J.A. Restrepo, D. Bartholomew, R.M. Cestero, E.N. Fedrizzi, A.L. Hirschberg, M.H. Mayrand,
K.S. Reisinger, J. Mehlsen, A. Chatterjee, O.E. Iversen, H.L. Sings, Q. Shou, A.M. Ruiz-Sternberg, J.T. Stapleton, D.J. Wiley, A. Ferenczy, R. Kurman,
T.A. Sausser, A. Saah, Long-term study of a quadrivalent human papillomavirus B.M. Ronnett, M.H. Stoler, J. Cuzick, S.M. Garland, S.K. Kjaer, O.M. Bautista,
vaccine, Pediatrics 134 (2014) e657–665. R. Haupt, E. Moeller, M. Ritter, C.C. Roberts, C. Shields, A. Luxembourg, Final
[84] P.S. Naud, C.M. Roteli-Martins, N.S. De Carvalho, J.C. Teixeira, P.C. de Borba, efficacy, immunogenicity, and safety analyses of a nine-valent human papillo-
N. Sanchez, T. Zahaf, G. Catteau, B. Geeraerts, D. Descamps, Sustained efficacy, mavirus vaccine in women aged 16-26 years: a randomised, double-blind trial,
immunogenicity, and safety of the HPV-16/18 AS04-adjuvanted vaccine: final Lancet 390 (2017) 2143–2159.
analysis of a long-term follow-up study up to 9.4 years post-vaccination, Hum. [97] R. Luckett, S. Feldman, Impact of 2-, 4- and 9-valent HPV vaccines on morbidity
Vaccines Immunother. 10 (2014) 2147–2162. and mortality from cervical cancer, Hum. Vaccines Immunother. 12 (2015)
[85] M. Nygard, A. Saah, C. Munk, L. Tryggvadottir, E. Enerly, M. Hortlund, 1332–1342.
L.G. Sigurdardottir, S. Vuocolo, S.K. Kjaer, J. Dillner, Evaluation of the long-term [98] T.J. Kemp, A. Hildesheim, M. Safaeian, J.G. Dauner, Y. Pan, C. Porras,
anti-human papillomavirus 6 (HPV6), 11, 16, and 18 immune responses generated J.T. Schiller, D.R. Lowy, R. Herrero, L.A. Pinto, HPV16/18 L1 VLP vaccine induces
by the quadrivalent HPV vaccine, Clin. Vaccine Immunol. 22 (2015) 943–948. cross-neutralizing antibodies that may mediate cross-protection, Vaccine 29
[86] A. Guevara, R. Cabello, L. Woelber, E.D. Moreira Jr., E. Joura, O. Reich, C. Shields, (2011) 2011–2014.
M.C. Ellison, A. Joshi, A. Luxembourg, Antibody persistence and evidence of im- [99] T. Malagon, M. Drolet, M.C. Boily, E.L. Franco, M. Jit, J. Brisson, M. Brisson, Cross-
mune memory at 5years following administration of the 9-valent HPV vaccine, protective efficacy of two human papillomavirus vaccines: a systematic review and
Vaccine 35 (2017) 5050–5057. meta-analysis, Lancet Infect. Dis. 12 (2012) 781–789.
[87] F.X. Bosch, C. Robles, M. Diaz, M. Arbyn, I. Baussano, C. Clavel, G. Ronco, [100] K.A. Lang Kuhs, C. Porras, J.T. Schiller, A.C. Rodriguez, M. Schiffman, P. Gonzalez,
J. Dillner, M. Lehtinen, K.U. Petry, M. Poljak, S.K. Kjaer, C.J. Meijer, S.M. Garland, S. Wacholder, A. Ghosh, Y. Li, D.R. Lowy, A.R. Kreimer, S. Poncelet, J. Schussler,
J. Salmeron, X. Castellsague, L. Bruni, S. de Sanjose, J. Cuzick, HPV-FASTER: W. Quint, L.J. van Doorn, M.E. Sherman, M. Sidawy, R. Herrero, A. Hildesheim,
broadening the scope for prevention of HPV-related cancer, Nature reviews, Clin. M. Safaeian, G. Costa, Rica Vaccine Trial, Effect of different human papillomavirus
Oncol. 13 (2016) 119–132. serological and DNA criteria on vaccine efficacy estimates, Am. J. Epidemiol. 180
[88] S.M. Garland, S.K. Kjaer, N. Munoz, S.L. Block, D.R. Brown, M.J. DiNubile, (2014) 599–607.
B.R. Lindsay, B.J. Kuter, G. Perez, G. Dominiak-Felden, A.J. Saah, R. Drury, R. Das, [101] A.R. Kreimer, A.C. Rodriguez, A. Hildesheim, R. Herrero, C. Porras, M. Schiffman,
C. Velicer, Impact and effectiveness of the quadrivalent human papillomavirus P. Gonzalez, D. Solomon, S. Jimenez, J.T. Schiller, D.R. Lowy, W. Quint,
vaccine: a systematic review of 10 Years of real-world experience, Clin. Infect. Dis. M.E. Sherman, J. Schussler, S. Wacholder, C.V.T.V. Group, Proof-of-principle
: Off. Publ. Infect. Dis. Soc. Am. 63 (2016) 519–527. evaluation of the efficacy of fewer than three doses of a bivalent HPV16/18
[89] S.M. Garland, J. Paavonen, U. Jaisamrarn, P. Naud, J. Salmeron, S.N. Chow, vaccine, J. Nat. Cancer. Inst. 103 (2011) 1444–1451.
D. Apter, X. Castellsague, J.C. Teixeira, S.R. Skinner, J. Hedrick, G. Limson, [102] T.F. Leung, A.P. Liu, F.S. Lim, F. Thollot, H.M.L. Oh, B.W. Lee, L. Rombo, N.C. Tan,
T.F. Schwarz, W.A. Poppe, F.X. Bosch, N.S. de Carvalho, M.J. Germar, K. Peters, R. Rouzier, S. De Simoni, P. Suryakiran, M. Hezareh, F. Thomas, N. Folschweiller,
M.R. Del Rosario-Raymundo, G. Catteau, D. Descamps, F. Struyf, M. Lehtinen, F. Struyf, Comparative immunogenicity and safety of human papillomavirus
G. Dubin, Prior human papillomavirus-16/18 AS04-adjuvanted vaccination pre- (HPV)-16/18 AS04-adjuvanted vaccine and 4vHPV vaccine administered ac-
vents recurrent high grade cervical intraepithelial neoplasia after definitive sur- cording to two- or three-dose schedules in girls aged 9-14years: results to month
gical therapy: post-hoc analysis from a randomized controlled trial, Int. J. Cancer 36 from a randomized trial, Vaccine 36 (2018) 98–106.
139 (2016) 2812–2826. [103] M. Jit, M. Brisson, J.F. Laprise, Y.H. Choi, Comparison of two dose and three dose
[90] J. Paavonen, P. Naud, J. Salmeron, C.M. Wheeler, S.N. Chow, D. Apter, human papillomavirus vaccine schedules: cost effectiveness analysis based on
H. Kitchener, X. Castellsague, J.C. Teixeira, S.R. Skinner, J. Hedrick, transmission model, BMJ 350 (2015) g7584.
U. Jaisamrarn, G. Limson, S. Garland, A. Szarewski, B. Romanowski, F.Y. Aoki, [104] s.w.i. World Health Organization, Electronic address, Human papillomavirus
T.F. Schwarz, W.A. Poppe, F.X. Bosch, D. Jenkins, K. Hardt, T. Zahaf, D. Descamps, vaccines: WHO position paper, May 2017-Recommendations, Vaccine 35 (2017)
F. Struyf, M. Lehtinen, G. Dubin, H.P.S. Group, Efficacy of human papillomavirus 5753–5755.
(HPV)-16/18 AS04-adjuvanted vaccine against cervical infection and precancer [105] S.R. Dobson, S. McNeil, M. Dionne, M. Dawar, G. Ogilvie, M. Krajden,
caused by oncogenic HPV types (PATRICIA): final analysis of a double-blind, C. Sauvageau, D.W. Scheifele, T.R. Kollmann, S.A. Halperin, J.M. Langley,
randomised study in young women, Lancet 374 (2009) 301–314. J.A. Bettinger, J. Singer, D. Money, D. Miller, M. Naus, F. Marra, E. Young,
[91] C.M. Wheeler, X. Castellsague, S.M. Garland, A. Szarewski, J. Paavonen, P. Naud, Immunogenicity of 2 doses of HPV vaccine in younger adolescents vs 3 doses in

99
R. Wang, et al. Cancer Letters 471 (2020) 88–102

young women: a randomized clinical trial, J. Am. Med. Assoc. 309 (2013) 88 (2003) 63–73.
1793–1802. [127] J.W. Wang, R.B. Roden, L2, the minor capsid protein of papillomavirus, Virology
[106] T. Puthanakit, L.M. Huang, C.H. Chiu, R.B. Tang, T.F. Schwarz, S. Esposito, 445 (2013) 175–186.
L. Frenette, C. Giaquinto, S. McNeil, P. Rheault, P. Durando, M. Horn, M. Klar, [128] C.B. Buck, N. Cheng, C.D. Thompson, D.R. Lowy, A.C. Steven, J.T. Schiller,
S. Poncelet, S. De Simoni, D. Friel, B. De Muynck, P.V. Suryakiran, M. Hezareh, B.L. Trus, Arrangement of L2 within the papillomavirus capsid, J. Virol. 82 (2008)
D. Descamps, F. Thomas, F. Struyf, Randomized open trial comparing 2-dose re- 5190–5197.
gimens of the human papillomavirus 16/18 AS04-adjuvanted vaccine in girls aged [129] R.B. Roden, W.H.t. Yutzy, R. Fallon, S. Inglis, D.R. Lowy, J.T. Schiller, Minor
9-14 Years versus a 3-dose regimen in women aged 15-25 years, J. Infect. Dis. 214 capsid protein of human genital papillomaviruses contains subdominant, cross-
(2016) 525–536. neutralizing epitopes, Virology 270 (2000) 254–257.
[107] B. Romanowski, T.F. Schwarz, L. Ferguson, K. Peters, M. Dionne, U. Behre, [130] E. Tumban, J. Peabody, D.S. Peabody, B. Chackerian, A universal virus-like par-
K. Schulze, P. Hillemanns, P. Suryakiran, F. Thomas, F. Struyf, Sustained im- ticle-based vaccine for human papillomavirus: longevity of protection and role of
munogenicity of the HPV-16/18 AS04-adjuvanted vaccine administered as a two- endogenous and exogenous adjuvants, Vaccine 31 (2013) 4647–4654.
dose schedule in adolescent girls: five-year clinical data and modeling predictions [131] K. Kalnin, S. Chivukula, T. Tibbitts, Y. Yan, S. Stegalkina, L. Shen, J. Cieszynski,
from a randomized study, Hum. Vaccines Immunother. 12 (2016) 20–29. V. Costa, R. Sabharwal, S.F. Anderson, N. Christensen, S. Jagu, R.B.S. Roden,
[108] R. Sankaranarayanan, P.R. Prabhu, M. Pawlita, T. Gheit, N. Bhatla, R. Muwonge, H. Kleanthous, Incorporation of RG1 epitope concatemers into a self-adjuvanting
B.M. Nene, P.O. Esmy, S. Joshi, U.R. Poli, P. Jivarajani, Y. Verma, E. Zomawia, Flagellin-L2 vaccine broaden durable protection against cutaneous challenge with
M. Siddiqi, S.S. Shastri, K. Jayant, S.G. Malvi, E. Lucas, A. Michel, J. Butt, diverse human papillomavirus genotypes, Vaccine 35 (2017) 4942–4951.
J.M. Vijayamma, S. Sankaran, T.P. Kannan, R. Varghese, U. Divate, S. Thomas, [132] T. Kanda, K. Kondo, Development of an HPV vaccine for a broad spectrum of high-
G. Joshi, M. Willhauck-Fleckenstein, T. Waterboer, M. Muller, P. Sehr, risk types, Hum. Vaccine 5 (2009) 43–45.
S. Hingmire, A. Kriplani, G. Mishra, S. Pimple, R. Jadhav, C. Sauvaget, [133] B. Huber, C. Schellenbacher, S. Shafti-Keramat, C. Jindra, N. Christensen,
M. Tommasino, M.R. Pillai, Immunogenicity and HPV infection after one, two, and R. Kirnbauer, Chimeric L2-based virus-like particle (VLP) vaccines targeting cu-
three doses of quadrivalent HPV vaccine in girls in India: a multicentre prospective taneous human papillomaviruses (HPV), PLoS One 12 (2017) e0169533.
cohort study, Lancet Oncol. 17 (2016) 67–77. [134] D. Wang, Z. Li, J. Xiao, J. Wang, L. Zhang, Y. Liu, F. Fan, L. Xin, M. Wei, Z. Kong,
[109] A.R. Kreimer, F. Struyf, M.R. Del Rosario-Raymundo, A. Hildesheim, S.R. Skinner, H. Yu, Y. Gu, J. Zhang, S. Li, N. Xia, Identification of broad-genotype HPV L2
S. Wacholder, S.M. Garland, R. Herrero, M.P. David, C.M. Wheeler, A. Costa Rica neutralization site for pan-HPV vaccine development by a cross-neutralizing an-
Vaccine Trial Study Group, P. Gonzalez, S. Jimenez, D.R. Lowy, L.A. Pinto, tibody, PLoS One 10 (2015) e0123944.
C. Porras, A.C. Rodriguez, M. Safaeian, M. Schiffman, J.T. Schiller, J. Schussler, [135] E. Medina, C.A. Guzman, Use of live bacterial vaccine vectors for antigen delivery:
M.E. Sherman, P.S.G. Authors, F.X. Bosch, X. Castellsague, A. Chatterjee, potential and limitations, Vaccine 19 (2001) 1573–1580.
S.N. Chow, D. Descamps, F. Diaz-Mitoma, G. Dubin, M.J. Germar, D.M. Harper, [136] M. Saxena, T.T. Van, F.J. Baird, P.J. Coloe, P.M. Smooker, Pre-existing immunity
D.J. Lewis, G. Limson, P. Naud, K. Peters, W.A. Poppe, B. Ramjattan, against vaccine vectors–friend or foe? Microbiology (Read.) 159 (2013) 1–11.
B. Romanowski, J. Salmeron, T.F. Schwarz, J.C. Teixeira, W.A. Tjalma, Efficacy of [137] G. Dietrich, S. Spreng, D. Favre, J.F. Viret, C.A. Guzman, Live attenuated bacteria
fewer than three doses of an HPV-16/18 AS04-adjuvanted vaccine: combined as vectors to deliver plasmid DNA vaccines, Curr. Opin. Mol. Ther. 5 (2003)
analysis of data from the Costa Rica Vaccine and PATRICIA Trials, Lancet Oncol. 10–19.
16 (2015) 775–786. [138] C. Schoen, D.I. Loeffler, A. Frentzen, S. Pilgrim, W. Goebel, J. Stritzker, Listeria
[110] Human Papillomavirus (HPV) Vaccine Safety, https://www.cdc.gov/ monocytogenes as novel carrier system for the development of live vaccines, Int. J.
vaccinesafety/vaccines/hpv-vaccine.html. Med. Microbiol. 298 (2008) 45–58.
[111] T.A. Suragh, P. Lewis, J. Arana, A. Mba-Jonas, R. Li, B. Stewart, T.T. Shimabukuro, [139] P. Guirnalda, L. Wood, Y. Paterson, Listeria monocytogenes and its products as
M. Cano, Safety of bivalent human papillomavirus vaccine in the US vaccine ad- agents for cancer immunotherapy, Adv. Immunol. 113 (2012) 81–118.
verse event reporting system (VAERS), 2009-2017, Br. J. Clin. Pharmacol. 84 [140] P. Schnupf, D.A. Portnoy, Listeriolysin O: a phagosome-specific lysin, Microb.
(2018) 2928–2932. Infect. 9 (2007) 1176–1187.
[112] Y. Jia, C. Zhu, J. Du, Y. Xiang, Y. Chen, W. Wang, C. Tao, Investigating safety [141] C. Watts, The exogenous pathway for antigen presentation on major histo-
profiles of human papillomavirus vaccine across group differences using VAERS compatibility complex class II and CD1 molecules, Nat. Immunol. 5 (2004)
data and MedDRA, PeerJ 7 (2019) e7490. 685–692.
[113] C. Scavone, C. Di Mauro, S. Brusco, M. Bertini, G. di Mauro, C. Rafaniello, [142] A. Wallecha, C. French, R. Petit, R. Singh, A. Amin, J. Rothman, Lm-LLO-based
L. Sportiello, F. Rossi, A. Capuano, Surveillance of adverse events following im- immunotherapies and HPV-associated disease, J. Oncol. 2012 (2012) 542851.
munization related to human papillomavirus vaccines: 12 years of vaccinovigi- [143] P.C. Maciag, S. Radulovic, J. Rothman, The first clinical use of a live-attenuated
lance in Southern Italy, Expert Opin. Drug Saf. 18 (2019) 427–433. Listeria monocytogenes vaccine: a Phase I safety study of Lm-LLO-E7 in patients
[114] D. Ward, N.M. Thorsen, M. Frisch, P. Valentiner-Branth, K. Molbak, A. Hviid, A with advanced carcinoma of the cervix, Vaccine 27 (2009) 3975–3983.
cluster analysis of serious adverse event reports after human papillomavirus (HPV) [144] P. Basu, A.O. Mehta, M.M. Jain, S. Gupta, R.V. Nagarkar, V. Kumar, S. Premkumar,
vaccination in Danish girls and young women, September 2009 to August 2017, R. Neve, S. John, R.G. Petit, ADXS11-001 immunotherapy targeting HPV-E7: final
Euro Surveill. 24 (2019) 1800380. results from a phase 2 study in Indian women with recurrent cervical cancer, J.
[115] H.I. Satari, J. Sundoro, A. Andrijono, S.R. Hadinegoro, S. Syafriyal, G. Tandy, Clin. Oncol. 32 (2014) 5610-5610.
S. Karolina, Post marketing surveillance study of 2nd dose quadrivalent human [145] K. Kawana, K. Adachi, S. Kojima, A. Taguchi, K. Tomio, A. Yamashita, H. Nishida,
papilloma virus vaccine in elementary school children in Jakarta, Indonesia: safety K. Nagasaka, T. Arimoto, T. Yokoyama, O. Wada-Hiraike, K. Oda, T. Sewaki,
result and implementation of school-based HPV immunization program, Asian Y. Osuga, T. Fujii, Oral vaccination against HPV E7 for treatment of cervical in-
Pac. J. Cancer Prev. APJCP : Asian Pac. J. Cancer Prev. APJCP 20 (2019) 869–875. traepithelial neoplasia grade 3 (CIN3) elicits E7-specific mucosal immunity in the
[116] M.P. Pereira, P. Flores, A.S. Neto, Neck and supraclavicular lymphadenopathy cervix of CIN3 patients, Vaccine 32 (2014) 6233–6239.
secondary to 9-valent human papillomavirus vaccination, BMJ Case Rep. 12 [146] A. Komatsu, S. Igimi, K. Kawana, Optimization of human papillomavirus (HPV)
(2019) e231582. type 16 E7-expressing lactobacillus-based vaccine for induction of mucosal E7-
[117] J.Y. Chung, S.J. Lee, B.S. Shin, H.G. Kang, Myasthenia gravis following human specific IFNgamma-producing cells, Vaccine 36 (2018) 3423–3426.
papillomavirus vaccination: a case report, BMC Neurol. 18 (2018) 222. [147] M. Santosuosso, S. McCormick, Z. Xing, Adenoviral vectors for mucosal vaccina-
[118] K. Zacharis, C.I. Messini, G. Anifandis, G. Koukoulis, M. Satra, A. Daponte, Human tion against infectious diseases, Viral Immunol. 18 (2005) 283–291.
papilloma virus (HPV) and fertilization: a mini review, Medicina 54 (2018) 50. [148] V. Cerullo, M.P. Seiler, V. Mane, N. Brunetti-Pierri, C. Clarke, T.K. Bertin,
[119] U. Bonde, J.S. Joergensen, R.F. Lamont, O. Mogensen, Is HPV vaccination in J.R. Rodgers, B. Lee, Toll-like receptor 9 triggers an innate immune response to
pregnancy safe? Hum. Vaccines Immunother. 12 (2016) 1960–1964. helper-dependent adenoviral vectors, Mol. Ther. 15 (2007) 378–385.
[120] E.O. Kharbanda, G. Vazquez-Benitez, H.S. Lipkind, S.S. Sheth, J. Zhu, [149] Z.C. Hartman, E.P. Black, A. Amalfitano, Adenoviral infection induces a multi-
A.L. Naleway, N.P. Klein, R. Hechter, M.F. Daley, J.G. Donahue, M.L. Jackson, faceted innate cellular immune response that is mediated by the toll-like receptor
A.T. Kawai, L. Sukumaran, J.D. Nordin, Risk of spontaneous abortion after inad- pathway in A549 cells, Virology 358 (2007) 357–372.
vertent human papillomavirus vaccination in pregnancy, Obstet. Gynecol. 132 [150] K.J. Ewer, T. Lambe, C.S. Rollier, A.J. Spencer, A.V. Hill, L. Dorrell, Viral vectors
(2018) 35–44. as vaccine platforms: from immunogenicity to impact, Curr. Opin. Immunol. 41
[121] A. Wang, C. Liu, Y. Wang, A. Yin, J. Wu, C. Zhang, M. Luo, L. Du, Y. Xiong, (2016) 47–54.
X. Zhao, Y. Huang, Pregnancy Outcomes after Human Papillomavirus Vaccination [151] E. Lopez-Gordo, I.I. Podgorski, N. Downes, R. Alemany, Circumventing antivector
in Periconceptional Period or during Pregnancy: A Systematic Review and Meta- immunity: potential use of nonhuman adenoviral vectors, Hum. Gene Ther. 25
Analysis, Human vaccines & immunotherapeutics, 2019, pp. 1–9. (2014) 285–300.
[122] L.D. Wise, J.J. Wolf, L.M. Plitnick, Evaluation of a 9-valent HPV vaccine in [152] S. Khan, K. Oosterhuis, K. Wunderlich, E.M. Bunnik, M. Bhaggoe, S. Boedhoe,
Sprague-Dawley rats: nonclinical studies assessing general, reproductive, and de- S. Karia, R.D.M. Steenbergen, L. Bosch, J. Serroyen, S. Janssen, H. Schuitemaker,
velopmental toxicity, Vaccine 36 (2018) 6401–6407. J. Vellinga, G. Scheper, R. Zahn, J. Custers, Development of a replication-deficient
[123] D.M. Parkin, F. Bray, Chapter 2: the burden of HPV-related cancers, Vaccine 24 adenoviral vector-based vaccine candidate for the interception of HPV16- and
(Suppl 3) (2006) S3/11-25. HPV18-induced infections and disease, Int. J. Cancer 141 (2017) 393–404.
[124] K.J. Hassett, N.M. Meinerz, F. Semmelmann, M.C. Cousins, R.L. Garcea, [153] N. Cuburu, S. Khan, C.D. Thompson, R. Kim, J. Vellinga, R. Zahn, D.R. Lowy,
T.W. Randolph, Development of a highly thermostable, adjuvanted human pa- G. Scheper, J.T. Schiller, Adenovirus vector-based prime-boost vaccination via
pillomavirus vaccine, Eur. J. Pharm. Biopharm. 94 (2015) 220–228. heterologous routes induces cervicovaginal CD8(+) T cell responses against
[125] R. Herrero, P. Gonzalez, L.E. Markowitz, Present status of human papillomavirus HPV16 oncoproteins, Int. J. Cancer 142 (2018) 1467–1479.
vaccine development and implementation, Lancet Oncol. 16 (2015) e206–216. [154] A. Volz, G. Sutter, Modified vaccinia virus Ankara: history, value in basic research,
[126] G.M. Clifford, J.S. Smith, M. Plummer, N. Munoz, S. Franceschi, Human papillo- and current perspectives for vaccine development, Adv. Virus Res. 97 (2017)
mavirus types in invasive cervical cancer worldwide: a meta-analysis, Br. J. Canc. 187–243.

100
R. Wang, et al. Cancer Letters 471 (2020) 88–102

[155] R. Alcock, M.G. Cottingham, C.S. Rollier, J. Furze, S.D. De Costa, M. Hanlon, vaccination therapy of patients with advanced or recurrent HPV16-induced gy-
A.J. Spencer, J.D. Honeycutt, D.H. Wyllie, S.C. Gilbert, M. Bregu, A.V. Hill, Long- necological carcinoma, a phase II trial, J. Transl. Med. 11 (2013) 88.
term thermostabilization of live poxviral and adenoviral vaccine vectors at su- [178] J. Banchereau, R.M. Steinman, Dendritic cells and the control of immunity, Nature
praphysiological temperatures in carbohydrate glass, Sci. Transl. Med. 2 (2010) 392 (1998) 245–252.
19ra12. [179] A. de Jong, T. O'Neill, A.Y. Khan, K.M. Kwappenberg, S.E. Chisholm, N.R. Whittle,
[156] R. Rosales, M. Lopez-Contreras, C. Rosales, J.R. Magallanes-Molina, R. Gonzalez- J.A. Dobson, L.C. Jack, J.A. St Clair Roberts, R. Offringa, S.H. van der Burg,
Vergara, J.M. Arroyo-Cazarez, A. Ricardez-Arenas, A. Del Follo-Valencia, J.K. Hickling, Enhancement of human papillomavirus (HPV) type 16 E6 and E7-
S. Padilla-Arriaga, M.V. Guerrero, M.A. Pirez, C. Arellano-Fiore, F. Villarreal, specific T-cell immunity in healthy volunteers through vaccination with TA-CIN,
Regression of human papillomavirus intraepithelial lesions is induced by MVA E2 an HPV16 L2E7E6 fusion protein vaccine, Vaccine 20 (2002) 3456–3464.
therapeutic vaccine, Hum. Gene Ther. 25 (2014) 1035–1049. [180] E.J. Davidson, R.L. Faulkner, P. Sehr, M. Pawlita, L.J. Smyth, D.J. Burt,
[157] I. Delany, R. Rappuoli, E. De Gregorio, Vaccines for the 21st century, EMBO Mol. A.E. Tomlinson, J. Hickling, H.C. Kitchener, P.L. Stern, Effect of TA-CIN (HPV 16
Med. 6 (2014) 708–720. L2E6E7) booster immunisation in vulval intraepithelial neoplasia patients pre-
[158] B. Yang, J. Jeang, A. Yang, T.C. Wu, C.F. Hung, DNA vaccine for cancer im- viously vaccinated with TA-HPV (vaccinia virus encoding HPV 16/18 E6E7),
munotherapy, Hum. Vaccines Immunother. 10 (2014) 3153–3164. Vaccine 22 (2004) 2722–2729.
[159] S.J. Lee, A. Yang, T.C. Wu, C.F. Hung, Immunotherapy for human papillomavirus- [181] S. Daayana, E. Elkord, U. Winters, M. Pawlita, R. Roden, P.L. Stern, H.C. Kitchener,
associated disease and cervical cancer: review of clinical and translational re- Phase II trial of imiquimod and HPV therapeutic vaccination in patients with
search, J. Gynecol Oncol. 27 (2016) e51. vulval intraepithelial neoplasia, Br. J. Canc. 102 (2010) 1129–1136.
[160] P. Vici, L. Pizzuti, L. Mariani, G. Zampa, D. Santini, L. Di Lauro, T. Gamucci, [182] R.M. Steinman, Decisions about dendritic cells: past, present, and future, Annu.
C. Natoli, P. Marchetti, M. Barba, M. Maugeri-Sacca, D. Sergi, F. Tomao, E. Vizza, Rev. Immunol. 30 (2012) 1–22.
S. Di Filippo, F. Paolini, G. Curzio, G. Corrado, A. Michelotti, G. Sanguineti, [183] A.D. Santin, S. Bellone, J.J. Roman, A. Burnett, M.J. Cannon, S. Pecorelli,
A. Giordano, R. De Maria, A. Venuti, Targeting immune response with therapeutic Therapeutic vaccines for cervical cancer: dendritic cell-based immunotherapy,
vaccines in premalignant lesions and cervical cancer: hope or reality from clinical Curr. Pharmaceut. Des. 11 (2005) 3485–3500.
studies, Expert Rev. Vaccines 15 (2016) 1327–1336. [184] Y.H. Ahn, S.O. Hong, J.H. Kim, K.H. Noh, K.H. Song, Y.H. Lee, J.H. Jeon,
[161] B. Afrough, S. Dowall, R. Hewson, Emerging viruses and current strategies for D.W. Kim, J.H. Seo, T.W. Kim, The siRNA cocktail targeting interleukin 10 re-
vaccine intervention, Clin. Exp. Immunol. 196 (2019) 157–166. ceptor and transforming growth factor-beta receptor on dendritic cells potentiates
[162] S.W. Tsen, A.H. Paik, C.F. Hung, T.C. Wu, Enhancing DNA vaccine potency by tumour antigen-specific CD8(+) T cell immunity, Clin. Exp. Immunol. 181 (2015)
modifying the properties of antigen-presenting cells, Expert Rev. Vaccines 6 164–178.
(2007) 227–239. [185] J.H. Kim, T.H. Kang, K.H. Noh, H.C. Bae, S.H. Kim, Y.D. Yoo, S.Y. Seong,
[163] T.J. Kim, H.T. Jin, S.Y. Hur, H.G. Yang, Y.B. Seo, S.R. Hong, C.W. Lee, S. Kim, T.W. Kim, Enhancement of dendritic cell-based vaccine potency by anti-apoptotic
J.W. Woo, K.S. Park, Y.Y. Hwang, J. Park, I.H. Lee, K.T. Lim, K.H. Lee, M.S. Jeong, siRNAs targeting key pro-apoptotic proteins in cytotoxic CD8(+) T cell-mediated
C.D. Surh, Y.S. Suh, J.S. Park, Y.C. Sung, Clearance of persistent HPV infection and cell death, Immunol. Lett. 122 (2009) 58–67.
cervical lesion by therapeutic DNA vaccine in CIN3 patients, Nat. Commun. 5 [186] K. Palucka, J. Banchereau, Cancer immunotherapy via dendritic cells, Nature re-
(2014) 5317. views, Cancer 12 (2012) 265–277.
[164] C.L. Trimble, M.P. Morrow, K.A. Kraynyak, X. Shen, M. Dallas, J. Yan, L. Edwards, [187] O.E. Rahma, V.E. Herrin, R.A. Ibrahim, A. Toubaji, S. Bernstein, O. Dakheel,
R.L. Parker, L. Denny, M. Giffear, A.S. Brown, K. Marcozzi-Pierce, D. Shah, S.M. Steinberg, R. Abu Eid, M. Mkrtichyan, J.A. Berzofsky, S.N. Khleif, Pre-im-
A.M. Slager, A.J. Sylvester, A. Khan, K.E. Broderick, R.J. Juba, T.A. Herring, mature dendritic cells (PIDC) pulsed with HPV16 E6 or E7 peptide are capable of
J. Boyer, J. Lee, N.Y. Sardesai, D.B. Weiner, M.L. Bagarazzi, Safety, efficacy, and eliciting specific immune response in patients with advanced cervical cancer, J.
immunogenicity of VGX-3100, a therapeutic synthetic DNA vaccine targeting Transl. Med. 12 (2014) 353.
human papillomavirus 16 and 18 E6 and E7 proteins for cervical intraepithelial [188] E. Zsiros, T. Tsuji, K. Odunsi, Adoptive T-cell therapy is a promising salvage ap-
neoplasia 2/3: a randomised, double-blind, placebo-controlled phase 2b trial, proach for advanced or recurrent metastatic cervical cancer, J. Clin. Oncol. 33
Lancet 386 (2015) 2078–2088. (2015) 1521–1522.
[165] K. Lundstrom, RNA viruses as tools in gene therapy and vaccine development, [189] S. Stevanovic, L.M. Draper, M.M. Langhan, T.E. Campbell, M.L. Kwong,
Genes (2019) 10. J.R. Wunderlich, M.E. Dudley, J.C. Yang, R.M. Sherry, U.S. Kammula, N.P. Restifo,
[166] S.S. Diebold, T. Kaisho, H. Hemmi, S. Akira, C. Reis e Sousa, Innate antiviral re- S.A. Rosenberg, C.S. Hinrichs, Complete regression of metastatic cervical cancer
sponses by means of TLR7-mediated recognition of single-stranded RNA, Science after treatment with human papillomavirus-targeted tumor-infiltrating T cells, J.
(N.Y.) 303 (2004) 1529–1531. Clin. Oncol. 33 (2015) 1543–1550.
[167] K. Lundstrom, Self-replicating RNA viruses for RNA therapeutics, Molecules [190] S.L. Doran, S. Stevanovic, S. Adhikary, J.J. Gartner, L. Jia, M.L.M. Kwong,
(2018) 23. W.C. Faquin, S.M. Hewitt, R.M. Sherry, J.C. Yang, S.A. Rosenberg, C.S. Hinrichs, T-
[168] K. Lundstrom, Replicon RNA viral vectors as vaccines, Vaccines 4 (2016) 39. cell receptor gene therapy for human papillomavirus-associated epithelial cancers:
[169] A.N. Varnavski, P.R. Young, A.A. Khromykh, Stable high-level expression of het- a first-in-human, phase I/II study, J. Clin. Oncol. 37 (2019) 2759–2768.
erologous genes in vitro and in vivo by noncytopathic DNA-based Kunjin virus [191] C.A. Ramos, N. Narala, G.M. Vyas, A.M. Leen, U. Gerdemann, E.M. Sturgis,
replicon vectors, J. Virol. 74 (2000) 4394–4403. M.L. Anderson, B. Savoldo, H.E. Heslop, M.K. Brenner, C.M. Rooney, Human pa-
[170] C.F. Hung, T.C. Wu, A. Monie, R. Roden, Antigen-specific immunotherapy of pillomavirus type 16 E6/E7-specific cytotoxic T lymphocytes for adoptive im-
cervical and ovarian cancer, Immunol. Rev. 222 (2008) 43–69. munotherapy of HPV-associated malignancies, J. Immunother. 36 (2013) 66–76.
[171] Q. Zeng, S. Peng, A. Monie, M. Yang, X. Pang, C.F. Hung, T.C. Wu, Control of [192] A.R. Cantelmo, L.C. Conradi, A. Brajic, J. Goveia, J. Kalucka, A. Pircher,
cervicovaginal HPV-16 E7-expressing tumors by the combination of therapeutic P. Chaturvedi, J. Hol, B. Thienpont, L.A. Teuwen, S. Schoors, B. Boeckx, J. Vriens,
HPV vaccination and vascular disrupting agents, Hum. Gene Ther. 22 (2011) A. Kuchnio, K. Veys, B. Cruys, L. Finotto, L. Treps, T.E. Stav-Noraas, F. Bifari,
809–819. P. Stapor, I. Decimo, K. Kampen, K. De Bock, G. Haraldsen, L. Schoonjans,
[172] R. Paris, S. Bejrachandra, P. Thongcharoen, S. Nitayaphan, P. Pitisuttithum, T. Rabelink, G. Eelen, B. Ghesquiere, J. Rehman, D. Lambrechts, A.B. Malik,
A. Sambor, S. Gurunathan, D. Francis, S. Ratto-Kim, C. Karnasuta, M.S. de Souza, M. Dewerchin, P. Carmeliet, Inhibition of the glycolytic activator PFKFB3 in en-
V.R. Polonis, A.E. Brown, J.H. Kim, H.A. Stephens, A.V.E.G. Thai, HLA class II dothelium induces tumor vessel normalization, impairs metastasis, and improves
restriction of HIV-1 clade-specific neutralizing antibody responses in ethnic Thai chemotherapy, Cancer Cell 30 (2016) 968–985.
recipients of the RV144 prime-boost vaccine combination of ALVAC-HIV and [193] S.J. Kim, K.H. Jung, M.K. Son, J.H. Park, H.H. Yan, Z. Fang, Y.W. Kang, B. Han,
AIDSVAX((R)) B/E, Vaccine 30 (2012) 832–836. J.H. Lim, S.S. Hong, Tumor vessel normalization by the PI3K inhibitor HS-173
[173] A.M. Solares, I. Baladron, T. Ramos, C. Valenzuela, Z. Borbon, S. Fanjull, enhances drug delivery, Cancer Lett. 403 (2017) 339–353.
L. Gonzalez, D. Castillo, J. Esmir, M. Granadillo, A. Batte, A. Cintado, M. Ale, [194] P. Rous, J.W. Beard, Carcinomatous changes in virus-induced papillomas of the
M.E. Fernandez de Cossio, A. Ferrer, I. Torrens, P. Lopez-Saura, Safety and im- skin of the rabbit, PSEBM (Proc. Soc. Exp. Biol. Med.) 32 (1935) 578–580.
munogenicity of a human papillomavirus peptide vaccine (CIGB-228) in women [195] P. Rous, J.G. Kidd, The carcinogenic effect of a papilloma virus on the tarred skin
with high-grade cervical intraepithelial neoplasia: first-in-human, proof-of-con- of rabbits : I. Description of the phenomenon, J. Exp. Med. 67 (1938) 399–428.
cept trial, ISRN Obstet. Gynecol. 2011 (2011) 292951. [196] H. zur Hausen, W. Meinhof, W. Scheiber, G.W. Bornkamm, Attempts to detect
[174] E.E. Sercarz, E. Maverakis, Mhc-guided processing: binding of large antigen virus-secific DNA in human tumors. I. Nucleic acid hybridizations with com-
fragments, Nat. Rev. Immunol. 3 (2003) 621–629. plementary RNA of human wart virus, Int. J. Cancer 13 (1974) 650–656.
[175] H.N. Coleman, W.W. Greenfield, S.L. Stratton, R. Vaughn, A. Kieber, [197] H. zur Hausen, Condylomata acuminata and human genital cancer, Cancer Res. 36
A.M. Moerman-Herzog, H.J. Spencer, W.C. Hitt, C.M. Quick, L.F. Hutchins, (1976) 794.
S.G. Mackintosh, R.D. Edmondson, S.W. Erickson, M. Nakagawa, Human pa- [198] A. Meisels, R. Fortin, Condylomatous lesions of the cervix and vagina. I. Cytologic
pillomavirus type 16 viral load is decreased following a therapeutic vaccination, patterns, Acta Cytol. 20 (1976) 505–509.
Cancer Immunol. Immunother.(CII) 65 (2016) 563–573. [199] M. Durst, L. Gissmann, H. Ikenberg, H. zur Hausen, A papillomavirus DNA from a
[176] G.G. Kenter, M.J. Welters, A.R. Valentijn, M.J. Lowik, D.M. Berends-van der Meer, cervical carcinoma and its prevalence in cancer biopsy samples from different
A.P. Vloon, F. Essahsah, L.M. Fathers, R. Offringa, J.W. Drijfhout, A.R. Wafelman, geographic regions, Proc. Natl. Acad. Sci. U.S.A. 80 (1983) 3812–3815.
J. Oostendorp, G.J. Fleuren, S.H. van der Burg, C.J. Melief, Vaccination against [200] M. Boshart, L. Gissmann, H. Ikenberg, A. Kleinheinz, W. Scheurlen, H. zur Hausen,
HPV-16 oncoproteins for vulvar intraepithelial neoplasia, N. Engl. J. Med. 361 A new type of papillomavirus DNA, its presence in genital cancer biopsies and in
(2009) 1838–1847. cell lines derived from cervical cancer, EMBO J. 3 (1984) 1151–1157.
[177] M.I. van Poelgeest, M.J. Welters, E.M. van Esch, L.F. Stynenbosch, G. Kerpershoek, [201] E. Schwarz, U.K. Freese, L. Gissmann, W. Mayer, B. Roggenbuck, A. Stremlau,
E.L. van Persijn van Meerten, M. van den Hende, M.J. Lowik, D.M. Berends-van H. zur Hausen, Structure and transcription of human papillomavirus sequences in
der Meer, L.M. Fathers, A.R. Valentijn, J. Oostendorp, G.J. Fleuren, C.J. Melief, cervical carcinoma cells, Nature 314 (1985) 111–114.
G.G. Kenter, S.H. van der Burg, HPV16 synthetic long peptide (HPV16-SLP) [202] C. Yee, I. Krishnan-Hewlett, C.C. Baker, R. Schlegel, P.M. Howley, Presence and

101
R. Wang, et al. Cancer Letters 471 (2020) 88–102

expression of human papillomavirus sequences in human cervical carcinoma cell and without abnormal cervical cytology, Lancet 2 (1987) 703–706.
lines, Am. J. Pathol. 119 (1985) 361–366. [207] J. Zhou, X.Y. Sun, D.J. Stenzel, I.H. Frazer, Expression of vaccinia recombinant
[203] B.A. Werness, A.J. Levine, P.M. Howley, Association of human papillomavirus HPV 16 L1 and L2 ORF proteins in epithelial cells is sufficient for assembly of HPV
types 16 and 18 E6 proteins with p53, Science (N.Y.) 248 (1990) 76–79. virion-like particles, Virology 185 (1991) 251–257.
[204] N. Dyson, P. Guida, K. Munger, E. Harlow, Homologous sequences in adenovirus [208] P.K. Magnusson, P. Sparen, U.B. Gyllensten, Genetic link to cervical tumours,
E1A and human papillomavirus E7 proteins mediate interaction with the same set Nature 400 (1999) 29–30.
of cellular proteins, J. Virol. 66 (1992) 6893–6902. [209] C.D. Harro, Y.Y. Pang, R.B. Roden, A. Hildesheim, Z. Wang, M.J. Reynolds,
[205] M. von Knebel Doeberitz, C. Rittmuller, H. zur Hausen, M. Durst, Inhibition of T.C. Mast, R. Robinson, B.R. Murphy, R.A. Karron, J. Dillner, J.T. Schiller,
tumorigenicity of cervical cancer cells in nude mice by HPV E6-E7 anti-sense RNA, D.R. Lowy, Safety and immunogenicity trial in adult volunteers of a human pa-
Int. J. Cancer 51 (1992) 831–834. pillomavirus 16 L1 virus-like particle vaccine, J. Nat. Cancer. Inst. 93 (2001)
[206] E.M. de Villiers, D. Wagner, A. Schneider, H. Wesch, H. Miklaw, J. Wahrendorf, 284–292.
U. Papendick, H. zur Hausen, Human papillomavirus infections in women with

102

You might also like