Next Article in Journal
Metabolism-Related Gene Expression in Circulating Tumor Cells from Patients with Early Stage Non-Small Cell Lung Cancer
Next Article in Special Issue
Impact of the Coronavirus Disease 2019 (COVID-19) Pandemic on Cervical Cancer Screening in Gynecological Practices in Germany
Previous Article in Journal
Screening a Targeted Panel of Genes by Next-Generation Sequencing Improves Risk Stratification in Real World Patients with Acute Myeloid Leukemia
Previous Article in Special Issue
Does the New FIGO 2018 Staging System Allow Better Prognostic Differentiation in Early Stage Cervical Cancer? A Dutch Nationwide Cohort Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gene Therapy for Malignant and Benign Gynaecological Disorders: A Systematic Review of an Emerging Success Story

by
Ekati Drakopoulou
1,
Nicholas P. Anagnou
1,* and
Kalliopi I. Pappa
1,2
1
Laboratory of Cell and Gene Therapy, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece
2
First Department of Obstetrics and Gynecology, University of Athens School of Medicine, 11528 Athens, Greece
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(13), 3238; https://doi.org/10.3390/cancers14133238
Submission received: 24 April 2022 / Revised: 21 June 2022 / Accepted: 27 June 2022 / Published: 30 June 2022
(This article belongs to the Special Issue Cervical Cancer Screening and Treatment in the 21st Century)

Abstract

:

Simple Summary

This review discusses all the major advances in gene therapy of gynaecological disorders, highlighting the novel and potentially therapeutic perspectives associated with such an approach. It specifically focuses on the gene therapy strategies against major gynaecological malignant disorders, such as ovarian, cervical, and endometrial cancer, as well as benign disorders, such as uterine leiomyomas, endometriosis, placental, and embryo implantation disorders. The above therapeutic strategies, which employ both viral and non-viral systems for mutation compensation, suicide gene therapy, oncolytic virotherapy, antiangiogenesis and immunopotentiation approaches, have yielded promising results over the last decade, setting the grounds for successful clinical trials.

Abstract

Despite the major advances in screening and therapeutic approaches, gynaecological malignancies still present as a leading cause of death among women of reproductive age. Cervical cancer, although largely preventable through vaccination and regular screening, remains the fourth most common and most lethal cancer type in women, while the available treatment schemes still pose a fertility threat. Ovarian cancer is associated with high morbidity rates, primarily due to lack of symptoms and high relapse rates following treatment, whereas endometrial cancer, although usually curable by surgery, it still represents a therapeutic problem. On the other hand, benign abnormalities, such as fibroids, endometriosis, placental, and embryo implantation disorders, although not life-threatening, significantly affect women’s life and fertility and have high socio-economic impacts. In the last decade, targeted gene therapy approaches toward both malignant and benign gynaecological abnormalities have led to promising results, setting the ground for successful clinical trials. The above therapeutic strategies employ both viral and non-viral systems for mutation compensation, suicide gene therapy, oncolytic virotherapy, antiangiogenesis and immunopotentiation. This review discusses all the major advances in gene therapy of gynaecological disorders and highlights the novel and potentially therapeutic perspectives associated with such an approach.

1. Introduction

The continuously growing amount of information regarding the molecular mechanisms underlying the emergence of tissue abnormalities from benign tumours to invasive malignancies has set the grounds for the use of gene therapy as a rational yet radical therapeutic approach for cancer therapy [1]. Gene therapy aims to introduce or modify genetic material into target cells, thus altering their function, usually by either restoring a lost function or initiating a new one. Although it was initially employed for the treatment of inherited genetic diseases, gene therapy was soon identified as an effective approach for the treatment of both gynaecological malignancies such as ovarian [2], cervical [3], and endometrial cancer [4] and certain benign gynaecological abnormalities, such as leiomyomas, endometriosis, placental, and embryo implantation disorders [5]. There are two main strategies for specific and efficient gene delivery to cancer and non-cancer cells, and these involve either viral or non-viral systems, as summarised in Figure 1 and described below in Section 2.
The main gene therapy strategies employ mutation compensation, primarily involving the replacement of a mutated tumour suppressor gene or the modification of aberrant gene expression in tumour cells, antiangiogenic, suicide gene therapy, and immunopotentiation approaches [5,6] (Figure 2). This review covers all major advances in gene therapy for gynaecological cancer and benign disorders.

2. Gene Delivery Systems

The main strategies for targeted gene therapy approaches for malignant and benign gynaecological disorders employ both viral and non-viral systems (Figure 1). This section provides a brief overview of the different viral and non-viral gene delivery systems.

2.1. Viral Systems

  • Retroviral vectors: Lentiviral vectors (LVs) are the most widely used retroviruses for therapeutic gene delivery, as they can efficiently transduce both quiescent and dividing cells and thus facilitate safe, efficient, and stable transgene expression [7,8]. They are enveloped, single-stranded RNA viruses with a packaging capacity of ~9 kb [7] and have already been employed in numerous successful clinical trials for the treatment of various diseases, such as hemoglobinopathies, metabolic and immune disorders, and various cancer types, including gynaecological malignancies [9]. Moreover, they have been instrumental in the development of chimeric antigen receptor (CAR) T cells, an approach with promising therapeutic perspectives in the field of oncology;
  • Adenoviral vectors (Ads): Adenoviral vectors have been extensively used as viral vector platforms primarily due to their broad tropism and the high transduction efficiency of both quiescent and dividing cells, as well as for their capacity to persist as episomal elements within the target cells [9]. They mainly derive from human serotypes-2 (Ad2) and -5 (Ad5) adenoviruses, which are non-enveloped, double-stranded DNA viruses with an icosahedral capsid able to accommodate up to 45 kb linear, double-stranded DNA [10]. However, since they are usually associated with potent immune responses [11] due to pre-existing immunity, extensive research has focused on generating less immunogenic Ad vectors, employing serotypes with low seroprevalence, such as Ad26 and Ad35 [12]. Furthermore, the generation of conditionally replicative adenoviral vectors (CRAds) by introducing tumour-specific promoters into the Ad genome can lead to higher gene expression [13]. Regarding gene therapy for malignant and benign gynaecological disorders, Ads have been extensively used as therapeutic vectors in both pre-clinical and clinical studies [9] for the delivery of vaccines, tumour suppressor genes, suicide genes, and immunomodulatory genes. All of these approaches are discussed in detail below;
  • Adeno-associated vectors (AAVs): Adeno-associated vectors are single-stranded DNA viruses with an icosahedral capsid of ~5 kb packaging capacity that depend on adenoviruses to complete their life cycle. The current AAV1 and AAV2 serotypes used are characterised by broad tropism, stable episomal persistence, and reduced immunogenicity, and therefore, they have been widely employed in gene therapy approaches [9]. Although AAV vectors have not been used yet in clinical gene therapy trials for gynaecological disorders, encouraging data from their pre-clinical assessment presented in this review point toward this direction;
  • Measles virus (MeV): The vaccine strain of MeV is a negative-strand RNA virus with a 16 kb-long genome that presents an attractive oncolytic platform, mainly due to its ability to selectively infect malignant cells, which overexpress the CD46 receptor [14]. Due to the fact that the monotherapy oncolytic approach using MeV is usually not sufficient to treat advanced-stage malignancies, combination approaches using radiotherapy or chemotherapy, MeV harbouring with therapeutic or immunomodulatory genes, and the use of carrier cells for MeV delivery into tumour cells can lead to successful therapeutic outcomes for a number of malignancies, including gynaecological cancers [15];
  • Herpes Simplex Virus (HSV): HSV-1 and HSV-2 members are enveloped, double-stranded DNA viruses with an icosahedral capsid, which demonstrate attractive vector features, such as a large accommodation capacity, easy production, and high titers [16]. Apart from their ability to mediate oncolysis upon delivery of the suicide thymidine kinase (TK) suicide gene into malignant cells, HSV vectors have also been used to deliver immunomodulatory cytokines into tumour cells and thus elicit a strong anti-tumour immune response. Extensive research has focused on the development of safer HSV vectors, carrying transcriptionally active promoter elements that overcome the latency of the viral genome, aiming to expand HSV pre-clinical and clinical applications [16].

2.2. Non-Viral Systems

The need for non-viral gene delivery systems arose primarily due to the immunogenicity and cytotoxicity concerns raised by some viral vectors, such as Adenoviruses [11]. Despite the relatively low transfection efficiency, their use in the field of gene therapy has gained a lot of ground [13,17]. The main non-viral approaches involve the delivery of the desired gene material, e.g., naked or plasmid DNA, either by physical methods, such as electroporation, ballistic DNA, injection, photoporation, magnetofection, sonoporation, and hydroporation [13,18], or chemically, by means of polymeric, lipid-based and inorganic nanoparticles (NPs) [13,19].

2.2.1. Physical Methods

Briefly, the most common types of non-viral physical methods involve the following approaches.
  • Electroporation: The electric pulse creates pores in the cell membrane, allowing the genetic material to enter the target cell. Depending on the target tissue, the electric pulse differs both in strength (high or low) and duration (short or long pulses), with cancer cells usually requiring low field strength (<700 V/cm) with long pulses (milliseconds) [13];
  • Ballistic DNA: Bombardment of DNA-coated heavy metal particles into target cells at a certain speed. The precision in DNA delivery makes it a method of choice for ovarian cancer [20];
  • Injection: The genetic material is directly injected into tissue by means of a needle. It represents the preferred approach for solid tumours; however, it has a relatively low efficiency [20];
  • Photoporation: A laser pulse causes cells to be permeable to DNA, with transfection efficiency being dependent on the focal point and laser frequency [13];
  • Magnetofection: It is mostly used for in vitro approaches; a gene-magnetic particle complex is introduced in cell culture, and electromagnets placed below the cell culture generate magnetic fields that mediate its effective sedimentation, thus leading to higher transfection efficiency [13];
  • Sonoporation: The generated ultrasound waves cause cell membranes to be permeable to micro-bubbles containing gene products [13];
  • Hydroporation: Hydrodynamic pressure generated by the injection of a large volume of DNA in a short period of time increases cell membrane permeability and mediates gene delivery [13].

2.2.2. Chemical Vectors

Non-viral chemical delivery systems employ particles of different shapes and sizes, known as nanoparticles (NPs), to encapsulate and efficiently transport genetic material, such as DNA and RNA, into target cells via endocytosis. These NPs are divided into the following three classes [13,19].
  • Polymeric NPs: These are stable, biodegradable, and water-soluble NPs, which make ideal candidates for drug delivery. They exist as monomers or polymers of different structures, with the most common being nanocapsules and nanospheres. According to shape, they are further divided into polymersomes, dendrimers, and micelles. Polymersomes are artificial vesicles with amphiphilic membranes that display increased cargo capacity and effective cytosol delivery [21], whereas micelles are quite effective for aqueous drug administration due to their hydrophilic core and hydrophobic coating. Polymeric micelles have already been employed in clinical trials, specifically in a study testing the effective delivery of paclitaxel (PTX), a drug widely used for ovarian cancer treatment [22]. Dendrimers are more complex, three-dimensional nanocarriers, commonly used for the delivery of nucleic acids and drugs in cancer therapy, usually in combination with poly amidoamine (PAMAM) and polyethylenimine (PEI) polymers [23]. Despite the toxicity associated with the surface amino groups, PAMAM was the first dendrimer used for gene delivery [17];
  • Lipid-based NPs: They are usually spherical vehicles with an internal aqueous compartment and an external lipid bilayer. They are divided into liposomes or lipoplexes, lipid emulsions (or nanogels), and lipid NPs [17]. Due to their positive charge, cationic liposomes bind to the anionic phosphate group of nucleic acids, forming lipoplexes and interacting with cell membranes, and thus, efficiently deliver nucleic acids into the cell [17]. Lipid emulsions, which are composed of oil, water, and a surface-active agent, show increased stability and serum resistance, whereas solid lipid NPs can protect nucleic acids from nucleases and are primarily employed for siRNA delivery [20];
  • Inorganic NPs: These nanoparticles are composed of inorganic materials, such as iron, gold, and silica, and therefore possess unique electrical, magnetic, and optical properties [13,19]. The most widely used inorganic NPs are gold NPs, primarily due to their low toxicity, while their photothermal properties establish them as good candidates for cancer therapy [24].

3. Gene Therapy for Gynaecological Cancers

3.1. Ovarian Cancer

Although usually considered a single disease, ovarian cancer is a highly heterogeneous group, comprising different histological subtypes with different underlying molecular mechanisms, clinical manifestations, and hence therapeutic approaches [25]. These include serous, endometrioid, clear-cell, and mucinous carcinoma [26] epithelial ovarian cancer types, which comprise the majority of ovarian cancers. Gene therapy is an emerging therapeutic approach for ovarian cancer [2] and is primarily based on viral and non-viral delivery systems (Figure 3) and on strategies such as suicide and antiangiogenic gene therapy, oncolytic virotherapy, mutation compensation, and immunopotentiation (Table 1).

3.1.1. Viral Vectors

The main viral vectors used for the gene therapy of ovarian cancer are summarized in Table 1 and Figure 3 and include retroviruses (primarily LVs), Ads, AAVs, HSV, and MeV. Highlighted below are the major advancers for each vector type.
Table 1. Targeted gene therapy approaches for ovarian cancer.
Table 1. Targeted gene therapy approaches for ovarian cancer.
Gene Therapy
Strategy
GeneDelivery SystemOutcomeModel/SpeciesReferences
Viral Vectors
Suicide gene therapyHSV-TKAdenoviral vectorEnhanced tumour inhibitory
effect, increased apoptosis and reduced micro-vessel density
in vitro (human)
in vivo (mouse)
[27,28]
Suicide gene therapyNTRAd-NTR/Ad-CB1954Increased survival in
ovarian-infected mouse models
in vitro (human)
in vivo (mouse)
[29]
Antiangiogenic
strategy
COL18A1
(endostatin)
Adenoviral vector
Ad-hEndo
Increase in apoptosis, tumour growth and vascularity reduction and prolonged survivalin vitro
(mouse/human)
in vivo (mouse)
[30,31]
Antiangiogenic
strategy
VEGFR2
Tie2
Adenoviral vector
Ad-VEGFR2/Ad-Tie2
Significant reduction in tumour weights and accumulation
of ascites
in vivo (mouse)[32]
Antiangiogenic
strategy
P125A-COL18A1Adeno-associated vector
rAAV
Inhibition of tumour growth and blood vessel formationin vivo (mouse)[33]
Antiangiogenic
strategy
kringle 5
(K5)
Adeno-associated vector
rAAV-K5
Inhibition of VEGF and tumour
angiogenesis
in vivo (mouse)[34]
Antiangiogenic
strategy
VEGF
inhibitor
Adeno-associated vector
AAVrh10.BevMab
Significant reduction in tumour growth, reduced angiogenesis, and increased survival in mouse modelin vivo (mouse)[35]
Mutation
compensation
Bim/BidAdenoviral vector
Ad-Bim, Ad-tBid
Strong anti-tumour effect and
tumour sensitization in cisplatin chemotherapy
following treatment with
Ad-tBid
in vitro (human)
in vivo (mouse)
[36]
Mutation
compensation
p53Adenoviral vector
rAd-53
p53-induced apoptosis and
taxol sensitization;
90% response rate and 100%
peritoneal effusion resolution
in vitro (human)
in vivo
(human/mouse)
[37,38]
Oncolytic virotherapyHER2/neu
EpCAM
Measles virus
(MeV)
Oncolytic potentialin vitro (human)
in vivo (mouse)
[39]
Oncolytic virotherapy
/Immunopotentiation
IL12Herpes simplex
virus
Cytotoxic effect, reduced
metastasis, increased CD8+
T-cell response
in vitro
(human/mouse)
in vivo (mouse)
[40]
Oncolytic virotherapy/
Immunopotentiation
GM-CSFHerpes simplex
virus
HF10
Reduction in tumour size and
increased anti-tumour immune response
in vitro (mouse)
in vivo (mouse)
[41]
Oncolytic virotherapyJak1/2
inhibitor
VSP-GPTransient tumour remissionin vitro (human)
in vivo (mouse)
[42]
Oncolytic virotherapySTAT3Adenoviral vector
M4
Inhibition of cell survival and
enhanced cisplatin-induced apoptosis
in vitro (human)
in vivo (mouse)
[43]
Oncolytic virotherapyZD55-MnSODAdenoviral vector
ZD55-MnSOD
Enhanced cisplatin-induced growth
suppression and apoptosis
in vitro (human)
in vivo (mouse)
[44]
Oncolytic virotherapyCD/5FUMeasles virus
MeV-SCD
Effective infection and lysis
of cancer cells
in vitro (human)[45]
ImmunopotentiationHER2/neuAdeno-associated vector
rAAV
Strong anti-tumour CTL
response
in vitro (human)[46]
ImmunopotentiationIL21Lentiviral vector
pUCMSCs-LV-IL-21
Reduction in tumour size by
inhibition of growth, elevation
of IFN-γ and TNF-α
in vitro (human)
in vivo (mouse)
[47]
Mutation
compensation
CD59Retroviral vector
shCD59
Enhanced complement-mediated cell damage, increased apoptosis and inhibition of tumour growthin vitro (human)
in vivo (mouse)
[48]
Mutation
compensation
NOB1Lentiviral vector
shRNA
Suppression of cancer cells growth and colony formationin vitro (human)[49]
Non-viral systems
Suicide gene therapyhEndoyCDFusion protein in plasmid vector (SV-
hEndoyCD)
Inhibition of tumour growth and
increased survival in xenograft mouse models
in vitro (human)
in vivo (mouse)
[50]
Suicide gene therapygelonin toxinHPEI nanogelsCancer cells growth reduction and induction of apoptosisin vitro (human)
in vivo (mouse)
[51]
Suicide gene therapyDT-ACationic polymerInhibition of tumour growth and prolonged lifespan in micein vitro (human)
in vivo (mouse)
[52,53]
Suicide gene therapyHSV-tTKTat/pDNA/C16TAB
nanoparticle
Increased targeted delivery in mouse model with human
ovarian cancer
in vitro human)
in vivo (mouse)
[54]
Suicide gene therapyVSV-MPF-LP/pMP
lipoplex
Inhibition of cancer cells
proliferation, induction of
apoptosis and suppression of
tumour angiogenesis
in vitro (human)
in vivo (mouse)
[55]
Suicide gene therapyCRB1PAMAM dendrimerIncreased survival in mice,
decreased proliferation, and
dissemination of cancer cells
in vitro (human)
in vivo (mouse)
[56]
ImmunopotentiationIL-21pIRES2-IL-21-EGFPReduction in tumour size,
increase in IFN-γ, TNF-α and
extension of survival
in vitro (human)
in vivo (mouse)
[57]
ImmunopotentiationpmIL-12pmIL-12/PPCElevation of IL-12 and IFN-γ,
significant decrease in VEGF
and survival improvement
in vitro (human)
in vivo (mouse)
[58]
Mutation
compensation
hPNAS-4cationic liposomeEfficient inhibition of growth,
induction of apoptosis,
inhibition of angiogenesis
in vitro (human)
in vivo (mouse)
[59]
Mutation
compensation
PTENPTEN plasmidApoptosis induction, G1 arrest, decrease in migration
and invasion
in vitro (human)[60]
Mutation
compensation
p16/
eEF1A2
pcDNA3.1-p16/
pcDNA3.1-vi5-hisA-eEF1A2 plasmids
Inhibition of cell growth by
downregulation of eEF1A2
in vitro (human)
in vivo (mouse)
[61,62]
Mutation
compensation
WWOXrecombinant plasmid/
pcDNA3.1-WWOX
Decreased proliferation, cell cycle arrest at G0/G1 phase, induction of apoptosis, inhibition of cell proliferation and autophagyin vitro (human)[63,64]
Mutation
compensation
WWOXpCMV-WWOX in liposomeDecreased ovarian cancer cell growth and induction
of apoptosis
in vitro (human)[65]
Mutation
compensation
EGFRsiRNA in nanogelsChemosensitisation to doxetacelin vitro (human)[66]
Mutation
compensation
MACC1shRNAInhibition of proliferation,
migration and invasion,
apoptosis induction
in vitro (human)[67]
Mutation
compensation
MTA1siRNAReduction in migration,
invasion, and adhesion,
induction of apoptosis
in vitro (human)[68]
Mutation
compensation
COX2siRNA/shRNAInhibition of cell proliferation, cell cycle arrest, reduced
invasion and migration,
growth suppression
in vitro (human)
in vivo (mouse)
[69,70]
Mutation
compensation
WT1ASODN in
liposomes
Inhibition of cell proliferation, cell cycle arrest, and increased
apoptosis
in vitro (human)[71]
Mutation
compensation
STAT3shRNA in liposomeInhibition of cell proliferation and induction of apoptosisin vitro (human)
in vivo (mouse)
[72]
Mutation
compensation
NOTCH1Cationic-cholesterol
derivative-based liposome
Inhibition of growth and
apoptosis induction
in vitro (human)[73]
Mutation
compensation
CLDN3siRNA in lipidoids
shRNA in nanoparticles
Inhibition of tumour growth,
reduction in cell proliferation and angiogenesis,
apoptosis induction
in vitro (human)
in vivo (mouse)
[74,75]
Mutation
compensation
HIF-1asiRNA with nanoparticlesEffective inhibition of
proliferation
in vitro (human)
in vivo (mouse)
[76]
Mutation
compensation
gDNMT1F-LP-delivered CRISPR/Cas9Inhibition of tumour growthin vitro (human)
in vivo (mouse)
[77]

Retroviruses

In a tumour-suppressor cervical-cancer gene therapy approach, Shi et al. employed shRNA to knock-down CD59, a negative regulator of complement activation, and demonstrated enhanced complement-mediated cell damage, increased apoptosis, and the inhibition of tumour growth in vitro and in vivo [48]. In another study, a lentiviral vector carrying shRNA for Nin one binding protein (NOB1p), a protein frequently overexpressed in ovarian cancer cells, led to the suppression of cancer cell growth and colony formation [49]. Lastly, Zhang et al. transplanted umbilical cord mesenchymal stem cells (MSCs) engineered to express IL-21 in SKOV3 ovarian cancer xenograft-bearing nude mice, utilising lentiviral vector pUCMSCs-LV-IL-21 and demonstrated a reduction in tumour size through the inhibition of growth and the elevation of IFN-γ and TNF-α [47].

Adenoviruses (Ads) and Adeno-Associated Viruses (AAVs)

These viral systems have been widely used in ovarian cancer gene therapy employed in suicide gene therapy, antiangiogenic, and oncolytic virotherapy, alone or together with immunopotentiation, as well as mutation compensation approaches. With regards to suicide gene therapy, Rawlinson et al. [27] developed an adenoviral vector carrying the HSV-TK gene under the tumour-specific promoter HE4 and demonstrated increased cell death by prodrug ganciclovir (GCV) treatment in vitro, in ovarian cancer cell lines. When Zhou et al. applied the above system in vivo in a murine ovarian cancer model, they observed an enhanced anti-tumour effect and increased apoptosis, as well as a reduction in micro-vessel density [28]. In a similar approach, but using the NTR-CB1954 system instead, White et al. performed an adenoviral delivery of both NTR and CB1954 in ovarian cancer cells and achieved increased survival in both in vitro and in vivo [29].
The antiangiogenic approach using Ads and AAVs has also yielded promising results in ovarian cancer, as demonstrated by several studies described in this section. Specifically, Tuppurainen et al. [32] used a combined strategy using adenoviral vectors expressing important genes in angiogenesis, such as vascular endothelial growth factor (VEGF) 2 receptor and angiopoietin growth factor receptor Tie2, with or without chemotherapy, and demonstrated reduced tumour growth and ascites formation. In a different approach, Hampl et al. [30] constructed the adenoviral vector, Ad-hEndo, and used it to mediate the transfer of human endostatin (COL18A1) in ovarian cancer ascites in mice, demonstrating a significant down-regulation in ascites formation, reduced tumour growth and vascularity, as well as prolonged animal survival [30]. In order to enhance transfection efficiency and escape antibody neutralisation in vivo, Yang et al. encapsulated the above vector into PEG-PE cationic liposomes and demonstrated the suppression of ovarian cancer growth in vivo, in ovarian cancer mouse models [31]. Similarly, the adeno-associated-mediated delivery of P125A- COL18A1, a mutated endostatin gene, led to the inhibition of tumour growth and blood vessel formation [33], while the administration of Kringle 5 (K5) of human plasminogen, a potent angiogenesis inhibitor, by means of a recombinant AAV vector led to the inhibition of VEGF and tumour angiogenesis [34]. Lastly, the use of AAVrh10.BevMab, a rhesus serotype 10 adeno-associated vector coding for bevacizumab, demonstrated a significant reduction in tumour growth, reduced angiogenesis, and increased survival in mice with ovarian cancer [35].
Regarding the oncolytic virotherapy strategy, apart from HSV, the Vesicular Stomatitis Virus (VSV), and MeV described below, adenoviruses have also been employed and led to significant anti-tumour effects. Specifically, Han et al. designed the novel oncolytic Ad-M4 targeting signal transducer and activator of transcription 3 (STAT3) in ovarian cancer and showed that the aberrant expression and constitutive activation of STAT3 were key factors in cisplatin resistance in both ovarian cancer cell lines and ovarian cancer tissues samples [43]. The authors showed significant and selective inhibition of ovarian cancer cell survival and cisplatin-induced apoptosis both in vitro and in vivo following Ad-M4 administration [43]. In a similar approach, Wang et al. constructed the manganese superoxide dismutase-armed oncolytic Ad ZD55-MnSOD and used it together with cisplatin, achieving enhanced cisplatin-induced growth suppression and apoptosis in ovarian cancer cells lines and ovarian tumour xenografts [44].
Furthermore, and regarding the mutational compensation approach, most studies utilising Ads target either the mutant p53, a protein with a variety of anti-cancer functions in gynaecological cancers [78], or key apoptosis pathway regulators, such as Bim and Bid. In the former approach, which made its way to Phase II/III clinical trials, the replacement of the altered p53 gene leads to significant anti-tumour effects [37,38]. Specifically, the in vitro and in vivo application of Ad-p53 resulted in the activation of apoptosis and taxol-sensitisation in ovarian cancer cells [38], while Gendicine, a gene therapy product approved for clinical use by the Chinese FDA in 2003, is now demonstrating very promising results, leading to a 90% patient response rate and 100% peritoneal effusion resolution [37]. In a recent study, Dai et al. [36] constructed two tumour-specific oncolytic adenoviruses coding for Bim (Ad-Bim) or truncated Bid (Ad-tBid) and performed gain-of-function assays in nine ovarian cancer cell lines. The authors observed increased cisplatin sensitisation and apoptosis following treatment with Ad-tBid in vitro and ex vivo, while following Ad-tBid intraperitoneal administration in mice with peritoneal disseminated ovarian cancer, they observed enhanced cisplatin-mediated anti-tumour effects [36]. Lastly, in regard to the immunopotentiation approach, HER2/neu, a protein shown to be overexpressed in ovarian cancer [79], was delivered to dendritic cells (DCs) by an AAV and primed these cells to express a self-tumour antigen. The above DC-loading elicited a strong MHC class I antigen presentation and led to a potent cytotoxic T lymphocyte response against ovarian cancer cells [46]. However, in view of the fact that the frequency of HER2 overexpression in ovarian cancer is low [80], the above approach may have limited applicability.

Herpes Simplex Virus (HSV), Measles Virus (MeV), and Vesicular Stomatitis Virus (VSV)

They comprise the main viral systems utilised in oncolytic virotherapy/suicide gene therapy approaches for ovarian cancer gene therapy. More specifically, Hartkopf et al. [45] designed a recombinant strain of MeV that carried a fusion cytosine deaminase (CD) protein, which enhanced the chemosensitivity of ovarian cancer cells in 5-fluoroucacil (5-FU) and used it in vitro and in vivo investigations. The authors demonstrated the effective infection and lysis of both human ovarian cancer cell lines and primary tumours [45]. In another study, Hanauer et al. [39], using a bispecific oncolytic MeV, with both HER2 and EpCAM as target receptors, demonstrated a significantly enhanced lysis of tumour cells in xenograft mice, highlighting the superiority of bispecific against monospecific oncolytic viruses [39]. Regarding HSV viruses, Goshima et al. [41] and Thomas et al. [40], in two independent studies, employed HSV in combination with immunostimulatory cytokines, such as GM-CSF and IL-12, respectively. The intraperitoneal injection (IP) of HSV-HF10 in a murine ovarian cancer murine model, together with the anti-tumour effect of GM-CSF, led to a reduction in tumour size and an increase in anti-tumour immune response [41]. Similarly, the intraperitoneal administration of HSV engineered to express IL-12 led to cytotoxic effects, reduced metastasis, and increased CD8+ T cell response [40]. Lastly, the use of VSV-GP, a potent oncolytic virus pseudotyped with the lymphocytic choriomeningitis virus (LCMV) envelope glycoprotein, together with the Jak1 and Jak2 inhibitor Ruxolitinib, led to transient tumour remission in mice following IP administration [42].

3.1.2. Non-Viral Systems

Strategies of this category utilise DNA/RNA plasmids to alter gene expression via transient transfection or nanoparticles to target a molecule expressed on the surface of cancer cells (Figure 3, Table 1).

Plasmids

The use of these approaches has yielded therapeutic effects through suicide gene therapy, immunopotentiation, and mutation compensation strategies. More specifically, Sher et al. [50] constructed the hEndoyCD fusion protein, composed of the antiangiogenic endostatin and the Escherichia coli cytosine deaminase (CD) domain which converts 5-fluorocytosine (5-FC) to 5-FU, and delivered it to ovarian cancer cells via a plasmid vector (SV-hEndoyCD). The authors reported tumour-specific growth inhibition and increased survival in xenograft mouse models [50]. Regarding immunopotentiation approaches, two reports provide evidence of survival improvement [57,58]. Specifically, Hu et al. employed human umbilical cord CD34+ stem cells transfected with the pIRES2-IL-21-EGFP plasmid, carrying the anti-tumour cytokine IL-21, and demonstrated a therapeutic effect in ovarian cancer xenografts, possibly due to tumour-specific NK cytotoxicity, as a result of elevated levels of IFN-γ and TNF-α. Despite the gradual decrease in IL-21 in mice tumour tissues, overall extended survival was observed in these mice [57]. Similarly, Fewell et al. employed a pmIL-12/PPC vehicle carrying the anti-cancer IL-12 cytokine and demonstrated an efficient treatment for disseminated ovarian cancer as a result of the significant VEGF decrease and IL-12 and IFN-γ increase following IP administration [58].
Regarding mutation compensation, a plethora of studies have demonstrated significant anti-tumour effects following this approach [60,61,62,63,64,65,67,68,69,70]. Specifically, Wu et al., demonstrated a reversal of the malignant phenotype in vitro following the transfection of A2780 epithelial ovarian cancer (EOC) cells with a plasmid carrying the wild-type phosphatase and tensin homolog (PTEN) gene [60]. The above cells showed apoptosis induction, G1 arrest, and a decrease in migration and invasion [60]. In a different study, transfection with pcDNA3.1-p16/pcDNA3.1-vi5-hisA-eEF1A2 plasmids led to eEF1A2 down-regulation through the increase in tumour-suppressor p16 and in turn inhibited cancer cell growth [61]. In a similar approach, Lu et al. [62] demonstrated a significant suppression of ovarian cell proliferation and migration in vitro and in vivo by the up-regulation of p16. The tumour-suppressing effects of the WW domain containing the oxidoreductase (WWOX) gene have been employed in targeted mutation compensation approaches in ovarian cancer, leading to significant results. Yan et al. [63] used a recombinant plasmid to transfect ovarian cancer stem cells and showed decreased proliferation, cell cycle arrest during the G0/G1 phase, and the induction of apoptosis through the up-regulation of caspase-3. A similar result was observed by Xiong et al. [65], employing WWOX delivery in liposomes, as described below. Recently Zhao et al. [64] showed that WWOX activation might inhibit chemoresistance and cell death induction in PTX-treated EOC cells. Specifically, the overexpression of WWOX in PTX-sensitive EOC cells using the pcDNA3.1-WWOX plasmid demonstrated the induction of apoptosis and the inhibition of autophagy and cell proliferation following PTX treatment [64]. The importance of the down-regulation of metastasis-associated in colon cancer 1 (MAAC1), metastasis-associated 1 (MTA1), and cyclooxygenase 2 (COC2) genes in ovarian cancer induction and progression was demonstrated by several studies, employing the down-regulation of their expression by shRNA/siRNA strategies [67,68,69,70]. Specifically, Zhang et al. used an shRNA plasmid to knock-down MACC1 in ovarian carcinoma (OVCAR)-3 cells to demonstrate the inhibition of proliferation, migration, invasion, and induction of apoptosis both in vitro and in vivo in mouse models [67], while the siRNA interference strategy to knock-down MTA1 in ovarian cancer cell line A2780 resulted in reduced migration, invasion, and adhesion and increased apoptosis in vitro [68]. Finally, the knock-down of cyclooxygenase 2 (COX2) by two independent groups [69,70] highlighted the role of the COX2 gene in ovarian cancer cell proliferation, invasion, migration, and growth.

Nanoparticles (NPs)

The use of these systems, primarily in suicide gene therapy and mutation compensation targeted approaches against ovarian cancer, has also yielded significant results. Specifically, Bai et al. transferred gelonin toxin in ovarian cancer cells using cationic heparin PEI (HPEI) nanogels and managed to reduce cancer cell growth and induce apoptosis [51]. Furthermore, Huang et al. [52] delivered diphtheria toxin subunit-A (DT-A) DNA in mice with ovarian tumours by means of cationic polymer administered IP, placing it under the control of a human epididymis protein 4 (HE4) promoter, whose activity is increased in ovarian cancer cells. The latter promoter was also used to drive the TK gene, leading to the inhibition of tumour growth and increased survival in mice upon delivery [52]. In a similar approach, Cocco et al. [53] employed the IP administration of NPs engineered to deliver a plasmid encoding for DT-A and succeeded in inhibiting ovarian tumour growth. Recently, Sun et al. [54] managed to successfully deliver a GCV-converted HSV-tTK in ovarian cancer cells employing Tat/pDNA/C16TAB (T-P-C) NPs, demonstrating targeted delivery in mouse models with human ovarian cancer.
Regarding the mutation compensation approach using nanoparticles, this involves the delivery of siRNA in NPs, targeting EGFR [66], the receptor of Clostridium perfringens enterotoxin claudin-3 (CLDN3) [74,75] and hypoxia-inducible factor 1-alpha (HIF1-a) [76]. To this end, Dickerson et al. knocked down EGFR, an oncogene highly expressed in ovarian cancer cells and implicated in cell proliferation, migration, and differentiation, using siRNA delivered in nanogels and showed increased sensitivity of these cells to docetaxel [66]. Similarly, the knock-down of CLDN3 by siRNA [74] or shRNA [75] led to the inhibition of tumour growth, a reduction in cell proliferation and angiogenesis, and the induction of apoptosis. Lastly, the silencing of HIF1-a, often overexpressed in ovarian cancer by means of siRNA in NPs, led to the effective inhibition of cell proliferation [76]. Regarding lipoplexes, dendrimers, and liposomes, a significant number of studies [55,56,59,71,72,73,77,81] have reported efficient delivery in ovarian cancer cells, highlighting the significant contribution of NPs in targeted gene therapy applications. Concerning the suicide gene therapy approach, He et al. [55] used FRα-targeted folate-modified lipoplex loaded with an hTERT promoter-regulated plasmid encoding VSV matrix protein (MP) (F-LP/pMP) and transfected SKOV3 ovarian cancer cells. The F-LP/pMP treatment resulted in the significant inhibition of tumour growth, as demonstrated by the inhibition of cell proliferation, apoptosis induction, and the suppression of tumour angiogenesis, as well as by the extended survival of mice in a SKOV-3 tumour model [55]. Moreover, Kobayashi et al. [56], employing a PAMAM dendrimer to deliver carbonyl reductase 1 (CRB1), which reduces micro-vessel density and leads to apoptosis, observed increased survival in mice and the decreased proliferation and dissemination of cancer cells.
Furthermore, in a study by Yang et al. [59], the delivery of the human pro-apoptotic gene hPNAS-4 into ovarian cancer cells using cationic liposomes led to the efficient inhibition of tumour growth, the induction of apoptosis, and the inhibition of angiogenesis both in vitro and in vivo. Furthermore, the inhibition of the Wilms tumour 1 (WT1) gene, overexpressed in ovarian carcinoma but not in normal tissue, by an antisense oligodeoxynucleotide (ASODN) delivered in liposomes, led to the inhibition of ovarian cancer cell proliferation, cell cycle arrest, and increased apoptosis in vitro [71]. The delivery of shRNA against STAT3 in liposomes resulted in a reduction in the growth of intraperitoneal ovarian cancer through the inhibition of cell proliferation and apoptosis induction [72], while the delivery of siRNA against NOTCH1 in cationic cholesterol derivative-based liposomes led to the inhibition of growth and apoptosis induction in vitro [73]. As mentioned above, Xiong et al. [65] delivered WWOX in ovarian cancer cells by means of liposomes and documented decreased cancer cell growth and apoptosis induction. Lastly, the utilisation of the CRISPR/Cas9 system in a folate receptor-targeted cationic liposome (F-LP) to target the DNA methyltransferase 1 (gDMNMT1) gene, whose overexpression results in inactivation or tumour suppressor genes and increased ovarian cancer cell resistance, resulted in the inhibition of ovarian cancer tumours both in vitro and in vivo [77].

miRNAs

Large-scale microarray analysis has highlighted the role of many microRNAs [miR(s)] in different types of cancer, including ovarian and cervical cancer [82]. miRNAs in ovarian cancer have shown to have either a tumour-promoting or tumour-suppressing role, depending on whether their expression is up-regulated or down-regulated, respectively, and hence can be employed both as therapeutic targets, as well as biomarkers for diagnosis and/or prognosis [83]. Regarding ovarian cancer, Iorio et al. [84] performed an initial and comprehensive miRNAs comparison between normal and ovarian cancer tissues and showed that miR-14, mir-199a, miR-200a, miR-200b, and miR-200c were up-regulated in ovarian cancer and thus acted as oncogenic miRs (oncomiRs), while miR-15, miR-16, mir-140, mir-145, mir-199a, and miR-125b1 were down-regulated, pointing toward a tumour-suppressing role (tumour suppressor miRs). Further studies highlighted novel players, such as miR-214, miR-150, miR-140-5P, miR-21, miR-29a, and let-7a and provided more insights into the role of different miRNAs in ovarian cancer.
More specifically, Yang et al. [85] showed that miR-214 and miR-150 are highly up-regulated in ovarian cancer and act as oncomiRs, inhibiting the expression of PTEN tumour suppressor gene, thus initiating the protein kinase B (ACT) pathway. On the other hand, the prevention of miR-PTEN interaction can inhibit ovarian cancer, primarily through the inhibition of cancer cell proliferation and chemotherapeutic drug resistance. Therefore, the down-regulation of both miR-214 and miR-150 expression could potentially have a therapeutic effect. Similarly, experiments by Lan et al. [86] showed that miR-140-5p expression is significantly decreased in ovarian cancer patients, while when up-regulated, it leads to the induction of ovarian cancer cell proliferation and migration. The authors also observed that miR-140-5p down-regulation could reduce sensitivity to cisplatin, and therefore, the up-regulation of the specific miRNA could lead to both the inhibition of proliferation and migration and an enhancement of cisplatin sensitivity. Regarding miR-21, Echevarria-Vargas et al. [87] showed that miR-21 is also associated with cisplatin resistance among ovarian cancer cells. Its expression was found to be up-regulated in cisplatin-resistant ovarian cancer cells, and therefore targeting miR-21 can lead to reduced tumour characteristics in the specific cells. The down-regulation of miR-29a in ovarian cancer tissues is associated with apoptosis [88] while restoring let-7A-3/let-b expression leads to a reduction in tumour growth both in vitro and in vivo and the inhibition of invasion and metastasis [89]. Furthermore, increased let-7a is associated with increased survival rates in patients receiving platinum-based chemotherapy [90]. Similarly, the up-regulation of miR-200 family members is associated with decreased metastasis, primarily through reduced epithelial–mesenchymal transition (EMT) and tumour angiogenesis. Pecot et al. [91] reported that the miR-200 family inhibits blood vessel formation by targeting IL-8 and CXCL-1; therefore, the expression of the miR-200 family to the tumour epithelium results in decreased tumour cell metastasis and angiogenesis. Moreover, the observation that miR-183 and miR-22 expression was elevated in the low, compared to the high form of metastatic cancer, coupled with the fact that increasing the expression of miR-183 and miR-22 resulted in EZRIN reduction, led to the conclusion that EZRIN inactivation through miR-183 and miR-22 can inhibit ovarian cancer metastasis [92]. Lastly, Aqeilan et al. [93] demonstrated that miR-15 and miR-16 are involved in the multi-drug resistance of ovarian cancer cells by acting on Bcl-2, while Luo et al. [94] reported the tumour suppressing role of miR-126 in cancer SKOV3 cells, by demonstrating a miR-126-mediated P21 (RAC1) activated kinase 4 (PAK4) down-regulation.

3.2. Cervical Cancer

Cervical cancer remains the fourth most common and most lethal cancer type in women, despite regular screening and prevention strategies [95]. Targeted gene therapy presents a promising approach for the treatment of the specific malignancy and focuses primarily on mutation compensation strategies, suicide gene therapy, oncolytic virotherapy, antiangiogenic strategies, immunopotentiation, and drug resistance therapies [3], employing both viral and non-viral systems.

3.2.1. Viral Vectors

The main viral vectors used for the gene therapy of cervical cancer are LVs, Ads, and AAVs (Figure 4, Table 2).

Lentiviruses

Cervical cancer gene therapy approaches employ LVs primarily for mutation compensation, antiangiogenic, and suicide gene therapy strategies. The restoration of important tumour suppressor genes’ expression, such as tyrosine phosphatase receptor J (PTPRJ), asparaginase and isoaspartyl peptidase 1 (ASRGL1), and homeobox-containing 1 (HMBOX1), has been successfully achieved with LVs. Specifically, Yan et al. [96] used pSicoR-PTPRJ LV to overexpress PTPRJ, a tumour suppressor gene whose expression is down-regulated in human cervical cancer tissues and demonstrated significant suppression of cell viability, migration, and growth in HPV-negative C33A cells. On the contrary, the knock-down of PTPRJ expression in the above cervical cancer cell line led to increased resistance to 5-FU-mediated apoptosis, verifying the importance of elevated PTPRJ expression for cervical cancer prevention. Moreover, Zhou et al. used a lentiviral shRNA to knock-down HMBOX1 expression in HeLa and C33A cancer cells and demonstrated increased radiosensitivity as a result of telomere shortening [97]. Lastly, the knock-down of ASRGL1 expression in SiHa cells by means of shRNA led to decreased proliferation, possibly through the reduced expression of CDK2 and cyclin A2 and the induction of apoptosis [98], which was characterised by the increased expression of the pro-apoptotic Bax and the decreased expression of anti-apoptotic Bcl-2. Regarding the antiangiogenic approach, Qi et al. used a lentiviral shRNA-VEGF construct to knock down VEGF expression in vitro and in vivo in nude mice and was able to inhibit tumour growth and tumour radiosensitivity [99]. One of the most promising approaches for cervical cancer gene therapy using lentiviruses is the genetic modification of T-cell receptors (TCRs) to target tumour-specific antigens. Based on the above, Jin et al. developed E7-specific T cells and achieved regression of HPV-positive mouse tumours [100]. The specific approach is currently under clinical trial NCT02379520 and is being used on patients with metastatic cervical cancer [101]. Regarding suicide gene therapy, the LV-mediated targeting of IAA and CD5 has been employed, while the GlNaTK retroviral vector was used to transfer the suicide TK gene into tumour cells. More specifically, Zhang et al. combined SEC3-ES LV targeting IAA and cisplatin and showed cell cycle arrest at the S phase, as well as the induction of apoptosis [102]. Similarly, Hao et al. studied the synergistic effect of two suicide gene systems, such as CD/5-FC and HSV-TK/ganciclovir (GCV), and demonstrated the inhibition of proliferation and enhanced apoptosis in vitro following lentiviral delivery combined with ultrasonic radiation [103]. Lastly, Chen et al. employed the GlNaTK retroviral vector to mediate TK transfer, which led to the inhibition of cell growth, increased apoptosis, and tumour radiosensitisation to cobalt-60 following GCV administration [104].
Table 2. Targeted gene therapy approaches for cervical cancer.
Table 2. Targeted gene therapy approaches for cervical cancer.
Gene Therapy StrategyGeneDelivery SystemOutcomeModel/SpeciesReferences
Viral vectors
Mutation compensation
p53Adenoviral vector
rAd-p53
Enhanced growth inhibition and
apoptosis; increased 5-year
overall survival rate
in vitro (human)
in vivo (human)
[38,105]
Mutation compensationE6Adenovirus +siRNASuccessful p53 transduction
following adenovirus treatment
in vitro (human)[106]
Mutation compensationE6/E7Adenoviral vector
Ad-ER-DN
Block of cell proliferation and
induction of apoptosis
in vitro (human)[107,108]
Suicide gene therapyhLF
(human
latoferin)
Adenoviral vector
rAd-hLF
Inhibition of tumour growth due to cell cycle arrest in G2/M phase and prolonged survival in xenograft modelsin vitro (human)
in vivo (mouse)
[109]
Oncolytic virotherapyE2Adenovirus
rAd-M5
Growth suspension and
increased apoptosis
in vitro (human)
in vivo (mouse)
[110]
Oncolytic virotherapyE6/E7Adenovirus
rAd-M6
Anti-tumour effectin vitro (human)
in vivo (mouse)
[111]
Oncolytic virotherapyEgr-1
TRAIL
Adenovirus
rAd-pE3-Erg/TRAIL
Increased apoptosis and
reduction in tumour size
in vitro (human)
in vivo (mouse)
[112]
Oncolytic virotherapyE6Adenovirus
rAd-H101
Down-regulation of HPV 16 E6
expression and increased
p53 expression
Enhancement of radiation
anti-tumour efficiency
in vitro (human)[113]
Oncolytic virotherapy/
Antiangiogenic strategy
VEGF
inhibitor
(VEGI- 251)
Adenoviral vector
VEGI-armed
Inhibition of tumour
neovascularization and
induction of apoptosis
in vitro (human
in vivo (mouse)
[114]
ImmunopotentiationIL-12Adenoviral vector
rAd-hIL12
Anti-tumour effect, inhibition of tumour growth and increase in survival ratesin vitro (mouse)
in vivo (mouse)
[115]
ImmunopotentiationIL-15Adeno-associated vector
rAAV2-hIL15
Inhibition of cell growthin vitro (human)
in vivo (mouse)
[116]
ImmunopotentiationTNF-14
(light)
Adeno-associated vector
rAAV2-LIGHT
Suppression of tumorigenesis in
a murine cervical cancer model
in vitro
(mice/hamsters)
in vivo (mouse)
[117]
Mutation compensationHMBOX1shRNA lentiviral
vector
Increased apoptosis rate,
increased radiosensitivity
in vitro (human)[97]
Mutation compensationASRGL1shRNA lentiviral vectorDecreased proliferation and apoptosis inductionin vitro (human)[98]
Mutation compensationPTPRJLentiviral vector
pSicoR-PTPRJ
Overexpression of PTPRJ led to significant suppression of
viability, migration, and growth of cancer cells; Knock-down of PTPRJ increased C33A resistance to 5-FU-induced apoptosis
in vitro (human)[96]
Antiangiogenic strategyVEGFLentiviral vector
shRNA-VEGF
Inhibition of cancer cell growthin vitro (human)
in vivo (mouse)
[99]
ImmunopotentiationE7-specific TCRLentiviral vectorRegression of HPV-positive mouse tumoursin vitro (human
in vivo
(human/mouse)
[100]
[101]
Suicide gene therapyCD5/TKLentiviral vectorInhibition of proliferation and
enhanced apoptosis
in vitro (human)[103]
Suicide gene therapyIAALentiviral vector
SEC3-ES
Cell cycle arrest at S phase
and apoptosis
in vitro (human)[102]
Suicide gene therapyHSV-TKGINaTK retroviral vectorCell growth inhibition, increased apoptosis, and tumour
radiosensitisation to cobalt-60
in vitro (human)
in vivo (mouse)
[104]
Mutation compensationc-MYCSendai virus (SeV)Decrease in c-Myc and significant anti-tumour effects and apoptosis inductionin vitro (human)
in vivo (mouse)
[118]
Non-viral systems
Mutation compensationHPV18 E6/E7CRISPR/Cas9—E6/E7-KORobust knockout of E6 and E7 in cervical cancer cells, increased apoptosis and tumour size
reduction
in vitro (human)
in vivo (mice)
[119]
Mutation compensationHPV16 E6/E7CRISPR/Cas9 -nanoliposomesSignificant reduction
in tumour size
in vitro (human)
in vivo (mouse)
[120]
Mutation compensationE6/E7CRISPR/Cas9, TALENGrowth inhibition, decreased
proliferation, and increased
apoptosis of HPV-positive
cervical cancer cells
in vitro (human)
in vivo (human)
[121,122]
[101]
Mutation compensationHPV16 E7Nanoparticles
composed of non-
viral PBAE546 and
CRISPR/Cas9 recombinant plasmid
Inhibition of growth of xenograft tumours and reversal of the
cervical epithelial malignant
phenotype of HPV16
transgenic mice
in vivo (mouse)[123]
Mutation compensationHPV16 E7CRISPR/Cas9Therapeutic effect
in K14-HPV16 mice
in vitro (human)
in vivo (mouse)
[124]
Mutation compensationE6/E7 and MCL1siRNA delivery by
PEG-lipoplexes
Successful and prolonged
delivery of
active siRNAs by lipoplexes
for HPV-induced lesions
in vitro (human)[125]
Mutation compensationp53Polyamidoamine derivative
AP-PAMAM
Anti-proliferative effect,
induction of cell-cycle arrest at
S phase, suppression of cancer cell migration and invasion
in vitro (human)[126]
Mutation compensationXIAPsiRNACleaved caspase-3 activation and apoptosis in tumour tissuein vivo (mouse)[127]
Mutation compensationhTERTsiRNATargeting hTERT induces growth inhibition and radiosensitivity;
Knock-down of hTERT inhibits
cervical cancer growth;
Knock-down of HMB0X1 increases the radiosensitivity of cervical
cancer cells
in vitro (human)
in vivo (mouse)
[97,128,129]
Mutation compensationPEDFFRα-targeted
liposome (FLP)
Significant anti-tumour activity, as demonstrated by significant growth inhibition and
suppression of adhesion,
invasion, and cancer cell migration
in vitro (human)
in vivo (mouse)
[130]
Mutation compensationRIZ1pcDna3.1(+)-RIZ1 plasmidUp-regulation of RIZ1 leads to reduction in cell proliferation and increased apoptosis;
in combination with
radiotherapy, it leads to increased apoptosis and DNA damage
in vitro (human)
[131]
Mutation compensationMMP9
PTX3
shRNAKnock-down of PTX3 inhibits cell migration and invasionin vitro (human)
in vivo (mice)
[132,133]
Antiangiogenic strategyNet1siRNAAngiogenesis and tumour growth reduction
through VEGF
down-regulation
in vitro (human)
in vivo (mouse)
[134]
ImmunopotentiationCXCL10Liposome-encapsulated plasmidGrowth and angiogenesis
inhibition, apoptosis induction,
arrest at G1 phase
in vitro (human)
in vivo (mouse)
[135,136]
ImmunopotentiationE6/E7pcDNA-3CRT/E7Potent CD8 T-cell response and
enhanced anti-tumour effects and survival
in vivo
(mouse/human)
[137]
[101]
ImmunopotentiationE6/E7pcDNA-3CRT/E7 + IL2Potent anti-tumour CD8 T-cellsin vivo (mouse)[138]
ChemoresistancehTERThTERT27Inhibition of cell proliferation and induction of apoptosisin vitro (human)[139]
ChemoresistanceBR1P1BRIP1 recombinant plasmidEnhanced anti-tumour activity of cisplatin, increased apoptosis
and angiogenesis suppression
in vitro (human)[140]
Figure 4. Gene therapy strategies for cervical cancer. They employ viral vectors, primarily adenoviral and adeno-associated viral vectors and lentiviral vectors, and non-viral systems, such as DNA plasmids, nanoparticles, CRISPR/CaS9, and shRNA/siRNA/miRNA approaches.
Figure 4. Gene therapy strategies for cervical cancer. They employ viral vectors, primarily adenoviral and adeno-associated viral vectors and lentiviral vectors, and non-viral systems, such as DNA plasmids, nanoparticles, CRISPR/CaS9, and shRNA/siRNA/miRNA approaches.
Cancers 14 03238 g004

Adenoviruses (Ad) and Adeno-Associated Viruses (AAV)

They comprise the majority of viral vectors used for cervical cancer gene therapy, often utilised for tumour suppressor gene restoration or blocking of oncogenic expression. The p53 gene, a key regulator of cell proliferation, apoptosis, and genetic stability [141], plays a fundamental role in most gynaecological cancers and has therefore been a major candidate for most targeted gene therapy approaches [78]. More specifically, since the p53 gene is often inactivated by HPV E6 protein in most cervical cancers [142]; either its delivery or inhibition of the E6 protein could result in significant antitumour effects. Indeed, Su et al. employing Genidicine®, a gene therapy product approved in 2003 by the China Food and Drug Administration (CFDA) for head and neck cancer gene therapy [143], showed that the injection of a recombinant human adenovirus engineered to express wild-type p53 gene (rArd-p53) in cervical cancer patients, may lead to an increased 5-year overall survival rate [105]. Moreover, Kajitani et al., using siRNA for E6 protein in HeLa cells, demonstrated successful p53 transduction following adenovirus treatment [106]. When the above transduction in HeLa cells was combined with PTX, it led to enhanced growth inhibition and apoptosis, as demonstrated by Liu et al. [38]. A further approach in tumour repressor gene restoration strategy is the use of rAd-ER-DN, an adenovirus expressing a dominant-negative estrogen receptor gene (DNER), to regulate estrogen imbalance, often associated with cervical cancer development. The use of rAd-ER-DN alone [107] or in combination with cisplatin and PTX [108] led to the blocking of cell proliferation and the induction of apoptosis in Ca Ski cervical cancer cells, primarily through the decrease in E6 and E7 oncoprotein expression. Regarding the oncolytic virotherapy approach, the use of five different Ads, i.e., rAd-M5, rAd-M6, rAd-53 [112], rAd-VEGFI-251, and rAd-H101 [113], demonstrated significant anti-tumour effects both in vitro and in vivo and led to increased apoptosis of cancer cells. rAd-M5 expressed the HPV E2 gene, which negatively regulated E6 and E7 expression and led to growth suspension and increased apoptosis [110], while rAd-M6 employing antisense HPV16 E6/E7 DNA inhibited the expression of E6 and E7 and induced apoptosis in vitro and in vivo [111]. In another study, Xiao et al. used an oncolytic adenovirus expressing the VEGF inhibitor VEGI-251 and demonstrated the induction of apoptosis and the inhibition of neovascularisation [114], while Wang et al., employing rAd-p53 carrying the promoter of early growth response (Egr-1), and the anti-apoptotic TNF-related apoptosis-inducing ligand (TRAIL) gene showed increased radiotherapy-induced apoptosis in cell lines and tumour size reduction in xenograft mice [112]. Recently, Duan et al., using rAd-H101 oncolytic adenovirus targeting the E6 gene demonstrated enhanced radiation anti-tumour activity, primarily in C33A cells [113]. As far as the suicide gene therapy approach is concerned, the use of an Ad carrying the human lactoferrin (hLf), a glycoprotein involved in solid tumour growth reduction, demonstrated the inhibition of tumour growth, due to cell cycle arrest in the G2/M phase in HeLa cells and a prolonged survival in mice with cervical cancer [109]. Lastly, immunopotentiation strategies in cervical cancer gene therapy show promising results and involve both adenoviruses and adeno-associated viruses. In particular, the delivery of hIL-12, a cytokine with potent anti-cancer activity via the adenoviral vector (rAd-hIL12), led to the inhibition of tumour growth and an increase in survival rates in murine models [115]. Similarly, the delivery of the anti-tumour hIL-15 cytokine using the rAAV2-hIL15 led to the inhibition of growth in HeLa cells in vitro and in vivo in murine models [116], while rAAV2-LIGHT, an AAV vector carrying LIGHT, a TNF ligand, succeeded in suppressing tumorigenesis in a murine cervical cancer model [117].

3.2.2. Non-Viral Systems

Strategies of this category utilise primarily NPs to target a molecule expressed on the surface of cancer cells, as well as DNA and RNA plasmids or miRNAs to alter gene expression via transient transfection (Figure 4, Table 2).

Nanoparticles (NPs)

Gene therapy strategies by means of nanoparticles focus primarily on mutation compensation and immunopotentiation strategies. Regarding the former approach, Liu et al. [144] developed polyethylene glycol-polylactic acid (PEG-PLA) NPs linked to folate and targeted cancer cells through the folate receptor α (FRα), a membrane-bound protein mediating folate uptake. Although the authors observed enhanced gene transfection efficiency, higher compared to naked DNA, and reduced cytotoxicity, the strategies targeting FRα receptors are hampered by its heterogeneous expression among cervical cancer patients [145]. A few years later, Yang et al. overexpressed FRα, previously shown to be highly expressed in cervical cancer, using an FRα-targeted liposome (FLP) to deliver a pigment epithelium-derived factor (PEDF) gene into HeLa cells and observed significant anti-tumour activity, as demonstrated by significant growth inhibition, the suppression of adhesion and invasion, and cancer cell migration in vitro [130]. Lechanteur et al. used PEG-lipoplexes to mediate siRNA delivery against E6 and E7 oncoproteins and the anti-apoptotic myeloid cell leukaemia 1 (MCL1) gene; this was followed by the successful and prolonged delivery of active siRNAs by PEGylated lipoplexes, suggesting that this approach can treat HPV-induced lesions in vitro and in vivo in murine models [125]. Moreover, Han et al. using an AP-PAMAM derivative, induced an anti-proliferative effect in vitro by suppressing cancer cell migration and invasion, as well as the induction of cell-cycle arrest at the S phase [126]. Recently, the use of CRISPR/Cas9 against the HPV16 E6 and E7 encoding genes delivered in nanoliposomes led to a significant reduction in tumour size in murine mouse models [120]. Nanoparticles, composed of non-viral vector PBAE546 and a CRISPR/Cas9 recombinant plasmid against HPV16 E7 protein, led to the inhibition of growth in xenograft tumours and the reversal of the cervical epithelial malignant phenotype of the HPV16 transgenic mice [123]. Finally, regarding the immunopotentiation approach, Wang et al. [135] and Yang et al. [136] employed a liposome-encapsulated plasmid encoding CXCL10, an important cytokine with a potential therapeutic role in cervical cancer; they demonstrated an inhibition of growth and angiogenesis, which was associated with induction of apoptosis, mediated by the modulation of E6 and E7 oncoproteins expression both in vitro and in vivo.

Plasmids

The use of different plasmids in cervical cancer gene therapy approaches focuses on mutational compensation, antiangiogenesis, immunopotentiation and chemoresistance. With regards to blocking oncogenic expression, Hu et al. employed the genome editing approach using both the CRISPR/Cas9 and transcription-activator-like nucleases (TALEN) systems to disrupt the HPV16 E7 oncoprotein coding gene [122]. The data documented induction of apoptosis and growth inhibition in HPV16-positive human cancer cells. The latter approach is being studied in Phase I clinical trials for the treatment of CIN-1 patients with HPV16 and HPV18 infection [101]. Recently, two groups demonstrated the therapeutic effect of targeting HPV E6 and E7 genes. Ling et al., using the CRISPR/Cas9 approach to delete the HPV18 E6 and E7 genes, achieved robust knock-out of these proteins and increased apoptosis and tumour size reduction [119]. In an attempt to compare the efficiency of CRISPR/Cas9 with the established TALEN approach, Gao et al. employed the former system against the HPV16 E7 gene and succeeded in reverting cervical carcinogenesis, both in vitro and in vivo [124]. Another tumour suppressor gene with clinical importance in cervical cancer gene therapy is the retinoblastoma protein zinc finger gene 1 (RIZ1) since it can induce apoptosis and cell cycle arrest. Cheng et al. demonstrated that overexpression of RIZ1 expression in HPV16-positive cervical cancer cells could lead to impaired cell proliferation and increased apoptosis [131]. When this overexpression is combined with radiotherapy, it leads to increased apoptosis and DNA damage in HeLa and SiHa cells [146]. Blocking oncogenic expression can also be mediated by plasmids, primarily via the siRNA and shRNA approaches. Important genes such as X-linked inhibitor of apoptosis (XIAP), matrix metalloproteinase (MMP), and telomerase reverse transcriptase (hTERT) were targeted using the above approaches with promising results. Specifically, Wang et al. employed an RNA interference approach to silence the anti-apoptotic XIAP and demonstrated cleaved caspase-3 activation and apoptosis in tumour tissues [127]. Similarly, targeting of hTERT, which is often upregulated in cervical cancer and implicated in tumour cell invasion and proliferation, resulted in induction of growth inhibition, along with enhanced radiosensitivity in vitro [128], reduced cell proliferation, migration and invasion of cancer cells in vitro, as well as in inhibition of cell growth and tumorigenesis in nude mice [129]. The direct knock-down of the MMP9 protein involved in metastasis [132] or the indirect targeting of PTX3 [133], a molecule necessary for MMP2 and MMP-9 expression using shRNA plasmids, led to a reduction in cell migration and invasion, both in vitro and in vivo. In addition to the use of siRNAs for mutational compensation, their use in antiangiogenic strategies has also been demonstrated. Specifically, Zhang et al. used RNA interference to knock-down Net1, shown to be overexpressed in cervical cancer tissues, and demonstrated decreased proliferation, migration, and angiogenesis in SiHa cells, as well as a reduction in angiogenesis and tumour growth in vivo, via VEGF down-regulation [134]. Regarding immunopotentiation, E6 and E7 oncoproteins have been the optimal targets for immunotherapy and DNA vaccine production. Sun et al. developed a DNA vaccine using a plasmid encoding calreticulin (CRT) and E7 protein and managed to evoke a potent CD8 T-cell response, which could ultimately lead to anti-tumour effects and survival [137]. The co-administration of DNA encoding IL-2 in tumour-bearing mice elicited a stronger CTL response and enhanced anti-tumour effects [138]. The above approach is under clinical trial NCT00988559 [101]. Lastly, the issue of chemoresistance is also addressed by gene therapy to induce sensitivity in cervical cancer cells to chemotherapy. Examples of such approaches with promising results include the up-regulation of hTERTC27 and BRACA1-interacting protein (BRIP1) expression. More specifically, Lin et al. demonstrated that the overexpression of hTERTC27 expression led to increased sensitivity in HeLa cells to 5-FU, followed by inhibition of cell proliferation and increased apoptosis, primarily through the down-regulation of anti-apoptotic Bcl-2 expression and the up-regulation of pro-apoptotic caspases 3 and 9 [69]. Similarly, Liu et al. found that the overexpression of BRP1 leads to enhanced cisplatin-mediated anti-tumour activity, increased apoptosis, and suppression of angiogenesis in vitro [140].

miRNAs

miRNAs involved in cervical cancer induction and development are classified into oncogenic miRNAs (oncomiRs) and tumour suppressor miRNAs (tumour suppressor miRs) [147,148] (Figure 4), and therefore, their down-regulation or overexpression, respectively, can be curative, while their presence in patients’ serum is considered as an important tool for cancer diagnosis and prognosis. More specifically and regarding oncomiRs, the most important ones include miR-10a, miR-19, miR-20, miR-21, miR-133b, and miR-886-5p [147]. Long et al. showed that miR-10a suppresses cell adhesion molecule L1, such as (CHL1), and thus leads to enhancement of tumour growth, metastasis, and invasion [149], while miR-19 is overexpressed in cervical cancer, and its silencing in SiHa cells led to a reduction in the proliferation and induction of apoptosis, through the up-regulation of Bax and down-regulation of Bcl-2 expression [150]. MiR-20 is a positive regulator of tyrosine kinase non-receptor 2 (TNKS2), an oncogene involved in metastasis and invasion [151]. In contrast, miR-21, which is the most well-known oncogenic miRNA, acts as a negative regulator of the tumour suppression gene programmed cell death 4 (PDCD4), which normally inhibits cell proliferation and induces apoptosis [152], thus promoting tumour growth. Silencing by means of siRNA in cervical cancer cell lines led to inhibition of cell proliferation and induction of cell death by autophagy and caspase 3/7-mediated apoptosis [153]. Moreover, through targeting and regulating CCL20, a gene involved in tumour differentiation and metastasis, miR-20 was implicated in cervical squamous carcinogenesis [152]. Lastly, miR-133b is implicated in malignant transformation, originating from ERK and AKT1 signalling pathways [154], while miR-886-5p is negatively regulating the expression of the apoptotic protein Bax, as its silencing in SiHa cells led to induction of Bax expression and sequentially to apoptosis [155]. Protective miRNAs with important tumour suppressor activity primarily include miR-143, miR-148b, miR-211, miR-187, miR-641, miR-664, miR-let-7a, and miR-129-5p. More specifically, Liu et al. showed that the overexpression of miR-143 in HeLa cells led to suppression of Bcl-2 expression and thus the inhibition of cell proliferation and the induction of apoptosis [156]. Similarly, the transfection of miR-187 in SiHa cells also led to the downregulation of Bcl-2 and, in turn, to a reduction in cell growth and increased apoptosis [157]. Mou et al. employing miR-148b overexpression in HeLa cells demonstrated induction of G1/S cell-cycle arrest and apoptosis, mediated by caspase-3 [158], while Liu et al. showed that the down-regulation of IAP expression in SiHa cells following miR-211 overexpression, led to the inhibition of cancer cell growth and apoptosis enhancement [159]. Furthermore, the overexpression of miR-641 [160] and mir-644 [161] led to the inhibition of cervical cancer cell proliferation, migration, and invasion, with a concomitant increase in apoptosis. Ultimately, the ectopic expression of miR-let-7a in SiHa and HeLa cells led to the inhibition of cancer cell proliferation, migration, invasion, and apoptosis induction [162], while miR-129-5p overexpression in HeLa cells resulted in the inhibition of cell proliferation, increased cell-cycle arrest at the G0/G1 phase, and promoted apoptosis [163].

3.3. Endometrial Cancer

Despite being usually curable following surgery, endometrial cancer still presents as one of the most common female reproductive tract cancer types [4]. Occasionally aggressive tumours, such as uterine papillary serous carcinomas (UPSC), are observed, with most of them demonstrating aberrant expression of p53 [164]. Gene therapy strategies toward endometrial cancer involve both viral and non-viral systems, with the former employing primarily adenoviral and retroviral vectors and the latter plasmid DNA/RNA.

3.3.1. Viral Vectors

Ramondetta et al. performed an adenovirus-mediated expression of p53 or p21 in a papillary serous endometrial carcinoma cell line and demonstrated growth inhibition and apoptotic cell death [165]. Similarly, Ural et al. performed an in vitro suicide gene therapy approach using HSV-TK and documented inhibition of endometrial cancer cell growth [166]. A similar approach, but with the use of the pNF-κB plasmid, along with the gonadotropin-releasing hormone receptor (GnRH-R) agonist triptorelin and the prodrug GCV, resulted in reduced cancer cell growth, both in vitro and in vivo in mice [167]. Recently, Xia et al. [168] employed next-generation sequencing (NGS) in four out of the twelve patients enrolled in the clinical trial using the Ad-p53 vector, Genidicine® [105] and observed reduced p53 expression in the tumours of three patients, all carrying mutations in tumour protein p53 CREB binding protein (CREBBP), cyclin-dependent kinase inhibitor 2A (CDKN2A), LYN proto-oncogene (LYN) and Janus kinase 2 (JAK2) genes. These data provide strong evidence that NGS can aid in the recruitment of suitable patients for Ad-53 uterine gene therapy.

3.3.2. Non-Viral Systems

Effective gene transfer with a significant inhibition of cancer cells in vitro was also achieved with non-viral approaches. Specifically, Maurice-Duelli et al. demonstrated that the transfer of PTEN, a frequently mutated gene in endometrial cancer [169], using PEI-photochemical irradiation could lead to a 44% inhibition in cancer cell growth [170]. Moreover, the role of certain miRNAs in endometrial cancer has gained a lot of ground, highlighting their potential diagnostic and therapeutic value. In a detailed review, Banno et al. [171] highlighted the role of several miRNAs differentially expressed in endometrial cancer. Specifically, miR-185, miR-106a, miR-181a, miR-210, miR-423, miR-107, miR-let7c, miR-205, miR-449, and miR-429 were found up-regulated, while miR-let7e, miR-221, miR-30c, miR-152, miR-193, miR-204, miR-99b and miR-193b were significantly down-regulated, suggesting a tumorigenic or tumour-suppressing activity, respectively [171]. miR-129-2 and miR-152 are involved in the development of endometrial cancer via DNA methylation; miR-125b, mir-30c, miR-200b/c, and miR-429 are related to cisplatin resistance, while miR-125b, miR-30c, miR-194 and miR-34b regulate proliferation, metastasis and invasion of endometrial cancer cells [171]. Similarly, in a recent systematic review, Donkers et al. [172] highlighted the importance of miR-205, miR-200 family (miR-200a, miR-200b, and miR-200c), miR-135b, miR-182, miR-183, and miR-223 in endometrial cancer prognosis [172]. As the above miRNAs are found to be up-regulated in most endometrial cancers, the down-regulation of their expression may have therapeutic outcomes. Regarding miRNAs, such as miR-137, miR-129-3p, and miR-410, whose expression is down-regulated in endometrial cancer, the authors observed little to no consensus [172], and therefore, their role in both predicting and treating endometrial cancer remains vague.

4. Gene Therapy for Benign Gynaecological Disorders

4.1. Uterine Leiomyomas or Fibroids

They are the most common benign tumours in reproductive-age women [173] and represent an attractive target for gene therapy, primarily due to their localised nature and slow growth rate [5,174]. The most common gene therapy approaches for fibroid treatment involve suicide gene therapy, antiangiogenic strategy, and mutation compensation and are summarised in Table 3 and Figure 5. The first attempt to employ gene therapy for uterine leiomyoma treatment came from Niu et al. 1998, who used a non-viral approach to deliver the suicide TK gene into leiomyoma cells [175]. Although the authors demonstrated significant cell death despite the low percentage of transfected leiomyoma cells, the above approach could only be applied in vitro. On the contrary, the use of improved Ads [174] with a leiomyoma-specific expression of critical therapeutic genes, such as DNER [176] and HSV1-TK [5], appear quite promising and present good candidates for human clinical trials. Furthermore, targeted and transduction-efficient Ads, such as Adenovirus-human somatostatin receptor subtype 2-arginine, glycine, and aspartate-thymidine kinase (Ad-SSTR-RGD-TK), given in combination with GCV, show promising results, both in vitro [177] and in vivo [178], as they lead to a significant reduction in proliferation and leiomyomas size, the induction of apoptosis, and the inhibition of angiogenic- and extracellular matrix-related genes. Recently, C-terminal Src kinase (CSK2) [179] and high mobility group AT-hook 2 (HMGA2) [180] were highlighted as key players in leiomyomas tumorigenesis. Specifically, CSK2 was shown to be highly expressed in uterine leiomyosarcomas compared to leiomyomas, and its overexpression was associated with tumour progression and poor prognosis, while CSK2 silencing using siRNA led to the inhibition of proliferation and cell cycle progression, decreased migration, invasion, and colony formation [179]. Additionally, the overexpression of HMGA2 in leiomyomas was shown to be associated with increased vasculature density, demonstrated by the increased expression of angiogenic factors and receptors, such as VEGFA, EGF, bFGF, TGFα, VEGFR1, and VEGFR2, which likely contribute to tumour growth [180]. Its important role in angiogenesis appears to be through IGF2BP2-mediated pAKT activity [180].

4.2. Endometriosis

It is a chronic, estrogen-dependent disease characterised by the presence of endometrial tissue outside the uterine cavity, often associated with subfertility [5,198]. In the early 2000s, the above benign gynaecological abnormality made its way to gene therapy applications (Table 3 and Figure 5). Specifically, Dabrosin et al. using a murine model of endometriosis and employing an adenovirus expressing the murine angiostatin gene managed to completely eradicate endometriotic lesions within two weeks [181]. A similar antiangiogenic strategy was followed by Sun et al., who used an AAV carrying endostatin (rAAv-endostatin-EGFP) and demonstrated the inhibition of angiogenesis in endometriotic lesions both in vitro and in vivo [182]. Similarly, but using an endostatin plasmid/PAMAM dendrimer instead, Wang et al. also demonstrated inhibition of endometriosis development following transfection [185]. Following the above, Othman and co-workers studied the effects of DNER gene transfer into endometriosis cells using an Ad-DNER vector and showed that it could induce significant cell death and reduce pro-inflammatory and angiogenic cytokine production by the specific cells [183]. Both of the above studies used an Ad5 viral vector; however, CRADs, such as Ad-SLP1-luc, Ad-hepanarase-luc [184], and Ad-sft-1 [186], achieved higher reporter gene expression and, therefore, may constitute better candidates for the gene therapy of endometriosis. In addition to antiangiogenic strategies, suicide gene therapy and mutation compensation have also been very efficient for endometriosis treatment. More specifically, the use of adenoviral vectors carrying the TK gene (Ad-TK) led to significant cell death induction and reduction in endometrial lesions in vivo [199]. Furthermore, regarding mutation compensation, Goncalves et al. employed Ad-p27, an adenovirus carrying p27kip1 coding gene, shown to be down-regulated in women with endometriosis [200] and demonstrated cell cycle arrest and reduced proliferation [188,189]. Lastly, Paupoo et al., employing CRAd-S-pK7, an adenoviral vector with dual advantages of infection enhancement and promoter specificity, demonstrated high replication rates and cell-killing efficiencies in vitro [190].

4.3. Placental Disorders

The potential contribution of gene therapy for placental abnormalities and functional restoration deficiencies was first introduced by Senut et al. [201] by injecting genetically modified cells into rat placenta, which demonstrated the increased secretion of gene products in fetal circulation. Furthermore, Xing et al. managed to successfully transfect rat placenta by either systemic administration or intra-placental needle injection of adenoviruses without altering the fetal genome [191]. However, as intra-placental needle injection may result in fetal-maternal barrier breakdown, Heikilla and co-workers performed a catheter-mediated intravascular gene transfer with adenoviruses, plasmid/liposomes and plasmid/PEI complexes and demonstrated that placental trophoblastic cells could be efficiently transfected with adenoviruses when delivered directly into uterine arteries [192]. On the contrary, and despite their ability to transfect fetal membranes and the basal plate more efficiently than adenoviruses, plasmid/PEI and plasmid/liposome complexes were not as efficient in transfecting placental trophoblastic cells [192]. Similarly, the Westwood group successfully used an adenovirus to deliver sense or antisense IGF-I or IGF-II cDNA into human primary placental fibroblasts (PPF), aiming to study the effects of overexpression or inhibition of these genes on placental cell proliferation, migration, and survival [193]. The authors reported that placenta cells could be efficiently transfected by adenoviruses, verifying the potential therapeutic outcome of gene therapy in placental disorders. Based on the above, Keswani et al. employed the adenoviral vector Ad-IGF-1 in vivo in mice, demonstrating the restoration of birth weight [194]. Similarly, Carr et al. showed that using Ad-VEGF led to an improvement in fetal growth and an increased fetal growth rate [195].

4.4. Embryo Implantation Disorders

The proof of principle that genetic manipulation of the adult uterine endometrium has therapeutic potential against implantation disorders came from Bagot et al. in early 2000 [196]. The authors first reported that the alteration of maternal Hoxa10 expression, previously shown to be expressed in human endometrium during implantation [202] by in vivo gene transfection, affects implantation in mice [196]. Based on this finding, they overexpressed the maternal homeobox A10 (Hoxa10) gene using a pcDNA3.1(+)/HOXA10a, a liposome plasmid that constitutively expresses Hoxa10, and achieved a significant increase in litter size. All of the mice pups were born following a normal gestation of 17–20 days, were normal in size and exhibited no morphological abnormalities. Similarly, Nakamura et al. [197], employing the hemagglutinating virus of Japan envelope vector (HVJ), showed that NF-κB expression determines the timing of implantation via control of LIF expression and that manipulation of its expression may alter the implantation outcome. Microarray-based bioinformatics experiments by Horcajadas et al. [203] led to the identification of a huge number of transcripts regulated during implantation and set the basis for many in vivo gene transfer approaches. Furthermore, a multi-omics approach for the identification of successful implantation biomarkers [204] can be a useful tool for predicting a successful implantation outcome.

5. Clinical Trials

Despite the plethora of published data regarding the pre-clinical evaluation of viral and non-viral gene therapy approaches toward gynaecological cancers, only a small percentage has reached the clinical phase, with most of them not progressing beyond Phase I/II. According to clinicaltrials.gov [101] and using the terms “ovarian cancer”, “cervix cancer”, and “endometrial cancer” in combination with “gene therapy” as keywords, followed by manual exclusion of trials not involving gene therapy approaches, there are 18 clinical trials for ovarian gene therapy, 11 for cervical gene therapy, and three for endometrial gene therapy, with only a few currently recruiting patients. There are no clinical trials for gene therapy approaches against benign gynaecological disorders. Table 4 summarises the clinical trials for ovarian, cervical, and endometrial cancer gene therapy.
Regarding ovarian cancer gene therapy clinical trials, out of the 18 studies, three employed mutation compensation, three suicide gene therapies, and one oncolytic virotherapy strategy, while the remaining 11 used immunopotentiation approaches. In detail, in Phase I NCT00003450 and NCT00003588 clinical trials, Ads alone were used to deliver the p53 gene, while in the Phase II NCT02435186 study, adenoviral-mediated p53 delivery was combined with cisplatin and PTX chemotherapy. None of the above clinical trials have been posted or had their results published yet. With regards to suicide gene therapy approaches, clinical trials NCT00964756, NCT01997190, and NCT00005025 employed adenoviral or HSV vectors to deliver the TK gene in patients with recurrent ovarian cancer. Published data from Phase I clinical trial NCT00964756 using the Ad5-SSTR/TK-R-GD vector highlighted its safety and effectiveness, with five out of twelve patients showing stable disease and one out of five demonstrating complete disease resolution [205]. In the ongoing Phase I/II clinical trial NCT02068794, MSCs are used as carrier cells for oncolytic measles virus encoding thyroidal sodium iodide symporter (MeV-NIS) in patients with ovarian cancer.
Regarding immunopotentiation approaches, in the Phase I clinical trial NCT00066404, adenoviral-mediated IFN-β transfer induced consistent anti-tumour immune responses; however, the rapid production of anti-adenoviral neutralizing antibodies following second dose administration led to minimized target cell transduction and IFN-β production [206]. To circumvent the above problem, the authors plan to administer a second dose as early as 3 days after the first one and combine adenoviral administration with chemotherapy or surgery.
The rest clinical trials involve administration of engineered T cells expressing either neoantigen-specific TCRs (NCT03412877, NCT05194735, NCT03970382, NCT04102436) or anti-carcinoembryonic antigen (CEA) (NCT00004178), anti-mesothelin (NCT01583686) or folate binding protein (NCT00019136) chimeric TCRs. Specifically, for patients with solid tumours, there are two clinical trials (NCT02366546 and NCT02096614) assessing the in vivo kinetics and safety of TBI-1301 (NY-ESO-1-specific TCR) and TBI-1201 (MAGE A4-specific TCR), in combination with cyclophosphamide and fludarabine. All the above approaches employing engineered T cells have gained a lot of ground, leading to promising results toward cancer immunotherapy [207].
Overall, out of the 18 clinical trials described above, 16 employed viral gene delivery systems, while two, specifically the NCT04102436 and NCT00381173 trials, used non-viral approaches, such as the Sleeping Beauty transposon/transposase system or plasmid DNA, respectively. Specifically, in the latter study, the administration of ZYC300, a DNA plasmid encoding the tumour-associated carcinogen activator cytochrome P450 1B1 (CYP1B1) [208], was used to treat 17 patients with advanced-stage progressive cancer and led to CYP1B1 immunity induction and marked response to next regimen in six of them [209].
Regarding the cervical cancer clinical trials, eight out of ten clinical trials employ immunopotentiation approaches, while only two and specifically NCT03544723 and NCT03057912, are based on mutation compensation strategies. Clinical trial NCT03544723 is a Phase II multi-centre study to evaluate adenoviral-mediated p53 delivery in combination with immune checkpoint inhibitors, such as anti-PD-1/anti-PD-L1 in patients with carcinomas approved for anti-PD-1 and anti-PD-L1 therapy. Accumulating evidence presented by Liu et al. shows that PD-1/PD-L1 inhibitors may also be a promising therapeutic approach for cervical cancer as well [210], and therefore the results of the above study (estimated completion date 31/12/2022) will contribute significantly toward efficient cervical cancer treatment. The NCT03057912 Phase I study assesses the safety and efficacy of the in vivo TALEN- and CRISPR/Cas9-mediated disruption of HPV16- and HPV18-E6/E7 oncoproteins.
Regarding clinical trials employing immunopotentiation approaches, in the ongoing clinical trial NCT04180215, checkpoint inhibitors are administered in combination with vectors expressing HPV16+ antigen (HB-201 and HB-202) to assess their efficacy against patients with metastatic HPV16+ cancers, while clinical trial NCT00988559 evaluates the safety and efficacy of different routes of administration of pnGVL4a-CRT/E7 (detox) DNA vaccine in patients with HPV16+ cervical intraepithelial neoplasia (CIN)2/3. While the results of the former study will not become available before summer 2025, results of the latter clinical trial indicate induction of CIN2/3 remission following intramuscular or intralesional delivery of pnGVL4a-CRT/E7 (detox) vaccine. The other cervical cancer clinical trials involve the infusion of T cells engineered to express TCRs against HPV16/18 E6/E7 oncoproteins (NCT02379520 and NCT022280811), DP0401/0402 restricted MAGE-A3 tumour antigen (NCT02153905, NCT02111850), CEA (NCT00004178), or mesothelin (NCT01583686), usually in combination with cyclosphosphamide or fludarabine. The results of the above clinical trials are not published yet; however, the therapeutic potential of the specific approach is quite promising [207]. Lastly, the anti-tumour effect of IFN-β transfer was clearly demonstrated in the NCT00066404 clinical trial [206], and it is expected that the revised experimental design will lead to an even more significant therapeutic outcome.
Regarding endometrial cancer clinical trials, all three (NCT05194735, NTC00004178, NCT00066404) employ immunopotentiation approaches discussed above and involve either infusion of CAR T cells or adenoviral-mediated IFN-β transfer.

6. Conclusions and Future Directions

The aim of this review was to summarise all the major advances regarding gene therapy for gynaecological malignant and benign disorders and highlight their therapeutic potential and their eventual clinical application. It is clear that targeted gene therapy approaches, such as mutation compensation, antiangiogenesis, immunopotentiation, suicide gene therapy, and oncolytic virotherapy, have yielded promising results, employing both viral and non-viral systems. However, most of these strategies either remain in the preclinical phase or show reduced effectiveness as monotherapies and usually require a combination of chemotherapy and radiotherapy to demonstrate a therapeutic outcome. Furthermore, gene therapy in combination with immunotherapy, e.g., targeting CTLA-4 and PD-1, or with the emerging therapeutic angiogenesis inhibitors, may result in increased effectiveness, especially for ovarian cancer in advanced stages, where still no effective therapies exist. Moreover, there is increasing evidence for the therapeutic potential of CAR T-cell technology also, and clinical trials are currently assessing its efficacy, primarily in ovarian cancer, by recruiting patients [101] (Table 4). The continuous development of safe and effective vectors, the discovery of novel diagnostic markers, such as miRNAs discussed above, or of new players, such as long non-coding RNAs (lncRNAs) [211] and circular RNAs (circRNAs) [212] and in combination with more clinical trials beyond Phase I/II are expected to lead to more effective and radical therapeutic outcomes.

Author Contributions

E.D.: drafting the manuscript; E.D.: figure preparation; E.D., N.P.A. and K.I.P.: Manuscript Revision & Editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by a Grant No. 3624 from the Hellenic Foundation for Research and Innovation (H.F.R.I.), Athens, Greece.

Acknowledgments

All figures were created with Biorender.com (accessed on 22 June 2022).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Brody, H. Gene therapy. Nature 2018, 564, S5. [Google Scholar] [CrossRef] [PubMed]
  2. Ayen, A.; Jimenez Martinez, Y.; Marchal, J.A.; Boulaiz, H. Recent Progress in Gene Therapy for Ovarian Cancer. Int. J. Mol. Sci. 2018, 19, 1930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Ayen, A.; Jimenez Martinez, Y.; Boulaiz, H. Targeted Gene Delivery Therapies for Cervical Cancer. Cancers 2020, 12, 1301. [Google Scholar] [CrossRef] [PubMed]
  4. Brooks, R.A.; Fleming, G.F.; Lastra, R.R.; Lee, N.K.; Moroney, J.W.; Son, C.H.; Tatebe, K.; Veneris, J.L. Current recommendations and recent progress in endometrial cancer. CA Cancer J. Clin. 2019, 69, 258–279. [Google Scholar] [CrossRef]
  5. Hassan, M.H.; Othman, E.E.; Hornung, D.; Al-Hendy, A. Gene therapy of benign gynecological diseases. Adv. Drug Deliv. Rev. 2009, 61, 822–835. [Google Scholar] [CrossRef] [Green Version]
  6. Brooks, R.A.; Mutch, D.G. Gene therapy in gynecological cancer. Expert Rev. Anticancer Ther. 2006, 6, 1013–1032. [Google Scholar] [CrossRef]
  7. Naldini, L.; Blomer, U.; Gallay, P.; Ory, D.; Mulligan, R.; Gage, F.H.; Verma, I.M.; Trono, D. In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector. Science 1996, 272, 263–267. [Google Scholar] [CrossRef] [Green Version]
  8. Naldini, L.; Blomer, U.; Gage, F.H.; Trono, D.; Verma, I.M. Efficient transfer, integration, and sustained long-term expression of the transgene in adult rat brains injected with a lentiviral vector. Proc. Natl. Acad. Sci. USA 1996, 93, 11382–11388. [Google Scholar] [CrossRef] [Green Version]
  9. Bulcha, J.T.; Wang, Y.; Ma, H.; Tai, P.W.L.; Gao, G. Viral vector platforms within the gene therapy landscape. Signal Transduct. Target 2021, 6, 53. [Google Scholar] [CrossRef]
  10. Greber, U.F. Adenoviruses—Infection, pathogenesis and therapy. FEBS Lett. 2020, 594, 1818–1827. [Google Scholar] [CrossRef]
  11. Muruve, D.A. The innate immune response to adenovirus vectors. Hum. Gene Ther. 2004, 15, 1157–1166. [Google Scholar] [CrossRef]
  12. Chen, H.; Xiang, Z.Q.; Li, Y.; Kurupati, R.K.; Jia, B.; Bian, A.; Zhou, D.M.; Hutnick, N.; Yuan, S.; Gray, C.; et al. Adenovirus-based vaccines: Comparison of vectors from three species of adenoviridae. J. Virol. 2010, 84, 10522–10532. [Google Scholar] [CrossRef] [Green Version]
  13. Ramamoorth, M.; Narvekar, A. Non viral vectors in gene therapy—An overview. J. Clin. Diagn. Res. 2015, 9, GE01. [Google Scholar] [CrossRef]
  14. Lin, L.T.; Richardson, C.D. The Host Cell Receptors for Measles Virus and Their Interaction with the Viral Hemagglutinin (H) Protein. Viruses 2016, 8, 250. [Google Scholar] [CrossRef]
  15. Engeland, C.E.; Ungerechts, G. Measles Virus as an Oncolytic Immunotherapy. Cancers 2021, 13, 544. [Google Scholar] [CrossRef]
  16. Lachmann, R. Herpes simplex virus-based vectors. Int. J. Exp. Pathol. 2004, 85, 177–190. [Google Scholar] [CrossRef]
  17. Zu, H.; Gao, D. Non-viral Vectors in Gene Therapy: Recent Development, Challenges, and Prospects. AAPS J. 2021, 23, 78. [Google Scholar] [CrossRef]
  18. Sung, Y.K.; Kim, S.W. Recent advances in the development of gene delivery systems. Biomater. Res. 2019, 23, 8. [Google Scholar] [CrossRef]
  19. Mitchell, M.J.; Billingsley, M.M.; Haley, R.M.; Wechsler, M.E.; Peppas, N.A.; Langer, R. Engineering precision nanoparticles for drug delivery. Nat. Rev. Drug. Discov. 2021, 20, 101–124. [Google Scholar] [CrossRef]
  20. Al-Dosari, M.S.; Gao, X. Nonviral gene delivery: Principle, limitations, and recent progress. AAPS J. 2009, 11, 671–681. [Google Scholar] [CrossRef] [Green Version]
  21. Zelmer, C.; Zweifel, L.P.; Kapinos, L.E.; Craciun, I.; Guven, Z.P.; Palivan, C.G.; Lim, R.Y.H. Organelle-specific targeting of polymersomes into the cell nucleus. Proc. Natl. Acad. Sci. USA 2020, 117, 2770–2778. [Google Scholar] [CrossRef] [Green Version]
  22. Lee, S.W.; Kim, Y.M.; Cho, C.H.; Kim, Y.T.; Kim, S.M.; Hur, S.Y.; Kim, J.H.; Kim, B.G.; Kim, S.C.; Ryu, H.S.; et al. An Open-Label, Randomized, Parallel, Phase II Trial to Evaluate the Efficacy and Safety of a Cremophor-Free Polymeric Micelle Formulation of Paclitaxel as First-Line Treatment for Ovarian Cancer: A Korean Gynecologic Oncology Group Study (KGOG-3021). Cancer Res. Treat. 2018, 50, 195–203. [Google Scholar] [CrossRef]
  23. Palmerston Mendes, L.; Pan, J.; Torchilin, V.P. Dendrimers as Nanocarriers for Nucleic Acid and Drug Delivery in Cancer Therapy. Molecules 2017, 22, 1401. [Google Scholar] [CrossRef] [Green Version]
  24. Vines, J.B.; Yoon, J.H.; Ryu, N.E.; Lim, D.J.; Park, H. Gold Nanoparticles for Photothermal Cancer Therapy. Front. Chem. 2019, 7, 167. [Google Scholar] [CrossRef] [Green Version]
  25. Matulonis, U.A.; Sood, A.K.; Fallowfield, L.; Howitt, B.E.; Sehouli, J.; Karlan, B.Y. Ovarian cancer. Nat. Rev. Dis. Primers 2016, 2, 16061. [Google Scholar] [CrossRef]
  26. Oswald, A.J.; Gourley, C. Low-grade epithelial ovarian cancer: A number of distinct clinical entities? Curr. Opin. Oncol. 2015, 27, 412–419. [Google Scholar] [CrossRef]
  27. Rawlinson, J.W.; Vaden, K.; Hunsaker, J.; Miller, D.F.; Nephew, K.P. Adenoviral-delivered HE4-HSV-tk sensitizes ovarian cancer cells to ganciclovir. Gene Ther. Mol. Biol. 2013, 15, 120–130. [Google Scholar]
  28. Zhou, X.L.; Shi, Y.L.; Li, X. Inhibitory effects of the ultrasound-targeted microbubble destruction-mediated herpes simplex virus-thymidine kinase/ganciclovir system on ovarian cancer in mice. Exp. Ther. Med. 2014, 8, 1159–1163. [Google Scholar] [CrossRef] [Green Version]
  29. White, C.L.; Menghistu, T.; Twigger, K.R.; Searle, P.F.; Bhide, S.A.; Vile, R.G.; Melcher, A.A.; Pandha, H.S.; Harrington, K.J. Escherichia coli nitroreductase plus CB1954 enhances the effect of radiotherapy in vitro and in vivo. Gene Ther. 2008, 15, 424–433. [Google Scholar] [CrossRef]
  30. Hampl, M.; Tanaka, T.; Albert, P.S.; Lee, J.; Ferrari, N.; Fine, H.A. Therapeutic effects of viral vector-mediated antiangiogenic gene transfer in malignant ascites. Hum. Gene Ther. 2001, 12, 1713–1729. [Google Scholar] [CrossRef]
  31. Yang, L.; Wang, L.; Su, X.Q.; Wang, L.; Chen, X.C.; Li, D.; Luo, S.T.; Shi, H.S.; Chen, L.J.; Wang, Y.S. Suppression of ovarian cancer growth via systemic administration with liposome-encapsulated adenovirus-encoding endostatin. Cancer Gene Ther. 2010, 17, 49–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Tuppurainen, L.; Sallinen, H.; Karvonen, A.; Valkonen, E.; Laakso, H.; Liimatainen, T.; Hytonen, E.; Hamalainen, K.; Kosma, V.M.; Anttila, M.; et al. Combined Gene Therapy Using AdsVEGFR2 and AdsTie2 with Chemotherapy Reduces the Growth of Human Ovarian Cancer and Formation of Ascites in Mice. Int. J. Gynecol. Cancer 2017, 27, 879–886. [Google Scholar] [CrossRef] [PubMed]
  33. Subramanian, I.V.; Ghebre, R.; Ramakrishnan, S. Adeno-associated virus-mediated delivery of a mutant endostatin suppresses ovarian carcinoma growth in mice. Gene Ther. 2005, 12, 30–38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Bui Nguyen, T.M.; Subramanian, I.V.; Xiao, X.; Nguyen, P.; Ramakrishnan, S. Adeno-associated virus-mediated delivery of kringle 5 of human plasminogen inhibits orthotopic growth of ovarian cancer. Gene Ther. 2010, 17, 606–615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Xie, Y.; Hicks, M.J.; Kaminsky, S.M.; Moore, M.A.; Crystal, R.G.; Rafii, A. AAV-mediated persistent bevacizumab therapy suppresses tumor growth of ovarian cancer. Gynecol. Oncol. 2014, 135, 325–332. [Google Scholar] [CrossRef] [Green Version]
  36. Dai, Y.; Zhao, X.J.; Li, F.; Yuan, Y.; Yan, D.M.; Cao, H.; Huang, X.Y.; Hu, Z.; Ma, D.; Gao, Q.L. Truncated Bid Regulates Cisplatin Response via Activation of Mitochondrial Apoptosis Pathway in Ovarian Cancer. Hum. Gene Ther. 2020, 31, 325–338. [Google Scholar] [CrossRef]
  37. Zhang, W.W.; Li, L.; Li, D.; Liu, J.; Li, X.; Li, W.; Xu, X.; Zhang, M.J.; Chandler, L.A.; Lin, H.; et al. The First Approved Gene Therapy Product for Cancer Ad-p53 (Gendicine): 12 Years in the Clinic. Hum. Gene Ther. 2018, 29, 160–179. [Google Scholar] [CrossRef] [Green Version]
  38. Liu, Y.G.; Zheng, X.L.; Liu, F.M. The mechanism and inhibitory effect of recombinant human P53 adenovirus injection combined with paclitaxel on human cervical cancer cell HeLa. Eur. Rev. Med. Pharmacol. Sci. 2015, 19, 1037–1042. [Google Scholar]
  39. Hanauer, J.R.; Gottschlich, L.; Riehl, D.; Rusch, T.; Koch, V.; Friedrich, K.; Hutzler, S.; Prufer, S.; Friedel, T.; Hanschmann, K.M.; et al. Enhanced lysis by bispecific oncolytic measles viruses simultaneously using HER2/neu or EpCAM as target receptors. Mol. Ther. Oncolytics 2016, 3, 16003. [Google Scholar] [CrossRef]
  40. Thomas, E.D.; Meza-Perez, S.; Bevis, K.S.; Randall, T.D.; Gillespie, G.Y.; Langford, C.; Alvarez, R.D. IL-12 Expressing oncolytic herpes simplex virus promotes anti-tumor activity and immunologic control of metastatic ovarian cancer in mice. J. Ovarian Res. 2016, 9, 70. [Google Scholar] [CrossRef]
  41. Goshima, F.; Esaki, S.; Luo, C.; Kamakura, M.; Kimura, H.; Nishiyama, Y. Oncolytic viral therapy with a combination of HF10, a herpes simplex virus type 1 variant and granulocyte-macrophage colony-stimulating factor for murine ovarian cancer. Int. J. Cancer 2014, 134, 2865–2877. [Google Scholar] [CrossRef]
  42. Dold, C.; Rodriguez Urbiola, C.; Wollmann, G.; Egerer, L.; Muik, A.; Bellmann, L.; Fiegl, H.; Marth, C.; Kimpel, J.; von Laer, D. Application of interferon modulators to overcome partial resistance of human ovarian cancers to VSV-GP oncolytic viral therapy. Mol. Ther. Oncolytics 2016, 3, 16021. [Google Scholar] [CrossRef]
  43. Han, Z.; Hong, Z.; Gao, Q.; Chen, C.; Hao, Z.; Ji, T.; Hu, W.; Yan, Y.; Feng, J.; Liao, S.; et al. A potent oncolytic adenovirus selectively blocks the STAT3 signaling pathway and potentiates cisplatin antitumor activity in ovarian cancer. Hum. Gene Ther. 2012, 23, 32–45. [Google Scholar] [CrossRef]
  44. Wang, S.; Shu, J.; Chen, L.; Chen, X.; Zhao, J.; Li, S.; Mou, X.; Tong, X. Synergistic suppression effect on tumor growth of ovarian cancer by combining cisplatin with a manganese superoxide dismutase-armed oncolytic adenovirus. Onco Targets Ther. 2016, 9, 6381–6388. [Google Scholar] [CrossRef] [Green Version]
  45. Hartkopf, A.D.; Bossow, S.; Lampe, J.; Zimmermann, M.; Taran, F.A.; Wallwiener, D.; Fehm, T.; Bitzer, M.; Lauer, U.M. Enhanced killing of ovarian carcinoma using oncolytic measles vaccine virus armed with a yeast cytosine deaminase and uracil phosphoribosyltransferase. Gynecol. Oncol. 2013, 130, 362–368. [Google Scholar] [CrossRef]
  46. Yu, Y.; Pilgrim, P.; Zhou, W.; Gagliano, N.; Frezza, E.E.; Jenkins, M.; Weidanz, J.A.; Lustgarten, J.; Cannon, M.; Bumm, K.; et al. rAAV/Her-2/neu loading of dendritic cells for a potent cellular-mediated MHC class I restricted immune response against ovarian cancer. Viral. Immunol. 2008, 21, 435–442. [Google Scholar] [CrossRef]
  47. Zhang, Y.; Wang, J.; Ren, M.; Li, M.; Chen, D.; Chen, J.; Shi, F.; Wang, X.; Dou, J. Gene therapy of ovarian cancer using IL-21-secreting human umbilical cord mesenchymal stem cells in nude mice. J. Ovarian. Res. 2014, 7, 8. [Google Scholar] [CrossRef] [Green Version]
  48. Shi, X.X.; Zhang, B.; Zang, J.L.; Wang, G.Y.; Gao, M.H. CD59 silencing via retrovirus-mediated RNA interference enhanced complement-mediated cell damage in ovary cancer. Cell. Mol. Immunol. 2009, 6, 61–66. [Google Scholar] [CrossRef] [Green Version]
  49. Lin, Y.; Peng, S.; Yu, H.; Teng, H.; Cui, M. RNAi-mediated downregulation of NOB1 suppresses the growth and colony-formation ability of human ovarian cancer cells. Med. Oncol. 2012, 29, 311–317. [Google Scholar] [CrossRef]
  50. Sher, Y.P.; Chang, C.M.; Juo, C.G.; Chen, C.T.; Hsu, J.L.; Lin, C.Y.; Han, Z.; Shiah, S.G.; Hung, M.C. Targeted endostatin-cytosine deaminase fusion gene therapy plus 5-fluorocytosine suppresses ovarian tumor growth. Oncogene 2013, 32, 1082–1090. [Google Scholar] [CrossRef]
  51. Bai, Y.; Gou, M.; Yi, T.; Yang, L.; Liu, L.; Lin, X.; Su, D.; Wei, Y.; Zhao, X. Efficient Inhibition of Ovarian Cancer by Gelonin Toxin Gene Delivered by Biodegradable Cationic Heparin-polyethyleneimine Nanogels. Int. J. Med. Sci. 2015, 12, 397–406. [Google Scholar] [CrossRef] [Green Version]
  52. Huang, Y.H.; Zugates, G.T.; Peng, W.; Holtz, D.; Dunton, C.; Green, J.J.; Hossain, N.; Chernick, M.R.; Padera, R.F., Jr.; Langer, R.; et al. Nanoparticle-delivered suicide gene therapy effectively reduces ovarian tumor burden in mice. Cancer Res. 2009, 69, 6184–6191. [Google Scholar] [CrossRef] [Green Version]
  53. Cocco, E.; Deng, Y.; Shapiro, E.M.; Bortolomai, I.; Lopez, S.; Lin, K.; Bellone, S.; Cui, J.; Menderes, G.; Black, J.D.; et al. Dual-Targeting Nanoparticles for In Vivo Delivery of Suicide Genes to Chemotherapy-Resistant Ovarian Cancer Cells. Mol. Cancer Ther. 2017, 16, 323–333. [Google Scholar] [CrossRef] [Green Version]
  54. Sun, Z.; Huang, J.; Su, L.; Li, J.; Qi, F.; Su, H.; Chen, Y.; Zhang, Q.; Zhang, Q.; Li, Z.; et al. Arf6-mediated macropinocytosis-enhanced suicide gene therapy of C16TAB-condensed Tat/pDNA nanoparticles in ovarian cancer. Nanoscale 2021, 13, 14538–14551. [Google Scholar] [CrossRef]
  55. He, Z.Y.; Deng, F.; Wei, X.W.; Ma, C.C.; Luo, M.; Zhang, P.; Sang, Y.X.; Liang, X.; Liu, L.; Qin, H.X.; et al. Ovarian cancer treatment with a tumor-targeting and gene expression-controllable lipoplex. Sci. Rep. 2016, 6, 23764. [Google Scholar] [CrossRef] [Green Version]
  56. Kobayashi, A.; Yokoyama, Y.; Osawa, Y.; Miura, R.; Mizunuma, H. Gene therapy for ovarian cancer using carbonyl reductase 1 DNA with a polyamidoamine dendrimer in mouse models. Cancer Gene Ther. 2016, 23, 24–28. [Google Scholar] [CrossRef] [Green Version]
  57. Hu, W.; Wang, J.; Dou, J.; He, X.; Zhao, F.; Jiang, C.; Yu, F.; Hu, K.; Chu, L.; Li, X.; et al. Augmenting therapy of ovarian cancer efficacy by secreting IL-21 human umbilical cord blood stem cells in nude mice. Cell Transplant. 2011, 20, 669–680. [Google Scholar] [CrossRef]
  58. Fewell, J.G.; Matar, M.M.; Rice, J.S.; Brunhoeber, E.; Slobodkin, G.; Pence, C.; Worker, M.; Lewis, D.H.; Anwer, K. Treatment of disseminated ovarian cancer using nonviral interleukin-12 gene therapy delivered intraperitoneally. J. Gene Med. 2009, 11, 718–728. [Google Scholar] [CrossRef]
  59. Yang, F.; Li, Z.; Deng, H.; Yang, H.; Yan, F.; Qian, Z.; Chen, L.; Wei, Y.; Zhao, X. Efficient inhibition of ovarian cancer growth and prolonged survival by transfection with a novel pro-apoptotic gene, hPNAS-4, in a mouse model. In vivo and in vitro results. Oncology 2008, 75, 137–144. [Google Scholar] [CrossRef]
  60. Wu, H.; Wang, S.; Weng, D.; Xing, H.; Song, X.; Zhu, T.; Xia, X.; Weng, Y.; Xu, G.; Meng, L.; et al. Reversal of the malignant phenotype of ovarian cancer A2780 cells through transfection with wild-type PTEN gene. Cancer Lett. 2008, 271, 205–214. [Google Scholar] [CrossRef]
  61. Lee, M.H.; Choi, B.Y.; Cho, Y.Y.; Lee, S.Y.; Huang, Z.; Kundu, J.K.; Kim, M.O.; Kim, D.J.; Bode, A.M.; Surh, Y.J.; et al. Tumor suppressor p16(INK4a) inhibits cancer cell growth by downregulating eEF1A2 through a direct interaction. J. Cell Sci. 2013, 126, 1744–1752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Lu, F.; Xu, H.; Wang, Q.; Li, M.; Meng, J.; Kuang, Y. Inhibition of enhancer of zeste homolog 2 increases the expression of p16 and suppresses the proliferation and migration of ovarian carcinoma cells in vitro and in vivo. Oncol. Lett. 2018, 15, 3233–3239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Yan, H.; Tong, J.; Lin, X.; Han, Q.; Huang, H. Effect of the WWOX gene on the regulation of the cell cycle and apoptosis in human ovarian cancer stem cells. Mol. Med. Rep. 2015, 12, 1783–1788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Zhao, Y.; Wang, W.; Pan, W.; Yu, Y.; Huang, W.; Gao, J.; Zhang, Y.; Zhang, S. WWOX promotes apoptosis and inhibits autophagy in paclitaxeltreated ovarian carcinoma cells. Mol. Med. Rep. 2021, 23, 1. [Google Scholar] [CrossRef] [PubMed]
  65. Xiong, Z.; Hu, S.; Wang, Z. Cloning of WWOX gene and its growth-inhibiting effects on ovarian cancer cells. J. Huazhong. Univ. Sci. Technol. Med. Sci. 2010, 30, 365–369. [Google Scholar] [CrossRef]
  66. Dickerson, E.B.; Blackburn, W.H.; Smith, M.H.; Kapa, L.B.; Lyon, L.A.; McDonald, J.F. Chemosensitization of cancer cells by siRNA using targeted nanogel delivery. BMC Cancer 2010, 10, 10. [Google Scholar] [CrossRef] [Green Version]
  67. Zhang, R.; Shi, H.; Chen, Z.; Wu, Q.; Ren, F.; Huang, H. Effects of metastasis-associated in colon cancer 1 inhibition by small hairpin RNA on ovarian carcinoma OVCAR-3 cells. J. Exp. Clin. Cancer Res. 2011, 30, 83. [Google Scholar] [CrossRef] [Green Version]
  68. Rao, Y.M.; Ji, M.; Chen, C.H.; Shi, H.R. Effect of siRNA targeting MTA1 on metastasis malignant phenotype of ovarian cancer A2780 cells. J. Huazhong Univ. Sci. Technol. Med. Sci. 2013, 33, 266–271. [Google Scholar] [CrossRef]
  69. Lin, Y.; Cui, M.; Xu, T.; Yu, W.; Zhang, L. Silencing of cyclooxygenase-2 inhibits the growth, invasion and migration of ovarian cancer cells. Mol. Med. Rep. 2014, 9, 2499–2504. [Google Scholar] [CrossRef] [Green Version]
  70. Guo, F.J.; Tian, J.Y.; Jin, Y.M.; Wang, L.; Yang, R.Q.; Cui, M.H. Effects of cyclooxygenase-2 gene silencing on the biological behavior of SKOV3 ovarian cancer cells. Mol. Med. Rep. 2015, 11, 59–66. [Google Scholar] [CrossRef] [Green Version]
  71. Huo, X.; Ren, L.; Shang, L.; Wang, X.; Wang, J. Effect of WT1 antisense mRNA on the induction of apoptosis in ovarian carcinoma SKOV3 cells. Eur. J. Gynaecol. Oncol. 2011, 32, 651–656. [Google Scholar]
  72. Jiang, Q.; Dai, L.; Cheng, L.; Chen, X.; Li, Y.; Zhang, S.; Su, X.; Zhao, X.; Wei, Y.; Deng, H. Efficient inhibition of intraperitoneal ovarian cancer growth in nude mice by liposomal delivery of short hairpin RNA against STAT3. J. Obstet. Gynaecol. Res. 2013, 39, 701–709. [Google Scholar] [CrossRef]
  73. Zhao, Y.C.; Zhang, L.; Feng, S.S.; Hong, L.; Zheng, H.L.; Chen, L.L.; Zheng, X.L.; Ye, Y.Q.; Zhao, M.D.; Wang, W.X.; et al. Efficient delivery of Notch1 siRNA to SKOV3 cells by cationic cholesterol derivative-based liposome. Int. J. Nanomed. 2016, 11, 5485–5496. [Google Scholar] [CrossRef] [Green Version]
  74. Huang, Y.H.; Bao, Y.; Peng, W.; Goldberg, M.; Love, K.; Bumcrot, D.A.; Cole, G.; Langer, R.; Anderson, D.G.; Sawicki, J.A. Claudin-3 gene silencing with siRNA suppresses ovarian tumor growth and metastasis. Proc. Natl. Acad. Sci. USA 2009, 106, 3426–3430. [Google Scholar] [CrossRef] [Green Version]
  75. Sun, C.; Yi, T.; Song, X.; Li, S.; Qi, X.; Chen, X.; Lin, H.; He, X.; Li, Z.; Wei, Y.; et al. Efficient inhibition of ovarian cancer by short hairpin RNA targeting claudin-3. Oncol. Rep. 2011, 26, 193–200. [Google Scholar] [CrossRef] [Green Version]
  76. Li, T.S.; Yawata, T.; Honke, K. Efficient siRNA delivery and tumor accumulation mediated by ionically cross-linked folic acid-poly(ethylene glycol)-chitosan oligosaccharide lactate nanoparticles: For the potential targeted ovarian cancer gene therapy. Eur. J. Pharm. Sci. 2014, 52, 48–61. [Google Scholar] [CrossRef] [Green Version]
  77. He, Z.Y.; Zhang, Y.G.; Yang, Y.H.; Ma, C.C.; Wang, P.; Du, W.; Li, L.; Xiang, R.; Song, X.R.; Zhao, X.; et al. In Vivo Ovarian Cancer Gene Therapy Using CRISPR-Cas9. Hum. Gene Ther. 2018, 29, 223–233. [Google Scholar] [CrossRef]
  78. Nakamura, M.; Obata, T.; Daikoku, T.; Fujiwara, H. The Association and Significance of p53 in Gynecologic Cancers: The Potential of Targeted Therapy. Int. J. Mol. Sci. 2019, 20, 5482. [Google Scholar] [CrossRef] [Green Version]
  79. Teplinsky, E.; Muggia, F. Targeting HER2 in ovarian and uterine cancers: Challenges and future directions. Gynecol. Oncol. 2014, 135, 364–370. [Google Scholar] [CrossRef]
  80. Bookman, M.A.; Darcy, K.M.; Clarke-Pearson, D.; Boothby, R.A.; Horowitz, I.R. Evaluation of monoclonal humanized anti-HER2 antibody, trastuzumab, in patients with recurrent or refractory ovarian or primary peritoneal carcinoma with overexpression of HER2: A phase II trial of the Gynecologic Oncology Group. J. Clin. Oncol. 2003, 21, 283–290. [Google Scholar] [CrossRef]
  81. Jiang, L.; Luo, R.Y.; Yang, J.; Cheng, Y.X. Knockdown of survivin contributes to antitumor activity in cisplatin-resistant ovarian cancer cells. Mol. Med. Rep. 2013, 7, 425–430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Lu, J.; Getz, G.; Miska, E.A.; Alvarez-Saavedra, E.; Lamb, J.; Peck, D.; Sweet-Cordero, A.; Ebert, B.L.; Mak, R.H.; Ferrando, A.A.; et al. MicroRNA expression profiles classify human cancers. Nature 2005, 435, 834–838. [Google Scholar] [CrossRef] [PubMed]
  83. Chen, S.N.; Chang, R.; Lin, L.T.; Chern, C.U.; Tsai, H.W.; Wen, Z.H.; Li, Y.H.; Li, C.J.; Tsui, K.H. MicroRNA in Ovarian Cancer: Biology, Pathogenesis, and Therapeutic Opportunities. Int. J. Environ. Res. Public Health 2019, 16, 1510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Iorio, M.V.; Visone, R.; Di Leva, G.; Donati, V.; Petrocca, F.; Casalini, P.; Taccioli, C.; Volinia, S.; Liu, C.G.; Alder, H.; et al. MicroRNA signatures in human ovarian cancer. Cancer Res. 2007, 67, 8699–8707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Yang, H.; Kong, W.; He, L.; Zhao, J.J.; O’Donnell, J.D.; Wang, J.; Wenham, R.M.; Coppola, D.; Kruk, P.A.; Nicosia, S.V.; et al. MicroRNA expression profiling in human ovarian cancer: miR-214 induces cell survival and cisplatin resistance by targeting PTEN. Cancer Res. 2008, 68, 425–433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Lan, H.; Chen, W.; He, G.; Yang, S. miR-140-5p inhibits ovarian cancer growth partially by repression of PDGFRA. Biomed. Pharmacother. 2015, 75, 117–122. [Google Scholar] [CrossRef]
  87. Echevarria-Vargas, I.M.; Valiyeva, F.; Vivas-Mejia, P.E. Upregulation of miR-21 in cisplatin resistant ovarian cancer via JNK-1/c-Jun pathway. PLoS ONE 2014, 9, e97094. [Google Scholar] [CrossRef]
  88. Dai, F.; Zhang, Y.; Zhu, X.; Shan, N.; Chen, Y. Anticancer role of MUC1 aptamer-miR-29b chimera in epithelial ovarian carcinoma cells through regulation of PTEN methylation. Target. Oncol. 2012, 7, 217–225. [Google Scholar] [CrossRef]
  89. Bussing, I.; Slack, F.J.; Grosshans, H. let-7 microRNAs in development, stem cells and cancer. Trends Mol. Med. 2008, 14, 400–409. [Google Scholar] [CrossRef]
  90. Lu, L.; Schwartz, P.; Scarampi, L.; Rutherford, T.; Canuto, E.M.; Yu, H.; Katsaros, D. MicroRNA let-7a: A potential marker for selection of paclitaxel in ovarian cancer management. Gynecol. Oncol. 2011, 122, 366–371. [Google Scholar] [CrossRef]
  91. Pecot, C.V.; Rupaimoole, R.; Yang, D.; Akbani, R.; Ivan, C.; Lu, C.; Wu, S.; Han, H.D.; Shah, M.Y.; Rodriguez-Aguayo, C.; et al. Tumour angiogenesis regulation by the miR-200 family. Nat. Commun. 2013, 4, 2427. [Google Scholar] [CrossRef] [Green Version]
  92. Li, J.; Liang, S.H.; Lu, X. Potential role of ezrin and its related microRNA in ovarian cancer invasion and metastasis. Zhonghua Fu Chan Ke Za Zhi 2010, 45, 787–792. [Google Scholar]
  93. Aqeilan, R.I.; Calin, G.A.; Croce, C.M. miR-15a and miR-16-1 in cancer: Discovery, function and future perspectives. Cell Death. Differ. 2010, 17, 215–220. [Google Scholar] [CrossRef] [Green Version]
  94. Luo, P.; Fei, J.; Zhou, J.; Zhang, W. microRNA-126 suppresses PAK4 expression in ovarian cancer SKOV3 cells. Oncol. Lett. 2015, 9, 2225–2229. [Google Scholar] [CrossRef] [Green Version]
  95. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
  96. Yan, C.M.; Zhao, Y.L.; Cai, H.Y.; Miao, G.Y.; Ma, W. Blockage of PTPRJ promotes cell growth and resistance to 5-FU through activation of JAK1/STAT3 in the cervical carcinoma cell line C33A. Oncol. Rep. 2015, 33, 1737–1744. [Google Scholar] [CrossRef] [Green Version]
  97. Zhou, S.; Xiao, Y.; Zhuang, Y.; Liu, Y.; Zhao, H.; Yang, H.; Xie, C.; Zhou, F.; Zhou, Y. Knockdown of homeobox containing 1 increases the radiosensitivity of cervical cancer cells through telomere shortening. Oncol. Rep. 2017, 38, 515–521. [Google Scholar] [CrossRef]
  98. Lv, X.F.; Hong, H.Q.; Liu, L.; Cui, S.H.; Ren, C.C.; Li, H.Y.; Zhang, X.A.; Zhang, L.D.; Wei, T.X.; Liu, J.J.; et al. RNAimediated downregulation of asparaginaselike protein 1 inhibits growth and promotes apoptosis of human cervical cancer line SiHa. Mol. Med. Rep. 2018, 18, 931–937. [Google Scholar] [CrossRef]
  99. Qi, L.; Xing, L.N.; Wei, X.; Song, S.G. Effects of VEGF suppression by small hairpin RNA interference combined with radiotherapy on the growth of cervical cancer. Genet. Mol. Res. 2014, 13, 5094–5106. [Google Scholar] [CrossRef]
  100. Jin, B.Y.; Campbell, T.E.; Draper, L.M.; Stevanovic, S.; Weissbrich, B.; Yu, Z.; Restifo, N.P.; Rosenberg, S.A.; Trimble, C.L.; Hinrichs, C.S. Engineered T cells targeting E7 mediate regression of human papillomavirus cancers in a murine model. JCI Insight 2018, 3, e99488. [Google Scholar] [CrossRef]
  101. U.S National Library of Medicine. ClinicalTrials.gov. Available online: https://clinicaltrials.gov (accessed on 29 May 2022).
  102. Zhang, H.; Liao, Z.K.; Sun, W.J.; Huang, C.; Xiong, J.; Zhou, F.X.; Xie, C.H.; Zhou, Y.F. Enhanced suicide gene therapy using a tumor-specific promoter in combination with cisplatin. Mol. Med. Rep. 2009, 2, 1017–1022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Hao, Y.; Guo, L.; Abudula, A.; Saidoula, W.; Guo, X. Proliferation inhibition and apoptosis enhancement of human cervical cancer cells by ultrasound-targeted microbubble destruction delivered double suicide genes. Int. J. Clin. Exp. Med. 2014, 7, 5330–5335. [Google Scholar] [PubMed]
  104. Chen, D.; Tang, Q. An experimental study on cervix cancer with combination of HSV-TK/GCV suicide gene therapy system and 60Co radiotherapy. BMC Cancer 2010, 10, 609. [Google Scholar] [CrossRef] [Green Version]
  105. Su, X.; Chen, W.J.; Xiao, S.W.; Li, X.F.; Xu, G.; Pan, J.J.; Zhang, S.W. Effect and Safety of Recombinant Adenovirus-p53 Transfer Combined with Radiotherapy on Long-Term Survival of Locally Advanced Cervical Cancer. Hum. Gene Ther. 2016, 27, 1008–1014. [Google Scholar] [CrossRef] [PubMed]
  106. Kajitani, K.; Honda, K.; Terada, H.; Yasui, T.; Sumi, T.; Koyama, M.; Ishiko, O. Human Papillomavirus E6 Knockdown Restores Adenovirus Mediated-estrogen Response Element Linked p53 Gene Transfer in HeLa Cells. Asian Pac. J. Cancer Prev. 2015, 16, 8239–8245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Au, W.W.; Abdou-Salama, S.; Al-Hendy, A. Inhibition of growth of cervical cancer cells using a dominant negative estrogen receptor gene. Gynecol. Oncol. 2007, 104, 276–280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Heo, M.Y.; Salama, S.A.; Khatoon, N.; Al-Hendy, A.; Au, W.W. Abrogation of estrogen receptor signaling augments cytotoxicity of anticancer drugs on CaSki cervical cancer cells. Anticancer Res. 2008, 28, 2181–2187. [Google Scholar]
  109. Li, W.Y.; Li, Q.W.; Han, Z.S.; Jiang, Z.L.; Yang, H.; Li, J.; Zhang, X.B. Growth suppression effects of recombinant adenovirus expressing human lactoferrin on cervical cancer in vitro and in vivo. Cancer Biother. Radiopharm. 2011, 26, 477–483. [Google Scholar] [CrossRef]
  110. Wang, W.; Xia, X.; Wang, S.; Sima, N.; Li, Y.; Han, Z.; Gao, Q.; Luo, A.; Li, K.; Meng, L.; et al. Oncolytic adenovirus armed with human papillomavirus E2 gene in combination with radiation demonstrates synergistic enhancements of antitumor efficacy. Cancer Gene Ther. 2011, 18, 825–836. [Google Scholar] [CrossRef]
  111. Wang, W.; Sima, N.; Kong, D.; Luo, A.; Gao, Q.; Liao, S.; Li, W.; Han, L.; Wang, J.; Wang, S.; et al. Selective targeting of HPV-16 E6/E7 in cervical cancer cells with a potent oncolytic adenovirus and its enhanced effect with radiotherapy in vitro and vivo. Cancer Lett. 2010, 291, 67–75. [Google Scholar] [CrossRef]
  112. Wang, H.; Song, X.; Zhang, H.; Zhang, J.; Shen, X.; Zhou, Y.; Fan, X.; Dai, L.; Qian, G.; Hoffman, A.R.; et al. Potentiation of tumor radiotherapy by a radiation-inducible oncolytic and oncoapoptotic adenovirus in cervical cancer xenografts. Int. J. Cancer 2012, 130, 443–453. [Google Scholar] [CrossRef] [Green Version]
  113. Duan, Y.; Bai, H.; Li, X.; Wang, D.; Wang, Y.; Cao, M.; Zhang, N.; Chen, H.; Wang, Y. Oncolytic Adenovirus H101 Synergizes with Radiation in Cervical Cancer Cells. Curr. Cancer Drug Targets 2021, 21, 619–630. [Google Scholar] [CrossRef]
  114. Xiao, T.; Fan, J.K.; Huang, H.L.; Gu, J.F.; Li, L.Y.; Liu, X.Y. VEGI-armed oncolytic adenovirus inhibits tumor neovascularization and directly induces mitochondria-mediated cancer cell apoptosis. Cell Res. 2010, 20, 367–378. [Google Scholar] [CrossRef]
  115. Garcia Paz, F.; Madrid Marina, V.; Morales Ortega, A.; Santander Gonzalez, A.; Peralta Zaragoza, O.; Burguete Garcia, A.; Torres Poveda, K.; Moreno, J.; Alcocer Gonzalez, J.; Hernandez Marquez, E.; et al. The relationship between the antitumor effect of the IL-12 gene therapy and the expression of Th1 cytokines in an HPV16-positive murine tumor model. Mediat. Inflamm. 2014, 2014, 510846. [Google Scholar] [CrossRef] [Green Version]
  116. Yiang, G.T.; Harn, H.J.; Yu, Y.L.; Hu, S.C.; Hung, Y.T.; Hsieh, C.J.; Lin, S.Z.; Wei, C.W. Immunotherapy: rAAV2 expressing interleukin-15 inhibits HeLa cell tumor growth in mice. J. Biomed. Sci. 2009, 16, 47. [Google Scholar] [CrossRef] [Green Version]
  117. Maitituoheti, M.; Li, Y.; Wang, W.; Wang, W.; Han, L.; Yang, R.; Wang, T.; Wu, Z.; Ma, D.; Wang, S. Adeno-associated virus-mediated local delivery of LIGHT suppresses tumorigenesis in a murine cervical cancer model. J. Immunother. 2011, 34, 581–587. [Google Scholar] [CrossRef]
  118. Matsushita, K.; Shimada, H.; Ueda, Y.; Inoue, M.; Hasegawa, M.; Tomonaga, T.; Matsubara, H.; Nomura, F. Non-transmissible Sendai virus vector encoding c-myc suppressor FBP-interacting repressor for cancer therapy. World J. Gastroenterol. 2014, 20, 4316–4328. [Google Scholar] [CrossRef]
  119. Ling, K.; Yang, L.; Yang, N.; Chen, M.; Wang, Y.; Liang, S.; Li, Y.; Jiang, L.; Yan, P.; Liang, Z. Gene Targeting of HPV18 E6 and E7 Synchronously by Nonviral Transfection of CRISPR/Cas9 System in Cervical Cancer. Hum. Gene Ther. 2020, 31, 297–308. [Google Scholar] [CrossRef]
  120. Zhen, S.; Liu, Y.; Lu, J.; Tuo, X.; Yang, X.; Chen, H.; Chen, W.; Li, X. Human Papillomavirus Oncogene Manipulation Using Clustered Regularly Interspersed Short Palindromic Repeats/Cas9 Delivered by pH-Sensitive Cationic Liposomes. Hum. Gene Ther. 2020, 31, 309–324. [Google Scholar] [CrossRef]
  121. Hu, Z.; Yu, L.; Zhu, D.; Ding, W.; Wang, X.; Zhang, C.; Wang, L.; Jiang, X.; Shen, H.; He, D.; et al. Disruption of HPV16-E7 by CRISPR/Cas system induces apoptosis and growth inhibition in HPV16 positive human cervical cancer cells. BioMed. Res. Int. 2014, 2014, 612823. [Google Scholar] [CrossRef]
  122. Hu, Z.; Ding, W.; Zhu, D.; Yu, L.; Jiang, X.; Wang, X.; Zhang, C.; Wang, L.; Ji, T.; Liu, D.; et al. TALEN-mediated targeting of HPV oncogenes ameliorates HPV-related cervical malignancy. J. Clin. Investig. 2015, 125, 425–436. [Google Scholar] [CrossRef] [Green Version]
  123. Xiong, J.; Tan, S.; Yu, L.; Shen, H.; Qu, S.; Zhang, C.; Ren, C.; Zhu, D.; Wang, H. E7-Targeted Nanotherapeutics for Key HPV Afflicted Cervical Lesions by Employing CRISPR/Cas9 and Poly (Beta-Amino Ester). Int. J. Nanomed. 2021, 16, 7609–7622. [Google Scholar] [CrossRef]
  124. Gao, C.; Wu, P.; Yu, L.; Liu, L.; Liu, H.; Tan, X.; Wang, L.; Huang, X.; Wang, H. The application of CRISPR/Cas9 system in cervical carcinogenesis. Cancer Gene Ther. 2021, 29, 466–474. [Google Scholar] [CrossRef]
  125. Lechanteur, A.; Furst, T.; Evrard, B.; Delvenne, P.; Piel, G.; Hubert, P. Promoting Vaginal Distribution of E7 and MCL-1 siRNA-Silencing Nanoparticles for Cervical Cancer Treatment. Mol. Pharm. 2017, 14, 1706–1717. [Google Scholar] [CrossRef]
  126. Han, H.; Chen, W.; Yang, J.; Liang, X.; Wang, Y.; Li, Q.; Yang, Y.; Li, K. Inhibition of cell proliferation and migration through nucleobase-modified polyamidoamine-mediated p53 delivery. Int. J. Nanomed. 2018, 13, 1297–1311. [Google Scholar] [CrossRef] [Green Version]
  127. Wang, P.; Yin, T.; Li, J.; Zheng, B.; Wang, X.; Wang, Y.; Zheng, J.; Zheng, R.; Shuai, X. Ultrasound-responsive microbubbles for sonography-guided siRNA delivery. Nanomedicine 2016, 12, 1139–1149. [Google Scholar] [CrossRef] [PubMed]
  128. Zhang, W.; Xing, L. RNAi gene therapy of SiHa cells via targeting human TERT induces growth inhibition and enhances radiosensitivity. Int. J. Oncol. 2013, 43, 1228–1234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Shi, Y.A.; Zhao, Q.; Zhang, L.H.; Du, W.; Wang, X.Y.; He, X.; Wu, S.; Li, Y.L. Knockdown of hTERT by siRNA inhibits cervical cancer cell growth in vitro and in vivo. Int. J. Oncol. 2014, 45, 1216–1224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Yang, Y.; He, L.; Liu, Y.; Xia, S.; Fang, A.; Xie, Y.; Gan, L.; He, Z.; Tan, X.; Jiang, C.; et al. Promising Nanocarriers for PEDF Gene Targeting Delivery to Cervical Cancer Cells Mediated by the Over-expressing FRalpha. Sci. Rep. 2016, 6, 32427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Cheng, H.Y.; Zhang, T.; Qu, Y.; Shi, W.J.; Lou, G.; Liu, Y.X.; Zhang, Y.Y.; Cheng, L. Synergism between RIZ1 gene therapy and paclitaxel in SiHa cervical cancer cells. Cancer Gene Ther. 2016, 23, 392–395. [Google Scholar] [CrossRef]
  132. Nowak, E.; Galilejczyk, A.; Sypniewski, D.; Bednarek, I. MMP-9 directed shRNAs as relevant inhibitors of matrix metalloproteinase 9 activity and signaling. Postepy. Hig. Med. Dosw. 2013, 67, 742–749. [Google Scholar] [CrossRef]
  133. Ying, T.H.; Lee, C.H.; Chiou, H.L.; Yang, S.F.; Lin, C.L.; Hung, C.H.; Tsai, J.P.; Hsieh, Y.H. Knockdown of Pentraxin 3 suppresses tumorigenicity and metastasis of human cervical cancer cells. Sci. Rep. 2016, 6, 29385. [Google Scholar] [CrossRef]
  134. Zhang, Y.; Xia, P.; Zhang, W.; Yan, M.; Xiong, X.; Yu, W.; Song, E. Short interfering RNA targeting Net1 reduces the angiogenesis and tumor growth of in vivo cervical squamous cell carcinoma through VEGF down-regulation. Hum. Pathol. 2017, 65, 113–122. [Google Scholar] [CrossRef]
  135. Wang, L.L.; Chen, P.; Luo, S.; Li, J.; Liu, K.; Hu, H.Z.; Wei, Y.Q. CXC-chemokine-ligand-10 gene therapy efficiently inhibits the growth of cervical carcinoma on the basis of its anti-angiogenic and antiviral activity. Biotechnol. Appl. Biochem. 2009, 53, 209–216. [Google Scholar] [CrossRef]
  136. Yang, L.L.; Wang, B.Q.; Chen, L.L.; Luo, H.Q.; Wu, J.B. CXCL10 enhances radiotherapy effects in HeLa cells through cell cycle redistribution. Oncol. Lett. 2012, 3, 383–386. [Google Scholar] [CrossRef] [Green Version]
  137. Sun, Y.; Peng, S.; Qiu, J.; Miao, J.; Yang, B.; Jeang, J.; Hung, C.F.; Wu, T.C. Intravaginal HPV DNA vaccination with electroporation induces local CD8+ T-cell immune responses and antitumor effects against cervicovaginal tumors. Gene Ther. 2015, 22, 528–535. [Google Scholar] [CrossRef] [Green Version]
  138. Sun, Y.; Peng, S.; Yang, A.; Farmer, E.; Wu, T.C.; Hung, C.F. Coinjection of IL2 DNA enhances E7-specific antitumor immunity elicited by intravaginal therapeutic HPV DNA vaccination with electroporation. Gene Ther. 2017, 24, 408–415. [Google Scholar] [CrossRef] [Green Version]
  139. Lin, G.; Chen, Q.; Yu, S.; Lin, S.; Yao, H.; Ding, Z.; Chen, S.; Lin, M.C.; Wang, X. Overexpression of human telomerase reverse transcriptase C-terminal polypeptide sensitizes HeLa cells to 5-fluorouracilinduced growth inhibition and apoptosis. Mol. Med. Rep. 2014, 9, 279–284. [Google Scholar] [CrossRef] [Green Version]
  140. Liu, Y.; Li, H.; Zhang, R.; Dang, H.; Sun, P.; Zou, L.; Zhang, Y.; Gao, Y.; Hu, Y. Overexpression of the BRIP1 ameliorates chemosensitivity to cisplatin by inhibiting Rac1 GTPase activity in cervical carcinoma HeLa cells. Gene 2016, 578, 85–91. [Google Scholar] [CrossRef]
  141. Li, Y.; Zhang, M.C.; Xu, X.K.; Zhao, Y.; Mahanand, C.; Zhu, T.; Deng, H.; Nevo, E.; Du, J.Z.; Chen, X.Q. Functional Diversity of p53 in Human and Wild Animals. Front. Endocrinol. 2019, 10, 152. [Google Scholar] [CrossRef]
  142. Doorbar, J. Molecular biology of human papillomavirus infection and cervical cancer. Clin. Sci. 2006, 110, 525–541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Jia, H.; Kling, J. China offers alternative gateway for experimental drugs. Nat. Biotechnol. 2006, 24, 117–118. [Google Scholar] [CrossRef] [PubMed]
  144. Liu, B.; Han, S.M.; Tang, X.Y.; Han, L.; Li, C.Z. Cervical cancer gene therapy by gene loaded PEG-PLA nanomedicine. Asian. Pac. J. Cancer Prev. 2014, 15, 4915–4918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Wu, M.; Gunning, W.; Ratnam, M. Expression of folate receptor type alpha in relation to cell type, malignancy, and differentiation in ovary, uterus, and cervix. Cancer Epidemiol. Biomarkers Prev. 1999, 8, 775–782. [Google Scholar] [PubMed]
  146. Yang, S.; Xing, L.; Gu, L.; Cheng, H.; Feng, Y.; Zhang, Y. Combination of RIZ1 Overexpression and Radiotherapy Contributes to Apoptosis and DNA Damage of HeLa and SiHa Cervical Cancer Cells. Basic Clin. Pharmacol. Toxicol. 2018, 123, 137–146. [Google Scholar] [CrossRef]
  147. Banno, K.; Iida, M.; Yanokura, M.; Kisu, I.; Iwata, T.; Tominaga, E.; Tanaka, K.; Aoki, D. MicroRNA in cervical cancer: OncomiRs and tumor suppressor miRs in diagnosis and treatment. Sci. World J. 2014, 2014, 178075. [Google Scholar] [CrossRef]
  148. Tornesello, M.L.; Faraonio, R.; Buonaguro, L.; Annunziata, C.; Starita, N.; Cerasuolo, A.; Pezzuto, F.; Tornesello, A.L.; Buonaguro, F.M. The Role of microRNAs, Long Non-coding RNAs, and Circular RNAs in Cervical Cancer. Front Oncol. 2020, 10, 150. [Google Scholar] [CrossRef] [Green Version]
  149. Long, M.J.; Wu, F.X.; Li, P.; Liu, M.; Li, X.; Tang, H. MicroRNA-10a targets CHL1 and promotes cell growth, migration and invasion in human cervical cancer cells. Cancer Lett. 2012, 324, 186–196. [Google Scholar] [CrossRef]
  150. Wang, Y.; Wang, Y.; Zhong, W.; Gulina, K. Correlation between miR-19a inhibition and radiosensitivity in SiHa cervical cancer cells. J. BUON 2017, 22, 1505–1508. [Google Scholar]
  151. Kang, H.W.; Wang, F.; Wei, Q.; Zhao, Y.F.; Liu, M.; Li, X.; Tang, H. miR-20a promotes migration and invasion by regulating TNKS2 in human cervical cancer cells. FEBS Lett. 2012, 586, 897–904. [Google Scholar] [CrossRef] [Green Version]
  152. Yao, Q.; Xu, H.; Zhang, Q.Q.; Zhou, H.; Qu, L.H. MicroRNA-21 promotes cell proliferation and down-regulates the expression of programmed cell death 4 (PDCD4) in HeLa cervical carcinoma cells. Biochem. Biophys. Res. Commun. 2009, 388, 539–542. [Google Scholar] [CrossRef]
  153. Peralta-Zaragoza, O.; Deas, J.; Meneses-Acosta, A.; De la O-Gómez, F.; Fernandez-Tilapa, G.; Gomez-Ceron, C.; Benitez-Boijseauneau, O.; Burguete-Garcia, A.; Torres-Poveda, K.; Bermudez-Morales, V.H.; et al. Relevance of miR-21 in regulation of tumor suppressor gene PTEN in human cervical cancer cells. BMC Cancer 2016, 16, 215. [Google Scholar] [CrossRef] [Green Version]
  154. Qin, W.; Dong, P.; Ma, C.; Mitchelson, K.; Deng, T.; Zhang, L.; Sun, Y.; Feng, X.; Ding, Y.; Lu, X.; et al. MicroRNA-133b is a key promoter of cervical carcinoma development through the activation of the ERK and AKT1 pathways. Oncogene 2012, 31, 4067–4075. [Google Scholar] [CrossRef] [Green Version]
  155. Li, J.H.; Xiao, X.; Zhang, Y.N.; Wang, Y.M.; Feng, L.M.; Wu, Y.M.; Zhang, Y.X. MicroRNA miR-886-5p inhibits apoptosis by down-regulating Bax expression in human cervical carcinoma cells. Gynecol. Oncol. 2011, 120, 145–151. [Google Scholar] [CrossRef]
  156. Liu, L.; Yu, X.; Guo, X.; Tian, Z.; Su, M.; Long, Y.; Huang, C.; Zhou, F.; Liu, M.; Wu, X.; et al. miR-143 is downregulated in cervical cancer and promotes apoptosis and inhibits tumor formation by targeting Bcl-2. Mol. Med. Rep. 2012, 5, 753–760. [Google Scholar] [CrossRef]
  157. He, C.Y.; Yang, J. miR-187 induces apoptosis of SiHa cervical carcinoma cells by downregulating Bcl-2. Genet. Mol. Res. 2017, 16, gmr16018969. [Google Scholar] [CrossRef]
  158. Mou, Z.; Xu, X.; Dong, M.; Xu, J. MicroRNA-148b Acts as a Tumor Suppressor in Cervical Cancer by Inducing G1/S-Phase Cell Cycle Arrest and Apoptosis in a Caspase-3-Dependent Manner. Med. Sci. Monit. 2016, 22, 2809–2815. [Google Scholar] [CrossRef] [Green Version]
  159. Liu, Y.L.; Wang, G.Q.; Cui, H.X.; Li, X.X.; Xu, Z.L.; Wang, X.Y. miRNA211 induces apoptosis of cervical cancer SiHa cells via down-regulation of inhibitor of apoptosis proteins. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 336–342. [Google Scholar] [CrossRef]
  160. Yao, R.; Zheng, H.; Wu, L.; Cai, P. miRNA-641 inhibits the proliferation, migration, and invasion and induces apoptosis of cervical cancer cells by directly targeting ZEB1. Onco Targets Ther. 2018, 11, 8965–8976. [Google Scholar] [CrossRef] [Green Version]
  161. Yang, Y.; Liu, H.; Wang, X.; Chen, L. Up-regulation of microRNA-664 inhibits cell growth and increases cisplatin sensitivity in cervical cancer. Int. J. Clin. Exp. Med. 2015, 8, 18123–18129. [Google Scholar]
  162. Guo, M.; Zhao, X.; Yuan, X.; Jiang, J.; Li, P. MiR-let-7a inhibits cell proliferation, migration, and invasion by down-regulating PKM2 in cervical cancer. Oncotarget 2017, 8, 28226–28236. [Google Scholar] [CrossRef] [Green Version]
  163. Zhang, J.; Li, S.; Yan, Q.; Chen, X.; Yang, Y.; Liu, X.; Wan, X. Interferon-beta induced microRNA-129-5p down-regulates HPV-18 E6 and E7 viral gene expression by targeting SP1 in cervical cancer cells. PLoS ONE 2013, 8, e81366. [Google Scholar] [CrossRef]
  164. Boyd, J. Molecular biology in the clinicopathologic assessment of endometrial carcinoma subtypes. Gynecol. Oncol. 1996, 61, 163–165. [Google Scholar] [CrossRef]
  165. Ramondetta, L.; Mills, G.B.; Burke, T.W.; Wolf, J.K. Adenovirus-mediated expression of p53 or p21 in a papillary serous endometrial carcinoma cell line (SPEC-2) results in both growth inhibition and apoptotic cell death: Potential application of gene therapy to endometrial cancer. Clin. Cancer Res. 2000, 6, 278–284. [Google Scholar]
  166. Ural, A.U.; Takebe, N.; Adhikari, D.; Ercikan-Abali, E.; Banerjee, D.; Barakat, R.; Bertino, J.R. Gene therapy for endometrial carcinoma with the herpes simplex thymidine kinase gene. Gynecol. Oncol. 2000, 76, 305–310. [Google Scholar] [CrossRef]
  167. Grundker, C.; Huschmand Nia, A.; Emons, G. Gonadotropin-releasing hormone receptor-targeted gene therapy of gynecologic cancers. Mol. Cancer Ther. 2005, 4, 225–231. [Google Scholar] [CrossRef]
  168. Xia, Y.; Li, X.; Sun, W. Applications of Recombinant Adenovirus-p53 Gene Therapy for Cancers in the Clinic in China. Curr. Gene Ther. 2020, 20, 127–141. [Google Scholar] [CrossRef]
  169. Maxwell, G.L.; Chandramouli, G.V.; Dainty, L.; Litzi, T.J.; Berchuck, A.; Barrett, J.C.; Risinger, J.I. Microarray analysis of endometrial carcinomas and mixed mullerian tumors reveals distinct gene expression profiles associated with different histologic types of uterine cancer. Clin. Cancer Res. 2005, 11, 4056–4066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Maurice-Duelli, A.; Ndoye, A.; Bouali, S.; Leroux, A.; Merlin, J.L. Enhanced cell growth inhibition following PTEN nonviral gene transfer using polyethylenimine and photochemical internalization in endometrial cancer cells. Technol. Cancer Res. Treat. 2004, 3, 459–465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Banno, K.; Yanokura, M.; Kisu, I.; Yamagami, W.; Susumu, N.; Aoki, D. MicroRNAs in endometrial cancer. Int. J. Clin. Oncol. 2013, 18, 186–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Donkers, H.; Bekkers, R.; Galaal, K. Diagnostic value of microRNA panel in endometrial cancer: A systematic review. Oncotarget 2020, 11, 2010–2023. [Google Scholar] [CrossRef]
  173. Evans, P.; Brunsell, S. Uterine fibroid tumors: Diagnosis and treatment. Am. Fam. Physician 2007, 75, 1503–1508. [Google Scholar]
  174. Hassan, M.H.; Khatoon, N.; Curiel, D.T.; Hamada, F.M.; Arafa, H.M.; Al-Hendy, A. Toward gene therapy of uterine fibroids: Targeting modified adenovirus to human leiomyoma cells. Hum. Reprod. 2008, 23, 514–524. [Google Scholar] [CrossRef] [Green Version]
  175. Niu, H.; Simari, R.D.; Zimmermann, E.M.; Christman, G.M. Nonviral vector-mediated thymidine kinase gene transfer and ganciclovir treatment in leiomyoma cells. Obstet. Gynecol. 1998, 91, 735–740. [Google Scholar] [CrossRef]
  176. Al-Hendy, A.; Lee, E.J.; Wang, H.Q.; Copland, J.A. Gene therapy of uterine leiomyomas: Adenovirus-mediated expression of dominant negative estrogen receptor inhibits tumor growth in nude mice. Am. J. Obstet. Gynecol. 2004, 191, 1621–1631. [Google Scholar] [CrossRef]
  177. Nair, S.; Curiel, D.T.; Rajaratnam, V.; Thota, C.; Al-Hendy, A. Targeting adenoviral vectors for enhanced gene therapy of uterine leiomyomas. Hum. Reprod. 2013, 28, 2398–2406. [Google Scholar] [CrossRef] [Green Version]
  178. Abdelaziz, M.; Sherif, L.; ElKhiary, M.; Nair, S.; Shalaby, S.; Mohamed, S.; Eziba, N.; El-Lakany, M.; Curiel, D.; Ismail, N.; et al. Targeted Adenoviral Vector Demonstrates Enhanced Efficacy for In Vivo Gene Therapy of Uterine Leiomyoma. Reprod. Sci. 2016, 23, 464–474. [Google Scholar] [CrossRef] [Green Version]
  179. Deng, Y.; Han, Q.; Mei, S.; Li, H.; Yang, F.; Wang, J.; Ge, S.; Jing, X.; Xu, H.; Zhang, T. Cyclin-dependent kinase subunit 2 overexpression promotes tumor progression and predicts poor prognosis in uterine leiomyosarcoma. Oncol. Lett. 2019, 18, 2845–2852. [Google Scholar] [CrossRef] [Green Version]
  180. Li, Y.; Qiang, W.; Griffin, B.B.; Gao, T.; Chakravarti, D.; Bulun, S.; Kim, J.J.; Wei, J.J. HMGA2-mediated tumorigenesis through angiogenesis in leiomyoma. Fertil. Steril. 2020, 114, 1085–1096. [Google Scholar] [CrossRef]
  181. Dabrosin, C.; Gyorffy, S.; Margetts, P.; Ross, C.; Gauldie, J. Therapeutic effect of angiostatin gene transfer in a murine model of endometriosis. Am. J. Pathol. 2002, 161, 909–918. [Google Scholar] [CrossRef] [Green Version]
  182. Sun, J.J.; Yin, L.R.; Mi, R.R.; Ma, H.D.; Guo, S.J.; Shi, Y.; Gu, Y.J. Human endostatin antiangiogenic gene therapy mediated by recombinant adeno-associated virus vector in nude mouse with endometriosis. Zhonghua Fu Chan Ke Za Zhi 2010, 45, 45–50. [Google Scholar]
  183. Othman, E.E.; Salama, S.; Ismail, N.; Al-Hendy, A. Toward gene therapy of endometriosis: Adenovirus-mediated delivery of dominant negative estrogen receptor genes inhibits cell proliferation, reduces cytokine production, and induces apoptosis of endometriotic cells. Fertil. Steril. 2007, 88, 462–471. [Google Scholar] [CrossRef] [PubMed]
  184. Othman, E.E.; Zhu, Z.B.; Curiel, D.T.; Khatoon, N.; Salem, H.T.; Khalifa, E.A.; Al-Hendy, A. Toward gene therapy of endometriosis: Transductional and transcriptional targeting of adenoviral vectors to endometriosis cells. Am. J. Obstet. Gynecol. 2008, 199, 117.e1–117.e6. [Google Scholar] [CrossRef] [Green Version]
  185. Wang, N.; Liu, B.; Liang, L.; Wu, Y.; Xie, H.; Huang, J.; Guo, X.; Tan, J.; Zhan, X.; Liu, Y.; et al. Antiangiogenesis therapy of endometriosis using PAMAM as a gene vector in a noninvasive animal model. BioMed Res. Int. 2014, 2014, 546479. [Google Scholar] [CrossRef]
  186. Koippallil Gopalakrishnan, A.R.; Pandit, H.; Metkari, S.M.; Warty, N.; Madan, T. Adenoviral vector encoding soluble Flt-1 engineered human endometrial mesenchymal stem cells effectively regress endometriotic lesions in NOD/SCID mice. Gene Ther. 2016, 23, 580–591. [Google Scholar] [CrossRef]
  187. Fortin, M.; Lepine, M.; Merlen, Y.; Thibeault, I.; Rancourt, C.; Gosselin, D.; Hugo, P.; Steff, A.M. Quantitative assessment of human endometriotic tissue maintenance and regression in a noninvasive mouse model of endometriosis. Mol. Ther. 2004, 9, 540–547. [Google Scholar] [CrossRef]
  188. Goncalves, G.A.; Camargo-Kosugi, C.M.; Bonetti, T.C.; Invitti, A.L.; Girao, M.J.; Silva, I.D.; Schor, E. p27kip1 overexpression regulates VEGF expression, cell proliferation and apoptosis in cell culture from eutopic endometrium of women with endometriosis. Apoptosis 2015, 20, 327–335. [Google Scholar] [CrossRef]
  189. Goncalves, G.A.; Invitti, A.L.; Parreira, R.M.; Kopelman, A.; Schor, E.; Girao, M.J. p27(kip1) overexpression regulates IL-1beta in the microenvironment of stem cells and eutopic endometriosis co-cultures. Cytokine 2017, 89, 229–234. [Google Scholar] [CrossRef]
  190. Paupoo, A.A.; Zhu, Z.B.; Wang, M.; Rein, D.T.; Starzinski-Powitz, A.; Curiel, D.T. A conditionally replicative adenovirus, CRAd-S-pK7, can target endometriosis with a cell-killing effect. Hum. Reprod. 2010, 25, 2068–2083. [Google Scholar] [CrossRef] [Green Version]
  191. Xing, A.; Boileau, P.; Cauzac, M.; Challier, J.C.; Girard, J.; Hauguel-de Mouzon, S. Comparative in vivo approaches for selective adenovirus-mediated gene delivery to the placenta. Hum. Gene Ther. 2000, 11, 167–177. [Google Scholar] [CrossRef]
  192. Heikkila, A.; Hiltunen, M.O.; Turunen, M.P.; Keski-Nisula, L.; Turunen, A.M.; Rasanen, H.; Rissanen, T.T.; Kosma, V.M.; Manninen, H.; Heinonen, S.; et al. Angiographically guided utero-placental gene transfer in rabbits with adenoviruses, plasmid/liposomes and plasmid/polyethyleneimine complexes. Gene Ther. 2001, 8, 784–788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Miller, A.G.; Aplin, J.D.; Westwood, M. Adenovirally mediated expression of insulin-like growth factors enhances the function of first trimester placental fibroblasts. J. Clin. Endocrinol. Metab. 2005, 90, 379–385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Keswani, S.G.; Balaji, S.; Katz, A.B.; King, A.; Omar, K.; Habli, M.; Klanke, C.; Crombleholme, T.M. Intraplacental gene therapy with Ad-IGF-1 corrects naturally occurring rabbit model of intrauterine growth restriction. Hum. Gene Ther. 2015, 26, 172–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Carr, D.J.; Wallace, J.M.; Aitken, R.P.; Milne, J.S.; Mehta, V.; Martin, J.F.; Zachary, I.C.; Peebles, D.M.; David, A.L. Uteroplacental adenovirus vascular endothelial growth factor gene therapy increases fetal growth velocity in growth-restricted sheep pregnancies. Hum. Gene Ther. 2014, 25, 375–384. [Google Scholar] [CrossRef]
  196. Bagot, C.N.; Troy, P.J.; Taylor, H.S. Alteration of maternal Hoxa10 expression by in vivo gene transfection affects implantation. Gene Ther. 2000, 7, 1378–1384. [Google Scholar] [CrossRef] [Green Version]
  197. Nakamura, H.; Kimura, T.; Ogita, K.; Koyama, S.; Tsujie, T.; Tsutsui, T.; Shimoya, K.; Koyama, M.; Kaneda, Y.; Murata, Y. Alteration of the timing of implantation by in vivo gene transfer: Delay of implantation by suppression of nuclear factor kappaB activity and partial rescue by leukemia inhibitory factor. Biochem. Biophys. Res. Commun. 2004, 321, 886–892. [Google Scholar] [CrossRef] [PubMed]
  198. Giudice, L.C.; Kao, L.C. Endometriosis. Lancet 2004, 364, 1789–1799. [Google Scholar] [CrossRef]
  199. Shubina, A.N.; Egorova, A.A.; Baranov, V.S.; Kiselev, A.V. Recent advances in gene therapy of endometriosis. Recent Pat. DNA Gene Seq. 2013, 7, 169–178. [Google Scholar] [CrossRef]
  200. Schor, E.; da Silva, I.D.; Sato, H.; Baracat, E.C.; Girao, M.J.; de Freitas, V. P27Kip1 is down-regulated in the endometrium of women with endometriosis. Fertil. Steril. 2009, 91, 682–686. [Google Scholar] [CrossRef]
  201. Senut, M.C.; Suhr, S.T.; Gage, F.H. Gene transfer to the rodent placenta in situ. A new strategy for delivering gene products to the fetus. J. Clin. Investig. 1998, 101, 1565–1571. [Google Scholar] [CrossRef] [Green Version]
  202. Taylor, H.S.; Arici, A.; Olive, D.; Igarashi, P. HOXA10 is expressed in response to sex steroids at the time of implantation in the human endometrium. J. Clin. Investig. 1998, 101, 1379–1384. [Google Scholar] [CrossRef] [Green Version]
  203. Horcajadas, J.A.; Riesewijk, A.; Martin, J.; Cervero, A.; Mosselman, S.; Pellicer, A.; Simon, C. Global gene expression profiling of human endometrial receptivity. J. Reprod. Immunol. 2004, 63, 41–49. [Google Scholar] [CrossRef]
  204. Hernandez-Vargas, P.; Munoz, M.; Dominguez, F. Identifying biomarkers for predicting successful embryo implantation: Applying single to multi-OMICs to improve reproductive outcomes. Hum. Reprod. Update 2020, 26, 264–301. [Google Scholar] [CrossRef]
  205. Kim, K.H.; Dmitriev, I.; O’Malley, J.P.; Wang, M.; Saddekni, S.; You, Z.; Preuss, M.A.; Harris, R.D.; Aurigemma, R.; Siegal, G.P.; et al. A phase I clinical trial of Ad5.SSTR/TK.RGD, a novel infectivity-enhanced bicistronic adenovirus, in patients with recurrent gynecologic cancer. Clin. Cancer Res. 2012, 18, 3440–3451. [Google Scholar] [CrossRef] [Green Version]
  206. Sterman, D.H.; Recio, A.; Haas, A.R.; Vachani, A.; Katz, S.I.; Gillespie, C.T.; Cheng, G.; Sun, J.; Moon, E.; Pereira, L.; et al. A phase I trial of repeated intrapleural adenoviral-mediated interferon-beta gene transfer for mesothelioma and metastatic pleural effusions. Mol. Ther. 2010, 18, 852–860. [Google Scholar] [CrossRef]
  207. Pinte, L.; Cunningham, A.; Trebeden-Negre, H.; Nikiforow, S.; Ritz, J. Global Perspective on the Development of Genetically Modified Immune Cells for Cancer Therapy. Front. Immunol. 2020, 11, 608485. [Google Scholar] [CrossRef]
  208. Maecker, B.; Sherr, D.H.; Vonderheide, R.H.; von Bergwelt-Baildon, M.S.; Hirano, N.; Anderson, K.S.; Xia, Z.; Butler, M.O.; Wucherpfennig, K.W.; O’Hara, C.; et al. The shared tumor-associated antigen cytochrome P450 1B1 is recognized by specific cytotoxic T cells. Blood 2003, 102, 3287–3294. [Google Scholar] [CrossRef] [Green Version]
  209. Gribben, J.G.; Ryan, D.P.; Boyajian, R.; Urban, R.G.; Hedley, M.L.; Beach, K.; Nealon, P.; Matulonis, U.; Campos, S.; Gilligan, T.D.; et al. Unexpected association between induction of immunity to the universal tumor antigen CYP1B1 and response to next therapy. Clin. Cancer Res. 2005, 11, 4430–4436. [Google Scholar] [CrossRef] [Green Version]
  210. Liu, Y.; Wu, L.; Tong, R.; Yang, F.; Yin, L.; Li, M.; You, L.; Xue, J.; Lu, Y. PD-1/PD-L1 Inhibitors in Cervical Cancer. Front. Pharmacol. 2019, 10, 65. [Google Scholar] [CrossRef] [Green Version]
  211. Luo, F.; Wen, Y.; Zhou, H.; Li, Z. Roles of long non-coding RNAs in cervical cancer. Life Sci. 2020, 256, 117981. [Google Scholar] [CrossRef]
  212. Shi, Y.; He, R.; Yang, Y.; He, Y.; Shao, K.; Zhan, L.; Wei, B. Circular RNAs: Novel biomarkers for cervical, ovarian and endometrial cancer (Review). Oncol. Rep. 2020, 44, 1787–1798. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Viral and non-viral systems for gene delivery.
Figure 1. Viral and non-viral systems for gene delivery.
Cancers 14 03238 g001
Figure 2. Main targeted approaches for the gene therapy of gynaecological malignant and benign disorders. They mainly involve (A) Mutation compensation employing primarily adenoviral vectors to correct the mutated phenotype or modify aberrant gene expression in tumour cells, usually leading to cancer cell death (B) Suicide gene therapy, usually with the delivery of a suicide gene, such as HSV-TK, leading to cell death following the production of toxic metabolite (C) Antiangiogenic gene therapy, by transferring an antiangiogenic gene, which inhibits cancer cell angiogenesis resulting in cancer cell death (D) Oncolytic virotherapy using oncolytic viruses that enter the cell, replicate and lead to oncolysis and cancer cell death. (E) Immunopotentiation by delivering a potent anti-cancer cytokine gene within the tumour using primarily adenoviruses and adeno-associated viral vectors, leading to reduction in tumour growth.
Figure 2. Main targeted approaches for the gene therapy of gynaecological malignant and benign disorders. They mainly involve (A) Mutation compensation employing primarily adenoviral vectors to correct the mutated phenotype or modify aberrant gene expression in tumour cells, usually leading to cancer cell death (B) Suicide gene therapy, usually with the delivery of a suicide gene, such as HSV-TK, leading to cell death following the production of toxic metabolite (C) Antiangiogenic gene therapy, by transferring an antiangiogenic gene, which inhibits cancer cell angiogenesis resulting in cancer cell death (D) Oncolytic virotherapy using oncolytic viruses that enter the cell, replicate and lead to oncolysis and cancer cell death. (E) Immunopotentiation by delivering a potent anti-cancer cytokine gene within the tumour using primarily adenoviruses and adeno-associated viral vectors, leading to reduction in tumour growth.
Cancers 14 03238 g002
Figure 3. Gene therapy strategies for ovarian cancer. These employ viral vectors, such as Herpes Simplex Virus, adenoviral and adeno-associated viral vectors, lentiviral vectors and Measles virus, and non-viral systems, such as shRNA/siRNA/miRNA, DNA plasmids, lipid-based and polymeric NPs and CRISPR/Cas9 approaches. Of note, some of the above approaches, e.g., shRNA, were also delivered via lentiviral vectors.
Figure 3. Gene therapy strategies for ovarian cancer. These employ viral vectors, such as Herpes Simplex Virus, adenoviral and adeno-associated viral vectors, lentiviral vectors and Measles virus, and non-viral systems, such as shRNA/siRNA/miRNA, DNA plasmids, lipid-based and polymeric NPs and CRISPR/Cas9 approaches. Of note, some of the above approaches, e.g., shRNA, were also delivered via lentiviral vectors.
Cancers 14 03238 g003
Figure 5. Gene therapy approaches toward benign gynaecological disorders. These include uterine fibroids, endometriosis, placental and embryo implantation disorders, using viral and non-viral systems. Successful approaches against uterine fibroids treatment employed primarily adenoviral vectors and plasmids, for endometriosis adenoviral vectors, adeno-associated vectors, and dendrimers, for placental disorders adenoviral vectors and plasmid/liposome complexes, while for embryo implantation disorders HVJ vectors and plasmid/liposomes complexes.
Figure 5. Gene therapy approaches toward benign gynaecological disorders. These include uterine fibroids, endometriosis, placental and embryo implantation disorders, using viral and non-viral systems. Successful approaches against uterine fibroids treatment employed primarily adenoviral vectors and plasmids, for endometriosis adenoviral vectors, adeno-associated vectors, and dendrimers, for placental disorders adenoviral vectors and plasmid/liposome complexes, while for embryo implantation disorders HVJ vectors and plasmid/liposomes complexes.
Cancers 14 03238 g005
Table 3. Targeted gene therapy approaches for benign gynaecological abnormalities.
Table 3. Targeted gene therapy approaches for benign gynaecological abnormalities.
Gene Therapy StrategyGeneDelivery SystemOutcomeModel/SpeciesReferences
Uterine leiomyomas or fibroids
Suicide gene
therapy
HSV-TKNon-viral
approach
Significant cell death of
leiomyoma cells
in vitro
(human/rat)
[175]
Suicide gene
therapy
HSV-TKAdenoviral
vector
Ad-HSV1TK/GCV
Significant reduction in uterine
fibroids volume, increased apoptosis and inhibition of cell proliferation
in Eker rats
in vitro (human)
in vivo (rat)
[174]
Suicide gene
therapy
Somatostatin receptor subtype 2/
HSV-TK
Adenoviral
vector
Ad-SSTR-RGD-TK
Significant reduction in leiomyomas size and proliferation, induction of apoptosis and inhibition of
angiogenic- and extracellular
matrix-related genes
in vitro (human)
in vivo (mouse)
[177,178]
Suicide gene
therapy/
Antiangiogenic gene
therapy
DNER/
HSV-TK
Adenoviral
vector
Ad-DNER
Leiomyoma-selective expression,
decreased apoptosis, and proliferation
in vitro
(human/rat)
[176]
Mutation
compensation
CKS2Plasmid
si-CKS2
Cell cycle arrest and inhibition of cell proliferation, colony formation,
migration, and invasion
in vitro (human)[179]
Endometriosis
Antiangiogenic strategyAngiostatinAdenoviral vectorEradication of
endometriosis lesions
in vitro (mouse)[181]
Antiangiogenic strategyCOL18A1
(endostatin)
Adeno-associated
vector
rAAV-endostatin-EGFP
Inhibition of angiogenesis in
endometriotic lesions
in vitro (human)
in vivo (mouse)
[182]
Antiangiogenic strategyDNERAdenoviral vector
Ad-DNER
Ad5 vector
Significant cell death and reduction in pro-inflammatory and angiogenic
cytokine production by
endometriosis cells
in vitro
(human)
[183,184]
Antiangiogenic strategyCOL18A1
(endostatin)
Endostatin
plasmid/PAMAM dendrimers
Inhibition of endometriosis
development
in vitro (human)
in vivo (mice)
[185]
Antiangiogenic strategyVEGF receptor 1Adenoviral vector
Ad-sft-1
Transduction flexibilityin vitro (human)
in vivo (mouse)
[186]
Antiangiogenic strategyHPSE
(heparanase)
SLP1
Adenoviral vector
Ad-heparanase-luc
Ad-SLP1-luc
Ad-heparanase-luc vector
demonstrated an “Endometriosis ON, liver OFF” phenotype
in vitro (human)[184]
Suicide gene
therapy
HSV-TKAdenoviral vector
Ad-TK
Induction of significant cell death
and reduction in endometrial
lesions in vivo
in vitro (human)
in vivo (mouse)
[187]
Mutation
compensation
p27kip1Adenoviral vector
Ad-p27
Cell cycle arrest and
reduced proliferation
in vitro (human)[188,189]
Mutation
compensation
VEGF promoterAdenoviral vector
CRAd-S-pk7
Endometriosis killing effectin vitro (human)[190]
Placental disorders
Mutation
compensation
Adenoviral vectorSuccessful transfection
of placenta cells
in vitro (human)
in vivo
(mouse/rabbit)
[191,192,193]
Mutation
compensation
IGF-1Adenoviral vector
Ad-IGF-1
Restoration of birth weightin vivo (rabbit)[194]
Mutation
compensation
VEGFAdenoviral vector
Ad-VEGF
Improvement of fetal growth,
increased fetal growth velocity
in vivo (sheep)[195]
Mutation
compensation
IGF-1/IGF-IIPlasmid/liposomesImportance of overexpression
or inhibition on placental cell proliferation,
migration and survival
in vitro (human)[192]
Embryo implantation disorders
Mutation
compensation
Hoxa10Plasmid/liposomeSignificant increase in litter sizein vivo (mouse)[196]
Mutation
compensation
NF-κBHVJ vectorNF-κB expression determines
the timing of implantation, via
control of LIF expression
in vitro
(pigs)
[197]
Table 4. Gene therapy clinical trials for ovarian, cervical and endometrial cancer available online clinicaltrials.gov (accessed on 29 May 2022) [101].
Table 4. Gene therapy clinical trials for ovarian, cervical and endometrial cancer available online clinicaltrials.gov (accessed on 29 May 2022) [101].
Therapeutic StrategyPhaseReferenceInterventionDate
(First–Last Posted)
Recruiting Status
Ovarian cancer gene therapy clinical trials
Mutation compensationPhase INCT00003450Delivery of p53 gene using
Ad5CMV-p53
5 December 2003–
2 February 2021
Completed
Mutation compensationPhase INCT00003588Delivery of p53 gene using Ad-5326 August 2004–
8 February 2013
Completed
Mutation compensationPhase IINCT02435186Delivery of p53 gene using Ad-53
+ cisplatin and PTX
6 May 2015–
6 May 2015
Unknown
Oncolytic virotherapyPhase I/IINCT02068794Administration of oncolytic
Measles virus-infected MSCs
21 February 2014–
15 March 2022
Recruiting
ImmunopotentiationPhase INCT00019136Transfer of anti-CD3 stimulated
peripheral blood lymphocytes
transduced with a folate binding
protein chimeric TCR
(MoV-gamma chimeric TCR)
27 January 2003–
29 April 2015
Completed
ImmunopotentiationPhase INCT00004178Transfer of recombinant CEA
immunoglobulin TCR (IgTCR)
19 April 2004–
10 June 2011
Completed
ImmunopotentiationPhase I/IINCT01583686Transduction of PBLs with retroviral vector and transfer of
anti-mesothelin CAR TCR
24 April 2012–
14 October 2019
Terminated
ImmunopotentiationPhase INCT00066404Delivery of human IFN-β gene using
recombinant adenovirus Ad.IFN-β
7 August 2003–
13 May 2020
Completed
ImmunopotentiationPhase IINCT03412877Infusion of autologous T cells
engineered to express
neoantigens-reactive TCRs
29 January 2018–
27 May 2022
Recruiting
ImmunopotentiationPhase I/IINCT05194735Infusion of autologous T cells
engineered to express neoantigens-reactive TCRs + IL-2
18 January 2022–
11 April 2022
Recruiting
ImmunopotentiationPhase IINCT04102436Infusion of autologous T cells
engineered with Sleeping Beauty
transposon/transposase system (non-viral approach) to express
neoantigens-reactive TCRs
25 September 2019–
27 May 2022
Recruiting
ImmunopotentiationPhase INCT00381173Administration of ZYC300 using plasmid DNA + cyclophosphamide27 September 2006–
14 May 2013
Completed
ImmunopotentiationPhase 1a/1bNCT03970382Infusion of autologous T cells
engineered to express neoantigen-
reactive TCR + nivolumab
31 May 2019–
8 February 2022
Active, not recruiting
ImmunopotentiationPhase INCT02366546Administration of TB1-1301
(NY-ESO-1 specific TCR) in solid tumours cyclophosphamide + fludarabine
19 February 2015–
24 October 2018
Active, not recruiting
ImmunopotentiationPhase INCT02096614Administration of TB1-1201 (MAGE-A4-specific TCR) in solid tumours + cyclophosphamide + fludarabine26 March 2014–
18 June 2021
Completed
Suicide gene therapyPhase INCT00964756Therapeutic gene delivery using Ad5.SSTR/TK.RGD + GCV25 August 2009–
13 February 2013
Completed
Suicide gene therapyPhase INCT01997190Administration of AdV-tk
+ valacyclovir
28 November 2013–
4 December 2020
Completed
Suicide gene therapyPhase IINCT00005025Administration of HSV-tK + GCV5 May 2003–
6 November 2013
Unknown
Cervical cancer gene therapy clinical trials
Mutation compensationPhase IINCT03544723Delivery of p53 gene using Ad-53 and anti-PD-1/anti-PD-L14 June 2018–
9 June 2020
Recruiting
Mutation compensationPhase INCT03057912Disruption of HPV16- and HPV18-E6/E7 oncoproteins by TALEN and CRISPR/Cas9 approaches20 February 2017–
9 June 2017
Unknown
ImmunopotentiationPhase I/IINCT04180215Administration of HB-201 alone or in combination with HB-202 in HPV 16 + patients + checkpoint inhibitors27 November 2019–
24 May 2022
Recruiting
ImmunopotentiationPilot studyNCT00988559Different routes of administration of DNA vaccine pnGVL4a-CRT/E72 October 2009–
9 July 2018
Completed
ImmunopotentiationPhase INCT02379520Administration of HPV-16/18 E6/E7-
specific T cells + fludarabine
5 March 2015–
11 January 2022
Active, not recruiting
ImmunopotentiationPhase I/IINCT022280811Infusion of T cells expressing an HPV E6-specific TCR +
cyclophosphamide and IL-2
2 November 2014–
6 September 2017
Completed
ImmunopotentiationPhase I/IINCT02153905Infusion of T cells engineered to
express an anti-MAGE A3 HLA-A*01-restricted TCR + fludarabine
3 June 2014–
17 June 2019
Terminated
ImmunopotentiationPhase I/IINCT02111850Transfer of anti-MAGE A3-DP4-
restricted TCR + IL-2
11 April 2014–
29 March 2022
Completed
ImmunopotentiationPhase INCT00004178Transfer of anti-CEA
immunoglobulin TCR (IgTCRs)
19 April 2004–
10 June 2011
Completed
ImmunopotentiationPhase INCT00066404Delivery of human IFN-β gene using
recombinant adenovirus (Ad.IFN-β)
7 August 2003–
13 May 2020
Completed
ImmunopotentiationPhase I/IINCT01583686Administration of anti-mesothelin
CAR TCR + fludarabine
24 April 2012–
14 October 2019
Completed
Endometrial cancer gene therapy clinical trials
ImmunopotentiationPhase I/IINCT05194735Infusion of autologous T cells
engineered to express neoantigens-reactive TCRs + IL-2
18 January 2022–
11 April 2022
Recruiting
ImmunopotentiationPhase INTC00004178Transfer of recombinant anti-CEA
immunoglobulin TCR (IgTCR)
19 April 2004–
10 June 2011
Completed
ImmunopotentiationPhase INCT00066404Delivery of human IFN-β gene using
recombinant adenovirus
7 August 2003–
13 May 2020
Completed
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Drakopoulou, E.; Anagnou, N.P.; Pappa, K.I. Gene Therapy for Malignant and Benign Gynaecological Disorders: A Systematic Review of an Emerging Success Story. Cancers 2022, 14, 3238. https://doi.org/10.3390/cancers14133238

AMA Style

Drakopoulou E, Anagnou NP, Pappa KI. Gene Therapy for Malignant and Benign Gynaecological Disorders: A Systematic Review of an Emerging Success Story. Cancers. 2022; 14(13):3238. https://doi.org/10.3390/cancers14133238

Chicago/Turabian Style

Drakopoulou, Ekati, Nicholas P. Anagnou, and Kalliopi I. Pappa. 2022. "Gene Therapy for Malignant and Benign Gynaecological Disorders: A Systematic Review of an Emerging Success Story" Cancers 14, no. 13: 3238. https://doi.org/10.3390/cancers14133238

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop