Keywords
adenovirus - cervical cancer - endometrial cancer - oncolytic virotherapy - ovarian
cancer - virus
Ad Adenovirus
CAR Chimeric antigen receptor
CICs Cancer-initiating cells
CSCs Cancer stem cells
CSCC Cervical squamous cell carcinoma
CSF Colony stimulating factor
CTLs Cytotoxic T lymphocytes
DAMP Damage-associated molecular patterns
DC Dendritic cell
ECM Extracellular matrix
HPV Human papillomavirus
HSV Herpes simplex virus
ICD Immunogenic cell death
ICIs Immune checkpoint inhibitors
IFN Interferon
IL Interleukin
JAK Janus kinase
NDV Newcastle disease virus
NK Natural killer
OVs Oncolytic viruses
OVT Oncolytic virotherapy
PAMP Pathogen-associated molecular patterns
PD-1 Programmed cell death receptor-1
PFS Progression-free survival
PRROC Platinum-resistant or refractory ovarian cancer
ROS Reactive oxygen species
RSV Respiratory syncytial virus
SV Sindbis virus
TAAs Tumor-associated antigens
TCR T-cell receptor
TGF Transforming growth factor
TILs Tumor-infiltrating lymphocytes
TLRs Toll-like receptors
TME Tumor microenvironment
TNF Tumor necrosis factor
TRAIL-R Tumor necrosis factor-related apoptosis-inducing ligand
receptor
VSV Vesicular stomatitis virus
VV Vaccinia virus
Introduction
Gynecologic malignancies, such as ovarian, cervical, and endometrial cancers, pose
significant threats to women’s health worldwide and account for a substantial
portion of the global cancer burden [1].
Recent data from 2022 reveals that gynecological cancers were responsible for
approximately 1,473,427 new cases and 680,372 deaths worldwide [2]. Despite advancements in the management
of gynecological cancers, significant challenges remain, highlighting the urgent
need for innovative and targeted treatment approaches [3].
Oncolytic viruses (OVs) have gained significant attention in the twenty-first century
due to their unique ability to induce direct tumor cell destruction (oncolysis) and
modulate the immune system against cancer. As a cutting-edge therapeutic strategy,
oncolytic virotherapy (OVT) leverages the selective replication of these viruses in
tumor cells, effectively eliminating malignant cells while sparing healthy tissues
[4]
[5].
Methodology
This narrative review was conducted to evaluate the current state and future
prospects of OVT in the management of gynecological cancers. A comprehensive
literature search was performed using PubMed, Scopus, and Web of Science databases,
focusing on studies published between December 2005 and December 2024. Keywords used
included “oncolytic virotherapy,” “gynecological cancers,” “ovarian cancer,”
“cervical cancer,” “endometrial cancer,” “oncolytic viruses,” and specific virus
types like “adenovirus,” “herpes simplex virus,” and “Newcastle disease virus.”
Inclusion criteria encompassed preclinical and clinical studies assessing the
efficacy, safety, and mechanisms of OVT in gynecological malignancies. Articles
discussing combinatorial approaches involving OVs and other therapies, such as
chemotherapy, radiotherapy, or immune checkpoint inhibitors, were also included.
Exclusion criteria involved non-English publications, studies lacking experimental
or clinical data, and those focusing solely on non-gynecological cancers. Data
extraction emphasized the types of OVs, therapeutic outcomes, immune modulation
effects, tumor microenvironment (TME) changes, and advancements in OV engineering,
focusing on studies reporting clinical efficacy. The present review explores the
transformative potential of OVT in gynecological cancers, providing comprehensive
insights into its therapeutic role.
Oncolytic Viruses: Types and Mechanisms of Action
Oncolytic Viruses: Types and Mechanisms of Action
OVs are classified based on their nucleic acid type into single- or double-stranded
RNA or DNA viruses. The most common types include double-stranded (ds) DNA viruses
such as adenovirus, vaccinia virus, and herpesvirus, and single-stranded (ss) RNA
viruses, which are further categorized into positive-sense (e. g., coxsackievirus,
Seneca Valley virus, poliovirus) and negative-sense (e. g., measles virus, Newcastle
disease virus, vesicular stomatitis virus). Positive-sense ssRNA viruses can
directly translate their genetic material into proteins, while negative-sense ssRNA
viruses require transcription into complementary RNA before translation. OVs are
also classified as naturally attenuated strains or genetically engineered viral
vectors based on their structure [4].
OVT has evolved across four generations. The first generation focused on genome
modifications to enhance tumor cell specificity while minimizing damage to normal
tissues. The second generation introduced OVs armed with viral and non-viral genes.
The third generation incorporated multiple coordinated genes to enhance tumor
immunotherapy. The fourth generation advanced further by engineering OVs to activate
T cells, including the use of bi-specific T cell activators (BiTA) to bolster the
immune response [6]
[7].
Mechanisms of Oncolytic Virus-Induced Tumor Destruction
Mechanisms of Oncolytic Virus-Induced Tumor Destruction
Oncolytic viruses (OVs) utilize the following mechanisms to achieve tumor cell
destruction:
Enhances cell death
Viral infections influence cell death through death receptor-mediated pathways,
where receptors like Fas, tumor necrosis factor-related apoptosis-inducing
ligand receptor (TRAIL-R), and tumor necrosis factor receptor (TNF-R) form
death-inducing signaling complexes to trigger apoptosis. Viruses activate the
caspase cascade by regulating the interaction between death receptors and their
ligands (including viral proteins), initiating extrinsic apoptosis. This
mechanism facilitates efficient cell death and aids in viral progeny
dissemination [8]
[9]. Another mechanism by which OVs
induce cell death is necroptosis, a programmed form of cell death that mimics
necrosis in morphology [10].
Necroptosis involves plasma membrane rupture, organelle swelling, leakage of
intracellular contents, and eventual cell death. Unlike uncontrolled necrosis,
necroptosis is a regulated process and is recognized as a common pathway of
OV-induced tumor cell elimination [9]
[11]
[12]. OVs can also induce cell death
through pyroptosis, a highly inflammatory form of programmed cell death [13]
[14]. Pyroptosis is characterized by the formation of pores in the
cell membrane, leading to membrane rupture, cell lysis, and death [15]. This process not only contributes
to tumor reduction but also triggers robust antitumor immune responses. During
pyroptosis, proinflammatory cytokines like Interleukin (IL)-1β and IL-18, along
with damage-associated molecular patterns, are released, serving as adjuvants to
activate and enhance the immune system’s attack on the tumor [9].
Immune-mediated action
The primary mechanism of OVs is immune-mediated oncolysis. Effective tumor
eradication with OVs primarily depends on activating systemic innate immunity
and tumor-specific adaptive immune responses [16]. Immunovirotherapy is an advanced therapeutic approach that
harnesses viruses to specifically target and destroy tumor cells while
simultaneously activating the immune system to mount a robust anti-tumor
response [17]
[18].
Cancer cells develop strategies to suppress antitumor immunity, creating a
non-immunogenic (“cold”) TME with minimal T-cell infiltration and low mutational
burden. OVs can transform these "cold" tumors into immunogenic (“hot”)
ones by promoting tumor antigen release from dying cancer cells, boosting T-cell
infiltration, and triggering robust antitumor immune responses [19]
[20]
[21]
[22]. Tumor cell lysis releases viral
progeny along with tumor-associated antigens (TAAs), pathogen-associated
molecular patterns (PAMPs), and damage-associated molecular patterns (DAMPs),
triggering tumor immunogenic cell death (ICD). PAMPs and DAMPs activate innate
immunity by binding to receptors like Toll-like receptors (TLRs), while mature
dendritic cells (DCs) and natural killer (NK) cells are stimulated to enhance
OV-driven tumor clearance [9]
[23]. Moreover, OVs, either independently
or as therapeutic platforms, can enhance the production of inflammatory
cytokines (e. g., IL-2, IL-12, IL-15, TNF-α) [20] and chemokines (e. g., CXCL9, CXCL10, CXCL11) within the TME
[24]. This immune activation
promotes T-cell migration and infiltration, amplifying the antitumor immune
response [9]. Hence, OVs are inherently
able to transform immunologically cold tumors into hot, immune-responsive ones
by promoting immune cell and cytokine infiltration. This effect can be further
amplified by engineering OVs with transgenes that enhance their
immunostimulatory properties, directing immune responses specifically toward
cancer cells for more targeted and potent antitumor activity [25]. OVs can be engineered to deliver
immunostimulatory molecules directly into the tumor, such as tumor antigens or
cytokines, further amplifying the antitumor immune response. Furthermore, OVs
can be strategically combined with other cancer immunotherapies, including
immune checkpoint inhibitors (ICI), chimeric antigen receptors (CARs),
antigen-specific T-cell receptors (TCRs), and autologous tumor-infiltrating
lymphocytes (TILs), to create a synergistic effect that enhances tumor targeting
and improves overall therapeutic outcomes [26]
[27]
[28]
[29]. Bi- and tri-specific antibodies that target tumor antigens and
activate T-cell receptor signaling have emerged as promising tools in cancer
immunotherapy. A novel approach combines these antibodies with OVs, creating
BiTE- or TriTE-armed OVs for targeted immunotherapy. BiTEs are bispecific
proteins consisting of two single-chain variable fragments (scFvs), one
targeting a TAA and the other binding to T-cell molecule CD3. TriTEs expand this
design by adding a third binding site, such as CD28, further enhancing T-cell
activation and anti-tumor response while bypassing the need for antigen
presentation by Antigen-presenting cells [30]
[31].
Anti-angiogenic action
OVs can be engineered to target tumor vasculature or endothelial cell receptors
specifically. Their action disrupts intratumoral blood flow by recruiting
neutrophils and inducing vascular collapse. This process results in fibrin
deposition and thrombosis, ultimately contributing to tumor destruction through
the shutdown of its blood supply [4]
[32]
[33]. Furthermore, OVs compromise tumor
vasculature by targeting tumor-associated endothelial cells and nearby tumor
cells, inducing an inflammatory cascade that releases TNF-α and Interferon
(IFN)-γ [16].
Other mechanisms
The extracellular matrix (ECM) serves as a critical growth niche for most solid
tumors, forming a physical barrier that contributes to cancer initiation,
progression, metastasis, and drug resistance [34]
[35]. OVs effectively
disrupt this structural barrier, bridging the gap between non-infiltrated immune
cells and the TME. Administered intratumorally, OVs propagate autonomously,
bypassing the need for vascular delivery and enhancing drug delivery efficiency.
By encoding modifiers of ECM-related molecules, OVs induce significant
alterations in the TME, triggering the release of inflammatory mediators and
cytotoxic proteases that degrade the ECM and facilitate immune cell infiltration
[4]
[33]
[36].
[Fig. 1] depicts the mechanism of
action of oncolytic viruses.
Fig. 1 Mechanism of action of Oncolytic Viruses.
Delivery Routes of OVs
OVs can be delivered through multiple routes, including intravenous, intratumoral,
intrapleural, intraperitoneal, aerosolized, and limb injections [6].
-
Direct Intratumoral Route: Intratumoral injection remains the most
commonly used method for delivering oncolytic viruses, offering precise
control over viral concentration at the tumor site while minimizing
off-target effects in healthy tissues. This localized approach ensures
sustained exposure of the tumor to the virus, enhancing therapeutic efficacy
[37]
[38]. However, its application is
largely limited to accessible or superficial tumors, making it unsuitable
for deep-seated or multifocal malignancies such as those in the brain or
pancreas. Additionally, effective intratumoral delivery requires the
presence of viable tumor cells to support viral replication and immune
activation. Accurate tumor localization and identification of optimal
injection sites are, therefore, critical to maximize treatment benefits
[38]
[39].
-
Intravenous route: Intravenous administration of oncolytic viruses
offers a more easy, straightforward, and less invasive route of delivery
[37]. This delivery route is
particularly advantageous for treating metastatic disease, as it enables
oncolytic viruses to circulate systemically and reach multiple tumor sites
throughout the body [38]. A recent
phase III clinical trial demonstrated that intravenous infusion of
paclitaxel in combination with oncolytic reovirus yielded promising
therapeutic outcomes in patients with recurrent ovarian, fallopian tube, and
primary peritoneal cancers [40].
Additionally, emerging research has investigated the potential of enhancing
intravenously delivered oncolytic virus vaccines by incorporating functional
peptides, aiming to improve their tumor-targeting specificity and
therapeutic efficacy [41].
Theoretically, systemic or intravenous delivery represents an optimal
strategy, as it allows widespread distribution of oncolytic viruses,
enabling them to target both primary tumors and distant metastases and is
amenable to repeated dosing. However, a major limitation is the rapid
clearance of viral particles by the host immune system, and poor penetration
into tumors [42]. Furthermore, this
strategy often requires the administration of high viral titers to ensure
adequate distribution and therapeutic efficacy, which may increase the risk
of off-target effects, including toxicity and unintended infection of
healthy tissues [38]
[43]. To overcome this challenge,
recent advances have explored the use of synthetic nanoparticle coatings to
shield oncolytic viruses, thereby enhancing their circulation time and
reducing neutralization by antiviral antibodies [39].
-
Intraperitoneal Route: Given the expansive surface area of the
peritoneum, drugs delivered via intraperitoneal injection are rapidly
absorbed, making this route particularly effective for targeting
intra-abdominal organs [37].
Intraperitoneal administration of oncolytic viruses offers a promising
strategy to directly reach peritoneal tumors, providing localized
therapeutic delivery to manage peritoneal metastases more efficiently [38]. In a preclinical study, the
combination of the oncolytic virus JX-594 with immune checkpoint inhibitors
(ICIs), administered intraperitoneally, demonstrated significant therapeutic
potential. This approach not only reversed the immunosuppressive environment
within the peritoneal cavity but also amplified the efficacy of immune
checkpoint blockade against colon cancer, resulting in substantial
suppression of peritoneal carcinomatosis and malignant ascites [38]
[44].
-
Other routes: Other less frequently utilized delivery routes for
oncolytic viruses include subcutaneous, intrathecal, intrapleural, oral, and
limb injections, primarily due to their limited efficiency and narrow
therapeutic scope. While subcutaneous injection is commonly employed in
small animal models, particularly when intravenous access is challenging, it
has limited clinical applicability [37]
[45]. Intrathecal and
intrapleural routes, meanwhile, are confined to specific anatomical regions,
with intrathecal injections targeting central nervous system malignancies
and intrapleural injections being applicable primarily to thoracic tumors
such as those in the lungs or pleura [37]. Furthermore, aerosolized and intranasal delivery of
oncolytic viruses are emerging as innovative approaches for cancer therapy,
particularly in the treatment of pulmonary malignancies such as lung cancer
[46].
Exploring the Efficacy of OVT in Gynecologic Cancer Therapy
Exploring the Efficacy of OVT in Gynecologic Cancer Therapy
OVT is emerging as a pivotal approach in managing gynecological malignancies,
demonstrating encouraging clinical outcomes. Various OVs are employed in treating
these cancers as standalone therapies or in synergy with other treatment modalities
[47]
[48]. Moreover, integrating OVs with other therapies significantly
improves cancer treatment outcomes. These viruses trigger localized tumor
inflammation, enhancing immune responses and amplifying the effects of
immunotherapy. This strategy helps the immune system identify and destroy cancer
cells while counteracting the tumor’s immune evasion tactics [16]. Studies have demonstrated that
combining oncolytic vaccinia virus (VV) therapy with conventional cancer treatments
yields a synergistic effect, proving more effective than standalone therapies [49]
[50].
Role of OVT in the Management of Ovarian Cancer
OVs present a highly promising strategy for the treatment of ovarian cancer,
especially for therapy-resistant ovarian cancers [51]. Despite debulking surgery and
chemotherapy treatment strategy, most patients experience relapse and develop
drug-resistant metastatic disease, often driven by cancer stem cells (CSCs) or
cancer-initiating cells (CICs). OVs offer a promising alternative by bypassing
traditional drug-resistance mechanisms, potentially providing a safe and
effective therapy for chemotherapy-resistant CSCs/CICs. Furthermore, Antibodies
against immune checkpoint proteins like anti-cytotoxic T-lymphocyte antigen-4
(CTLA-4) and programmed death protein-1 (PD-1) have been approved for ovarian
cancer treatment, showing durable clinical benefits. However, their efficacy
often depends on a pre-existing active immune TME. Integrating oncolytic
therapies with checkpoint inhibitors provides a synergistic strategy, boosting
tumor-specific immune responses while counteracting immune suppression, thereby
enhancing overall therapeutic outcomes [52].
Adenoviruses and vesicular stomatitis virus (VSV) are among the most widely
studied and commonly employed vectors in virotherapy for ovarian cancers for
their ability to preferentially replicate in cancer cells and induce oncolysis
[53]
[54]. The role of various OVs in managing
advanced ovarian cancer is explored in the following studies:
-
Adenovirus: A study showed that two ovarian oncolytic
adenoviruses, OvAd1, developed using Matrigel cultures, and OvAd2, from
traditional monolayers, effectively target platinum-resistant ovarian
cancer cell lines while sparing normal cells, offering a 200-fold
therapeutic window. Unlike adenovirus-5 (Ad5)-based therapies, neither
virus caused peritoneal adhesions, making OvAd1 and OvAd2 promising
candidates for treating aggressive ovarian cancer [55]. In another study, Ad-5 was
engineered into Ad5NULL-A20, a tumor-selective virotherapy designed to
target αvβ6 integrin, a marker overexpressed in aggressive epithelial
ovarian cancers. This advanced vector enables precise local and systemic
targeting of αvβ6-positive tumors, offering a versatile platform for
delivering tumor-specific anticancer therapies, including ICIs [56]. A study explored the impact
of oncolytic adenovirus on malignant ascites in a mouse model with
advanced ovarian cancer. OV effectively reduced ascites formation and
extended overall survival. Immune profiling revealed that OV treatment
enhanced T cell infiltration, activation, and effector differentiation,
reprogrammed macrophages to an M1-like phenotype, and improved
CD8+/CD4+T cell and M1/M2 macrophage ratios. It was observed that
combining OV with the colony-stimulating factor 1 receptor (CSF-1R)
inhibitor PLX3397 and anti-PD1 therapy significantly delayed ascites
progression, further amplifying T cell infiltration, activation, and
proliferation [57]. A recent
study engineered an oncolytic adenovirus, Ad5/3-E2F-d24-aMUC1aCD3-IL-2
(TILT-322), armed with a human aMUC1aCD3 T-cell engager and
interleukin-2 (IL-2), and evaluated its efficacy on ascites samples
derived from ovarian cancer patients with peritoneal carcinomatosis.
This treatment enhanced T cell cytotoxicity, increasing granzyme B,
perforin, and IFN-γ levels. Immune profiling showed that TILT-322 also
activated gamma delta T cells and impacted NK and NK-like T cells.
Moreover, it reduced the proportion of exhausted CD8+T cells in ovarian
ascites. Hence, TILT-322 shows great promise as a novel anti-tumor agent
for clinical use [58]. A
similar study utilizing the oncolytic adenovirus
Ad5/3-E2F-D24-hTNFa-IRES-hIL2 (TILT-123) to deliver TNF-α and IL-2
demonstrated its potential to overcome immunosuppression and boost
antitumor T cell infiltrates (TILs) in ovarian cancer. The treatment
reprogrammed the TME to enhance TIL reactivity, potentially improving
the clinical outcomes of adoptive TIL therapy in patients with advanced
ovarian cancer [59].
Furthermore, A novel oncolytic adenovirus, AR2011, was developed to
target ovarian tumors and demonstrated potent lytic effects in vitro on
human ovarian cancer cell lines and ascitic fluid-derived malignant
cells. When preloaded into menstrual blood stem cells (MenSCs), AR2011’s
activity was enhanced, overcoming the inhibitory effects of ascitic
fluids. MenSC-AR treatment in nude mice with peritoneal carcinomatosis
effectively inhibited tumor growth, showing that MenSCs can amplify the
oncolytic effects of AR2011. This strategy offers a promising approach
to overcoming viral treatment barriers in ovarian cancer [60]. A study evaluating the
efficacy of Enadenotucirev, a tumor-selective and blood-stable
adenoviral vector, in recurrent platinum-resistant ovarian cancer,
reported promising results. Administering enadenotucirev intravenously
in combination with paclitaxel showed manageable tolerability, an
encouraging median progression-free survival (PFS), and enhanced
immune-cell infiltration within tumors, highlighting its potential as a
therapeutic option for platinum-resistant ovarian cancer [61]. A similar study
demonstrated that intravenous administration of enadenotucirev combined
with nivolumab showed manageable tolerability, improved overall
survival, and induced immune cell infiltration and activation in
patients with advanced or metastatic epithelial ovarian cancer,
suggesting its potential as a promising treatment approach [62].
-
Vesicular stomatitis virus (VSV): A study evaluated the oncolytic
VSV in patient-derived ovarian cancer cell lines across all epithelial
subtypes. The findings revealed that combining VSV with Janus kinase
(JAK) inhibitors, such as ruxolitinib, significantly enhanced
therapeutic efficacy. These results suggest that VSV, either alone or in
combination with JAK inhibitors, holds promise as a potent treatment
option for ovarian cancer [63].
A similar study revealed that in metastatic breast and ovarian cancer
models, combining oncolytic VSV or reovirus with NK-T cell activation
via DCs loaded with α-galactosylceramide significantly reduced tumor
burden. This synergistic approach enhanced survival and decreased
metastases more effectively than either treatment alone, highlighting
its potential as a powerful therapeutic strategy [64].
-
Herpes simplex virus: A study evaluated an oncolytic herpes
simplex virus (oHSV) armed with murine IL-12, demonstrating its efficacy
against ovarian cancer cell lines in vitro. Mice treated with the
IL-12-expressing oHSV showed enhanced tumor antigen-specific CD8+T-cell
responses in the omentum and peritoneal cavity, resulting in better
control of ovarian cancer metastases and improved survival. These
findings underscore the potential of IL-12-expressing oHSV to reduce
metastasis and improve outcomes in ovarian cancer [65].
-
Poxvirus: A study revealed that CF17, a chimeric poxvirus created
from nine orthopoxvirus species, has enhanced oncolytic properties. In
ovarian cancer, CF17 replicated and induced cytotoxicity in human and
mouse cell lines. It also demonstrated strong antitumor effects in a
syngeneic ovarian cancer mouse model at doses as low as 6×10⁶
plaque-forming units. These findings suggest CF17’s potential as a
promising treatment for aggressive ovarian cancer [66]. A similar study explored
the immunotherapeutic potential of Parapoxvirus ovis (OrfV) using the
ID8 orthotopic mouse model of advanced epithelial ovarian carcinoma. The
findings demonstrated that OrfV effectively reduced tumor progression as
a monotherapy in an advanced-stage cancer model. The antitumor effects
of OrfV were heavily reliant on NK cells, as their depletion eliminated
CD8+T-cell-mediated responses against the tumor. These results highlight
OrfV as a promising NK cell-stimulating immunotherapy for the treatment
of advanced epithelial ovarian cancer [67]. Another recent study on the
OrfV highlighted its dual role as an oncolytic agent and immune
modulator. In a preclinical advanced epithelial ovarian cancer model,
OrfV enhanced immune cell infiltration into the ascites TME, boosting
activation markers and effector cytokines. This immune activation
correlated with reduced tumor burden and prolonged survival,
underscoring its potential as a multifaceted therapy for advanced-stage
ovarian cancer [68]. A study
evaluating intraperitoneal olvimulogene nanivacirepvec (Olvi-Vec), a
modified vaccinia virus in patients with platinum-resistant or
refractory ovarian cancer (PRROC) demonstrated encouraging results. The
treatment showcased a favorable safety profile, notable clinical
activity, and robust immune activation, highlighting its potential as a
therapeutic option for managing PRROC [69]. A related trial
investigated the efficacy and safety of intraperitoneal Olvi-Vec
virotherapy in combination with platinum-based chemotherapy, with or
without bevacizumab, in patients with PRROC. The findings revealed that
Olvi-Vec, followed by immunochemotherapy, achieved encouraging objective
response rates and PFS while maintaining a manageable safety profile,
offering a potential therapeutic strategy for PRROC patients [70].
-
Sindbis virus: The Sindbis virus-based vaccine platform has
emerged as a promising immunotherapy candidate for managing ovarian
cancer. Armed oncolytic Sindbis virus (SV) vectors have demonstrated
unique and compelling properties in vivo, sparking considerable interest
in their potential applications in recent years [71]. A recent study highlighted
the potential of the SV vector platform, specifically SVIL-12, combined
with an agonistic OX40 antibody, in eradicating ovarian cancer in a
Mouse Ovarian Surface Epithelial Cell model. Remarkably, this
combination also prevented tumor recurrence in mice rechallenged with
tumor cells after approximately five months. Additionally, engineering a
single SV vector, SV.IgGOX40.IL-12, to co-express IL-12 and anti-OX40
enabled precise local delivery of immunomodulatory agents to tumors,
significantly enhancing the antitumor immune response [72].
-
Maraba virus: A study demonstrated that a prime-boost strategy
combining a vaccine with antigen-armed oncolytic Maraba virus elicited
strong tumor-specific CD8+T cell responses, improving tumor control in
ovarian cancer. While adaptive resistance led to T cell suppression in
the TME, adding PD-1 blockade restored T cell function and enhanced
outcomes [73].
Hence, OVs show immense potential as a therapeutic strategy for managing ovarian
cancers, with ongoing research continuously uncovering new possibilities.
Role of OVT in the Management of Cervical Cancer
OVs have shown promise in the management of advanced stages of cervical cancer,
acting as potent biological agents that selectively infect and destroy cancer
cells while sparing healthy tissues [74]. In cervical cancer OVT, the primary viruses utilized include
adenoviruses, herpes simplex viruses, parvoviruses, and Newcastle disease virus
[74]. The role of various OVs in
managing advanced cervical cancer is explored in the following studies:
-
Adenovirus: The genetically engineered oncolytic adenovirus CRAd
AdCB016-mp53(268 N) demonstrated remarkable specificity in targeting
Human papillomavirus (HPV)-positive cells by combining selective
replication in HPV E6/E7-expressing cells with a p53 variant resistant
to E6-mediated degradation. This virus exhibited 10- to 1000-fold
greater efficacy in HPV-positive cervical cancer and dysplastic cell
lines while sparing healthy keratinocytes, underscoring its precision
and significant therapeutic potential [75]. Another study introduced
the novel recombinant adenovirus M5, designed for tumor-specific
replication and targeted expression of the HPV16 E2 gene. This virus
effectively silenced HPV E6 and E7 oncogenes in cervical cancer cells,
demonstrating strong antitumor activity both in vitro and in vivo. Its
therapeutic potency was significantly enhanced when combined with
radiation therapy [76]. A
similar study on the E1A-mutant adenovirus (M6) with antisense HPV16
E6/E7 DNA showed selective replication in HPV16-positive cervical cancer
cells, suppressing E6/E7 expression, inducing apoptosis, and reducing
invasiveness. Combined with radiotherapy, M6 enhanced tumor inhibition,
increased apoptosis, and significantly improved survival in
tumor-bearing mice compared to monotherapy or Adv5/dE1A with radiation
[77]. Recently it was
reported that oncolytic adenoviruses effectively infect and lyse
cervical cancer cells, triggering tumor cell disruption and immune
activation. These viruses can deliver tumor suppressor genes like p53
and Rb, restoring their normal function in cancer cells. Adenoviruses
targeting E6 and E7 oncoproteins enhance immune responses, particularly
activating cytotoxic T lymphocytes (CTLs). Additionally, the integration
of antisense RNAs and microRNAs (miRNAs) into adenoviral vectors
specifically inhibits HPV E6 and E7 expression, effectively suppressing
these oncoproteins in cervical cancer cells [78]. Another study investigated
the use of adenovirus types 26 and 35 vectors expressing HPV16 E6 and E7
oncoproteins in mice through intramuscular priming followed by
intravaginal (Ivag) boosting. This combined immunization strategy
significantly enhanced HPV-specific CD8+T cell responses, evidenced by
increased production of IFN-γ and TNF-α, along with upregulation of CD69
and CD103, markers of tissue-resident memory cells. Additionally, it
induced circulating HPV-specific CD8+T cells, highlighting the potential
of Ivag immunization with adenoviral vectors as a promising therapeutic
approach for HPV infections and cervical intraepithelial neoplasia [79].
-
Herpes simplex virus (HSV): A recent study highlighted that
therapy with a triple-mutated oncolytic HSV (T-01) significantly
enhances tumor immunogenicity and boosts the efficacy of ICIs in
treating HPV-associated cervical cancer [80]. A related study
investigated the use of a triple-mutated oncolytic HSV (T-01) for
treating HPV-associated cervical cancer. The findings revealed that T-01
exhibited potent cytotoxicity across all tested cell lines. In both the
HeLa xenograft and TC-1 syngeneic mouse models, T-01 significantly
suppressed tumor growth. Additionally, tumor-bearing mice treated with
T-01 demonstrated a marked increase in CD8+T-cell precursors compared to
control mice, highlighting the therapeutic potential of T-01 for
HPV-related cervical cancer [81]. A recent study introduced SONC103, a recombinant
oncolytic HSV-1 armed with CRISPR/Cas9, to disrupt integrated HPV16
genes in cervical cancer cells. SONC103 effectively knocked down HPV16
oncogenes, reducing cell proliferation and inducing apoptosis. It
eliminated HPV16 DNA probes from chromosomes, downregulated E6/E7
oncoproteins, and upregulated tumor suppressor proteins p53 and pRB. In
a murine cervical cancer model, SONC103 significantly inhibited tumor
growth, highlighting its potential as a targeted therapy for
HPV16-positive cervical cancer [82].
-
Newcastle disease virus (NDV): A study investigated the Hitchner
B1 (HB1) strain of NDV as an oncolytic agent for cervical cancer. The
findings showed that HB1 NDV infection significantly reduced TC-1 cell
viability in a dose-dependent manner. The virus-induced reactive oxygen
species (ROS) production, apoptosis, and autophagy. Additionally, NDV
treatment upregulated cytochrome-C expression while downregulating
survivin levels. These results position the HB1 strain of NDV as a
promising candidate for cervical cancer therapy [83]. Another study on the
oncolytic NDV vaccine strain LaSota in TC-1 cells expressing HPV-16
E6/E7 antigens revealed that NDV significantly reduced cell viability
and suppressed growth by inducing apoptosis through ROS production.
These findings suggest, NDV as a promising selective antitumor agent for
cervical cancer therapy [84]. A
similar study highlighted the therapeutic potential of NDV in enhancing
the efficacy of Doxorubicin for cervical cancer in mouse models. The
combined treatment significantly improved survival rates, slowed tumor
progression, and increased nitric oxide and lactate dehydrogenase levels
in splenocytes. Additionally, it elevated TNF-α, IL-12, and IFN-γ levels
while reducing TGF-β and IL-4 secretion compared to NDV or Doxorubicin
alone. These findings reveal that NDV is a potent adjunct to
chemotherapy for cervical cancer [85].
-
Coxsackievirus: A recent study highlighted the oncolytic potential
of coxsackievirus B3 strain 2035 A (CVB3/2035 A) in cervical squamous
cell carcinoma (CSCC). The virus demonstrated potent anti-tumor activity
in CSCC cell lines, xenografts, and patient-derived tissue cultures and
organoids. Notably, CVB3/2035 A exhibited synergistic effects when
combined with paclitaxel, enhancing its therapeutic impact. These
findings suggest that CVB3/2035 A could be a promising alternative or
complement to existing CSCC chemotherapy regimens [86].
-
Respiratory syncytial virus: A recent study explored the antitumor
activity and underlying molecular mechanisms of the oncolytic
Respiratory syncytial virus- A2 (RSV-A2) in TC-1 cancer cells, a model
for HPV-related cervical cancers. The results demonstrated that RSV-A2
exhibited potent cytotoxic effects on HPV-associated cervical cancer
cells. It induced apoptosis and autophagy, activated caspase-3, promoted
ROS generation, and inhibited the cell cycle in the TC-1 cell line.
These findings highlight RSV-A2 as a promising candidate for the
treatment of cervical cancer [87].
Role of OVT in the Management of Endometrial Cancer
The potential of OVT in endometrial cancer remains largely underexplored, with
only a limited number of studies investigating its therapeutic applications.
Despite this, OVT is rapidly emerging as a promising and innovative approach for
managing endometrial cancer, offering new avenues for treatment and highlighting
the need for further research in this area [88]. Few OVs have been explored to treat endometrial cancer. One
study investigated the oncolytic potential of coxsackievirus B3 strain 2035 A
(CV-B3/2035 A) as a novel therapeutic option. The findings demonstrated that
CV-B3/2035 A exhibited strong oncolytic activity in human endometrial cancer
cell lines both in vitro and in vivo, as well as in patient-derived endometrial
cancer samples ex vivo. These results suggest that CV-B3/2035 A holds promise as
an alternative virotherapy agent for endometrial cancer treatment [89]. Another study compared the efficacy
of the Edmonston strain of measles virus and VSV against endometrial cancer and
found VSV to be more effective. Intratumoral VSV led to faster tumor regression
than measles virus, while intravenous VSV achieved complete tumor control in all
treated mice [90].
[Table 1] depicts the role of OVT in
gynecological cancers.
Table 1 Role of Oncolytic Viruses in the Management of
Gynecological Cancers.
Author & Year of Study
|
Country of Study
|
OV and its type
|
Nucleic acid type
|
Study Model
|
Delivery Route
|
Main Results
|
Reference
|
OVARIAN CANCER
|
Kuhn, et al., 2016
|
United States of America & United Kingdom
|
Adenovirus (OvAd1 and OvAd2)
|
dsDNA
|
Ovarian cancer cell line SKOV3
|
Intratumoral (Virus incubated with cell lines)
|
-
Developed OvAd1 and OvAd2 OVs.
-
Effective against platinum-resistant ovarian cancer
cell lines.
-
Neither virus caused peritoneal adhesions as observed
with Ad-5.
|
[55]
|
Thomas, et al., 2016
|
United States of America
|
Herpes simplex virus armed" with murine IL-12
|
dsDNA
|
Murine and Human ovarian cancer cell lines
|
Intraperitoneal
|
-
Enhanced tumor antigen-specific CD8+T-cell responses
in the omentum and peritoneal cavity
-
Better control of ovarian cancer metastases
-
Improved overall survival
|
[65]
|
Alfano, et al., 2017
|
Argentina
|
Adenovirus (AR2011)
|
dsDNA
|
Human ovarian cancer cell lines
|
Intratumoral (Virus incubated with cell lines)
|
|
[60]
|
Uusi-Kerttula, et al., 2018
|
United Kingdom
|
Adenovirus (Ad5NULL-A20)
|
dsDNA
|
Mice model and SKOV3 xenograft model of human epithelial
ovarian cancer
|
Intravenous
|
-
OVs reduced ascites formation
-
Extended overall survival.
-
Enhanced T cell infiltration, activation, and
effector differentiation, reprogrammed macrophages,
and improved CD8+/CD4+T cell and M1/M2 macrophage
ratios.
|
[56]
|
McGray, et al., 2019
|
Canada
|
Maraba virus (antigen-armed)
|
Negative-sense ssRNA
|
Mice model
|
Intraperitoneal/Intravenous
|
|
[73]
|
Santos, et al., 2020
|
Finland
|
Adenovirus (Ad5/3-E2F-D24-hTNFa-IRES-hIL2)
|
dsDNA
|
Cultures derived from patients with advanced ovarian
cancer
|
Intratumoral
|
|
[59]
|
Hamma, et al., 2020
|
United States of America
|
Parapoxvirus ovis (CF17)
|
dsDNA
|
Human and mouse cancer cell lines
|
Human cell lines and Intraperitoneal injection in mice.
|
|
[66]
|
Shi, et al., 2021
|
China
|
Adenovirus
|
dsDNA
|
Mice
|
Intraperitoneal
|
-
Reduced ascites development in advanced ovarian
cancer
-
Prolonged overall survival.
-
Ascitic immune microenvironment revealed promoted T
cell infiltration, activation, and differentiation
into effector phenotype, reprogrammed macrophages,
and improved CD8+/CD4+T cell and M1/M2 macrophage
ratios.
|
[57]
|
Moreno, et al., 2021
|
Spain
|
Adenovirus (Enadenotucirev plus paclitaxel)
|
dsDNA
|
Humans
|
Intraperitoneal
|
|
[61]
|
Gebremeske, et al., 2021
|
Canada
|
Vesicular stomatitis virus
|
Negative-sense ssRNA
|
Mice
|
Intravenous
|
|
[64]
|
Manyam, et al., 2021
|
United States of America
|
Modified vaccinia virus (Olvi-Vec)
|
dsDNA
|
Humans
|
Intraperitoneal
|
|
[69]
|
van Vloten, et al., 2022
|
Canada
|
Parapoxvirus ovis
|
dsDNA
|
Orthotopic mouse model of end-stage epithelial ovarian
carcinoma
|
Intraperitoneal
|
|
[67]
|
Opp, et al., 2022
|
United States of America
|
Sindbis virus
|
Positive-sense ssRNA
|
Mouse Ovarian Surface Epithelial Cell Line model
|
Intraperitoneal
|
|
[72]
|
Fakih, et al., 2023
|
United States of America
|
Adenovirus (Enadenotucirev plus nivolumab)
|
dsDNA
|
Humans
|
Intravenous
|
|
[62]
|
Minott, et al., 2023
|
Canada
|
Parapoxvirus ovis
|
dsDNA
|
Murine model of late-stage ovarian cancer
|
Intraperitoneal
|
|
[68]
|
Holloway, et al., 2023
|
United States of America
|
Modified vaccinia virus (Olvi-Vec)
|
dsDNA
|
Humans
|
Intraperitoneal
|
|
[70]
|
Basnet, et al., 2024
|
Finland
|
Adenovirus (Ad5/3-E2F-d24-aMUC1aCD3-IL-2 armed with human
aMUC1aCD3 T cell engager and IL-2)
|
dsDNA
|
Patient-derived ovarian cancer xenograft models
|
Intravenous
|
-
Enhanced T cell cytotoxicity, increasing granzyme B,
perforin, and IFN-γ levels.
-
Immune profiling revealed activated gamma delta T
cells and NK and NK-like T cells.
-
Reduced exhausted CD8+T cells in ovarian ascites.
|
[58]
|
Geoffroy, et al., 2024
|
Canada
|
Vesicular stomatitis virus
|
Negative-sense ssRNA
|
Patient-derived ovarian cancer cell lines
|
Intratumoral (Virus incubated with cell lines)
|
|
[63]
|
CERVICAL CANCER
|
Heideman, et al., 2005
|
Amsterdam
|
Adenovirus (CRAd AdCB016)
|
dsDNA
|
Human cervical carcinoma cell lines
|
Intratumoral (Virus incubated with cell lines)
|
|
[75]
|
Wang, et al., 2010
|
China
|
Adenovirus (M6)
|
dsDNA
|
HPV16-positive cervical cancer cell lines
|
Intravenous
|
|
[77]
|
Wang, et al., 2011
|
China
|
Adenovirus (M5)
|
dsDNA
|
Human cervical carcinoma cell lines
|
Intratumoral (Virus incubated with cell lines)
|
|
[76]
|
Çuburu, et al., 2018
|
United States of America & Netherlands
|
Adenovirus types 26 and 35
|
dsDNA
|
Mice
|
Intramuscular and/or Intravaginal
|
|
[79]
|
Mozaffari Nejad, et al., 2020
|
Iran
|
Newcastle disease virus (Hitchner B1 strain)
|
Negative-sense ssRNA
|
Murine TC-1 cell line
|
Intratumoral (Virus incubated with cell lines)
|
|
[83]
|
Keshavarz, et al., 2020
|
Iran
|
Newcastle disease virus (vaccine strain LaSota)
|
Negative-sense ssRNA
|
Murine TC-1 cells
|
Intratumoral (Virus incubated with cell lines)
|
|
[84]
|
Kagabu, et al., 2021
|
Japan
|
Herpes simplex virus (T-01)
|
dsDNA
|
Human cervical cancer cell lines
|
Intratumoral
|
-
Potent cytotoxicity
-
Suppressed tumor growth
|
[81]
|
Kagabu, et al., 2023
|
Japan
|
Herpes simplex virus (T-01)
|
dsDNA
|
Murine model
|
Intratumoral
|
|
[80]
|
Samadi, et al., 2023
|
Iran
|
Respiratory syncytial virus (RSV-A2)
|
Negative-sense ssRNA
|
TC-1 cancer cell lines
|
Intratumoral (Virus incubated with cell lines)
|
-
Potent cytotoxic effects on HPV-associated cervical
cancer cells
-
Induced apoptosis and autophagy, activated caspase-3,
promoted ROS generation and inhibited the cell
cycle
|
[87]
|
Hu, et al., 2024
|
China
|
Herpes simplex virus (SONC103, armed with a CRISPR/Cas9 gene
editing system)
|
dsDNA
|
Murine xenograft cervical cancer model
|
Intratumoral
|
-
Effectively knocked down HPV16 oncogenes
-
Reduced cell proliferation
-
Induced apoptosis and inhibited tumor growth
|
[82]
|
Rasekhi Kazerun, et al., 2024
|
Iran
|
Newcastle disease virus
|
Negative-sense ssRNA
|
Mice
|
Intratumoral
|
-
Enhances efficacy of Doxorubicin
-
Combined treatment with Doxorubicin significantly
improved survival rates, slowed tumor
progression
|
[85]
|
Lin, et al., 2024
|
China
|
Coxsackievirus (CVB3/2035 A)
|
Positive-sense ssRNA
|
Human Cervical squamous cell carcinoma cell lines in vitro
and mouse xenograft models in vivo
|
Intratumoral/ Intravenous
|
|
[86]
|
ENDOMETRIAL CANCER
|
Liu, et al., 2018
|
United States of America
|
Edmonston strain of measles virus and vesicular stomatitis
virus
|
Negative-sense ssRNA
|
Mice
|
Intratumoral/ Intravenous
|
|
[90]
|
Lin, et al., 2018
|
China
|
Coxsackievirus (CV-B3/2035 A)
|
Positive-sense ssRNA
|
Human endometrial cancer cell lines
|
Intratumoral/ Intravenous
|
|
[89]
|
Safety issues and Limitations of Oncolytic virotherapy
Safety issues and Limitations of Oncolytic virotherapy
While OVT shows great potential, several hurdles limit its effectiveness. Effective
delivery of OVs faces significant challenges. Systemic administration often leads
to
rapid immune clearance, while intratumoral injection is impractical for inaccessible
or metastatic tumors [16]
[91]
[92]. The immunosuppressive TME further restricts OV spread and
replication [93]. Additionally,
pre-existing immunity to common viruses can neutralize OVs before they reach tumor
cells, and tumors may develop resistance mechanisms, reducing therapeutic efficacy
over time [16].
In addition to these, there are safety concerns associated with OVT. The most common
side effects of OVT include low-grade systemic symptoms and local reactions at the
injection site. Fever is the most frequently observed treatment-related adverse
event, with additional symptoms such as chills, nausea, vomiting, flu-like symptoms,
fatigue, and pain also reported [94].
Delivery challenges and the activation of immune checkpoints after therapy can
suppress immune responses, leading to resistance. Moreover, the restricted
accessibility of viral receptors in tight junctions, robust interferon-mediated
antiviral defenses, and abnormalities in gene expression required for viral
replication and infection reduce the ability of OVs to target and destroy tumor
cells [95]. Other challenges include poor
penetration into tumor masses, immune responses that neutralize the virus,
off-target infections, adverse conditions within the TME, and the lack of dependable
predictive or therapeutic biomarkers [96].
Monotherapies often show limited effectiveness in cancer management. OVT can be
combined with other treatment modalities such as chemotherapy, immunotherapy,
targeted therapies, ICIs, and adoptive cell therapies to enhance therapeutic
outcomes. These combinations aim to improve response rates and overall efficacy
[9]
[16]. Additionally, ongoing research is exploring the potential of OVT and
viral oncogenes in targeting CSCs, aiming to improve cancer management strategies
[97].
Conclusion
Oncolytic virotherapy represents a groundbreaking strategy for treating gynecological
cancers, with various virus platforms such as adenoviruses, herpes simplex viruses,
vaccinia viruses, and reoviruses showing promising therapeutic potential. These
viruses not only selectively lyse tumor cells but also reprogram the TME to enhance
anti-tumor immunity. Significant progress has been made in ovarian and cervical
cancers, particularly with combination therapies involving ICIs and chemotherapeutic
agents, resulting in improved survival and durable responses. However, the
application of OVT in endometrial cancer remains underexplored, potentially due to
the relatively lower mutation burden of endometrial tumors and the challenges of
optimizing virus delivery and efficacy in this context. Future efforts should focus
on tailoring virus platforms to address tumor-specific challenges, improving
delivery mechanisms, and combining virotherapy with emerging immunomodulatory
agents. Comprehensive preclinical and clinical investigations are essential to
unlock the full potential of OVT across all gynecological cancers, paving the way
for innovative and personalized treatment options for patients with advanced or
resistant diseases.
Study Limitations
The review is based on existing literature, which may be influenced by publication
bias or the lack of unpublished studies. Future systematic reviews and meta-analyses
are essential for conducting quantitative assessments and addressing potential gaps
in the current research to enhance the accuracy and comprehensiveness of
findings.
Declaration of use of AI
During the writing process of this paper, the author(s) used ChatGPT in order to
improve the English language and clarity of the manuscript. The assistance provided
was limited to enhancing the quality of writing. The author(s) reviewed and edited
the text and take(s) full responsibility for the content of the paper.
Authors’ contributions
NK: Conceptualization, Literature search, Data collection, Formal analysis,
Data interpretation, writing original drafts, writing review and editing, final
review, and approval of the manuscript.