Next Article in Journal
Solute Transport across the Lymphatic Vasculature in a Soft Skin Tissue
Previous Article in Journal
Plant–Microbes Interaction: Exploring the Impact of Cold-Tolerant Bacillus Strains RJGP41 and GBAC46 Volatiles on Tomato Growth Promotion through Different Mechanisms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

TIME Is Ticking for Cervical Cancer

1
Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), 1700 Tulane Avenue, New Orleans, LA 70012, USA
2
Louisiana Children’s Medical Center Cancer Center, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), 1700 Tulane Avenue, New Orleans, LA 70012, USA
*
Authors to whom correspondence should be addressed.
Biology 2023, 12(7), 941; https://doi.org/10.3390/biology12070941
Submission received: 8 June 2023 / Revised: 23 June 2023 / Accepted: 27 June 2023 / Published: 30 June 2023
(This article belongs to the Topic Advances in Tumor Microenvironment)

Abstract

:

Simple Summary

Cervical cancer (CC) is a major health problem in women of childbearing age. For example, CC is the fourth most common cancer in women across the world with an estimated 604,000 new cases in 2020. CC comprised 342,000 deaths worldwide, mainly in women in low- and middle-income countries (LMICs). Frequent CC screening and human papillomavirus (HPV) vaccination programs have significantly decreased the incidence of CC in the developed world. However, the last three years of the coronavirus disease-19 (COVID-19) pandemic have significantly increased CC incidence by disrupting the vaccination program. Understanding the immunological network and environment in the CC tumor microenvironment (TME) called the tumor immune microenvironment (TIME) will help to design immune cell-specific immunotherapeutic approaches for CC patients at different stages. The current article introduces the epidemiology socioeconomic burden of CC, the immune microenvironment in the cervix of healthy women, and its involvement in CC immunopathogenesis. Later sections discuss different immune cells, which comprise CC TIME and its targeting for immunotherapeutic approaches. Therefore, it is critical to understand CC TIME to save our women’s lives from CC throughout the world.

Abstract

Cervical cancer (CC) is a major health problem among reproductive-age females and comprises a leading cause of cancer-related deaths. Human papillomavirus (HPV) is the major risk factor associated with CC incidence. However, lifestyle is also a critical factor in CC pathogenesis. Despite HPV vaccination introduction, the incidence of CC is increasing worldwide. Therefore, it becomes critical to understand the CC tumor immune microenvironment (TIME) to develop immune cell-based vaccination and immunotherapeutic approaches. The current article discusses the immune environment in the normal cervix of adult females and its role in HPV infection. The subsequent sections discuss the alteration of different immune cells comprising CC TIME and their targeting as future therapeutic approaches.

1. Introduction

Cervical cancer (CC) is a common cancer affecting women worldwide. For example, 604,000 women across the world suffered and 342,000 women died (mainly in low- and middle-income countries (LMICs)) from CC in 2020 [1]. Although it can be prevented, it still affects an average of 7.7 out of every 100,000 women in the US and is a leading cause of death in young women [1]. In recent years, the incidence of CC in US women between 30 and 34 years old has increased, reaching 11.60/100,000 in 2019 [2]. Unfortunately, the incidence of stage IV CC is higher among Black women as compared to White women [3] and the cost of treating CC is approximately $56,250 for the first year alone [4].
Squamous-cell carcinoma (SCC) accounts for 80% of cases, while cervical adenocarcinoma (CAC) is less common but more severe, making up 15% of all cases [5]. Lifestyle factors can also impact the risk of CC, with an increased number of full-term pregnancies associated with a higher incidence of high-risk precancerous lesions. Additionally, prolonged use of oral contraceptives is associated with high-risk lesions and invasive CC [6]. However, the most significant risk factor for CC is human papillomavirus (HPV), which is associated with 99.7% of all cases [7,8]. Although the introduction of HPV vaccines in 2006 has likely helped reduce the prevalence of CC, it remains a significant cause of cancer deaths [9].
Recent studies have shown that the Cervarix HPV vaccine (GlaxoSmith Kline) protects females from specific HPV-induced CC but may increase their susceptibility to HPV genotypes not targeted by the vaccine [10]. In addition, women who receive the vaccine may still develop grade 2 or 3 cervical intraepithelial neoplasia (CIN) due to nonpreventable HPV types at higher rates than unvaccinated women [11,12,13]. Despite the strong correlation between CC, HPV infection, and the immune system, little is known about the tumor immune microenvironment (TIME) and potential novel CC immunotherapeutics. Therefore, it is critical to understand the local immune environment in the cervix and its role in preventing CC. This review explores the role of various immune cells in CC pathogenesis, its TIME, and the potential for successful immunotherapeutic treatments.

2. The Immune Environment of the Healthy Cervix

The cervix is a vital part of the female reproductive tract (FRT) that connects the uterus to the vagina and it is crucial in maintaining pregnancy by protecting against invading microbes [2]. It has two distinct parts: the endocervix, which leads to the uterus and contains glandular cells, and the ectocervix, the outer part of the cervix that opens into the vagina and comprises squamous cells. The transformation zone (TZ), where the endocervix and ectocervix meet, is the most common location for CC to originate from, and its position changes with age and number of pregnancies (Figure 1A). The FRT, similar to the gastrointestinal tract, relies on mucus and mucins for innate immune functions. Therefore, protecting against invading microbes is a crucial function of the cervix, particularly during pregnancy [4,5,6].
The endocervix is protected from pathogens and supports reproductive function through mucus-producing glands known as pseudoglands with goblet cells. On the other hand, the ectocervix, which has low mucus and is frequently exposed to external pathogens, is more vulnerable to microorganisms. This results in a unique immune environment [7,8,9]. Mucin 4 (MUC4), MUC5 (5A and B), and MUC6 are significant components of the endocervical mucus, critical innate immunity components found at mucosal surfaces, including the FRT [10,11,12]. The cervix also expresses various pattern recognition receptors (PRRs), including toll-like receptors (TLRs) such as TLR1 to TLR9 [13,14]. In addition, nucleotide-binding oligomerization domain (NOD) proteins, NOD1 and NOD2, are present in cervical epithelial cells (CECs) and are critical for inflammasome signaling to release proinflammatory IL-1α, IL-18, and IL-33. The cervix with its CECs also has many other humoral innate immune components, including complement proteins, different antimicrobial peptides (AMPs, including defensins), lysozyme, and lactoferrins, along with secretory antibodies (IgA and IgG) of the adaptive immune system. These components complement the mucus-associated innate immunity against invading pathogens, including HPV [15,16,17].
Epithelial cells, particularly CECs, serve as crucial innate immune cells that act as a physical barrier against pathogens in the upper FRT (uterus, fallopian tubes, and ovaries) to prevent ascending infections (as shown in Figure 1A) [15,18,19]. Specialized immune cells in the area also provide innate and adaptive immune responses [15]. CECs play an additional role in promoting the immunological functions of epithelial cervicovaginal Langerhans cells (LCs) and dendritic cells (DC) to fight against invading microbes/pathogens (as shown in Figure 1A). Keratinocytes in the ectocervix also act as innate immune cells, expressing various PRRs such as different TLRs, NOD-like receptors (NLRs), C-type lectin receptors (CLRs), absent in melanoma-2 (AIM-2), and stimulating interferon genes (STING), a downstream adaptor protein for the cyclic guanosine-monophosphate (GMP)-adenosine monophosphate (AMP) synthase (cGAS)/STING signaling pathway (as shown in Figure 1A) [20,21,22,23,24,25].
Cytokines released from activated keratinocytes are known as cytokinocytes. Syndecan-1, a transmembrane heparan sulfate proteoglycan (HSPG) that carries heparan-sulfate (HS) and chondroitin-sulfate glycosaminoglycans on its ectodomain, is also present in ectocervix keratinocytes [26,27]. Syndecan-1 regulates various cellular events, including migration, adhesion, proliferation, and growth, by acting as a coreceptor for different growth factors, cytokines, and chemokines [26]. Interferons (IFNs) secreted by cervical fibroblasts stimulate IFN-stimulated genes (ISGs), indicating cervical immune cell involvement in innate immunity by recognizing and responding to pathogens [28]. Cytotoxic T-lymphocytes (CTLs) in the cervix play a critical protective role and maintain their high levels despite menstrual status [28,29]. However, CD3+, CD4+, and CD8+ cell populations are significantly smaller in the endocervix than in the ectocervix (as shown in Figure 1A). Conversely, NK cell populations do not differ significantly between the endo- and ectocervix (as shown in Figure 1A) [30]. Therefore, even in a healthy cervix, immune cells (CECs, DCs, and T cells) have a dynamic composition (as shown in Figure 1A).
The population of immune cells varies between the cervix and the rest of the FRT [31]. Women who are healthy and do not have any reproductive tract infections or inflammation tend to have many antigen-presenting cells (APCs) and T cells in their cervical TZ. Leukocytes are more common in the upper reproductive tract than the cervix, and macrophages are less common in the cervix than in other tissues, comprising only 10% of all leukocytes in the reproductive tract [28,32,33]. During menstruation and childbirth, the cervix dilates, making the upper reproductive tract particularly vulnerable, and a flexible immune response is necessary [15]. The immune system is programmed to respond by recruiting macrophages to the cervix as it softens during labor [34,35]. Depletion of CD8+ T, B, and NK cells following menopause suggests some hormonal regulation on the cervical immune environment [15]. However, not all cervical immune cells are influenced by menstrual status. For example, CD8+ T cell activity decreases in the fallopian tubes and endometrium during the secretory phase of the menstrual cycle but remains unchanged in the cervix [36]. It is essential to understand the cervical immune landscape to comprehend cervical carcinogenesis.

3. CC Immunopathogenesis

CC is mainly caused by high-risk strains of HPV, such as HPV16 and HPV18. This correlation is more significant in women older than 35, as younger women tend to have shorter HPV infections [37]. The type of HPV strain also affects the development of cancerous or precancerous lesions [38]. HPV16 is the most common strain, followed by HPV18 [38,39]. Women with HPV16 infection are at a higher risk of developing CC and more severe lesions than women with HPV18 [40]. HPV16 is especially prevalent in Italian women [41] and Chinese patients with SCC [42], while HPV18 is more common in Iranian women [43]. Nevertheless, HPV infection dramatically increases women’s risk of CC or precancerous lesions.
The lifecycle of high-risk HPV is discussed elsewhere [44,45,46], but this section focuses on its immunopathogenesis and immune escape mechanisms. HPV infection suppresses the immune response and thereby increases the risk of CC through escaping cancer immunosurveillance and cancer immunoediting. Authors have discussed cancer immunosurveillance failure and cancer immunoediting elsewhere in detail [47]. For instance, HPV implies different strategies to escape immune surveillance for initiating and progressing the CC. For example, after infecting epithelial cells, HPV DNA integrates into gene-dense chromosomal regions responsible for transcriptional activities [48]. HPV can remain latent for years without clinical manifestation [49] and, during this time, it suppresses the host’s immune response. The epithelial-cell infection of HPV hijacks its innate immune-cell function and suppresses the proinflammatory immune response that is required to generate cell-mediated immunity (CMI) to clear the HPV infection (Figure 2) [50,51]. This characteristic of the HPV replicative cycle has been exquisitely shaped by its coevolution with the host [52] and enables HPV to avoid immune recognition and clearance.
HPV primarily avoids immune recognition during the initial infection phase by escaping the cytosolic cGAS/STING signaling-mediated recognition in epithelial cells and keratinocytes due to its unique vesicular trafficking pathway (Figure 2) [53]. Additionally, cervical keratinocytes with HPV16 are refractory to tumorigenic transformation by oncogenic H-Ras (Harvey rat sarcoma virus, a cellular GTPase that signals through mitogen-activated protein kinases (MAPK), phosphatidylinositol 3 kinase (PI3K), and Ral-GEF pathways expression (Figure 2) [54]. Along with the genetic integration of HPV DNA with the host cell DNA, the loss of the E2 episome (circular viral DNA that remains unintegrated and competent for viral transcription and replication during latency loss) is critical for cervical carcinogenesis [55,56].
During HPV infection of cervical keratinocytes, the loss of the E2 episome chronically stimulates antiviral genes. This stimulation is induced by the cGAS/STING signaling-dependent proliferation of type 1 IFNs and NF-κB-responsive genes, which promotes a chronic proinflammatory environment that supports tumor growth [55,57]. The HPV 16 and 18 E7 oncoprotein inhibits this signaling and the release of type 1 IFNs in CECs, keratinocytes, and fibroblasts (Figure 2) [57,58], while blocking cGAS/STING-dependent NF-κB-mediated immune responses. However, E7 does not affect IRF3 activation responsible for type 1 IFN generation in CECs (Figure 2) [59]. Chronic cGAS/STING-dependent type 1 IFN release can have a protumorigenic effect, as evidenced by the association of the rs311678 polymorphism in the cGAS gene with increased genetic susceptibility to HPV-induced precancerous cervical lesions [60]. E6 increases the synthesis and release of various cytokines in differentiating cervical keratinocytes, including IL-8, regulated upon activation, normal T cells expressed and secreted (RANTES or CCL5), macrophage inflammatory protein-1α (MIP-1α or CCL3), and interferon-γ-induced protein-10 (IP-10 or CXCL10) [57]. Keratinocytes also produce IL-10 to create a tumor-supportive chronic inflammatory environment (Figure 2) [61]. E6 and E7 further enhance this environment by inhibiting the release of transforming growth factor beta (TGF-β) from keratinocytes [62]. The HPV16 E7 oncoprotein can cause cervical keratinocytes to become more sensitive to apoptosis and release type 1 IFNs, leading to a chronic tumor-supportive inflammatory environment [63]. However, HPV16 E7 in CECs can prevent apoptosis and support uncontrolled growth and cancer development (Figure 2) [64]. HR-HPVs can evade phagocytosis by APCs and the antigen presentation to T and B cells [52], decreasing the chances of systemic immunity development. HPV can also suppress apoptosis and initiate squamous intraepithelial lesion development (Figure 2) [65]. In addition, p53 loss or mutation can suppress STING activation, which is critical to induce apoptosis of the infected cell and the release of type 1 IFNs for antiviral and antitumor immunity [66].
Patients with CC overexpress the regulator of G protein signaling 1 (RGS1) oncogene [67], which can inhibit proinflammatory immune-cell infiltration, reduce Th1 and cytotoxic CD8+ T-cell survival, and promote T-cell exhaustion, leading to an immunosuppressive environment [67,68,69,70]. RGS1 overexpression in CC correlates with increased HPV+ E6 in AC and cervical SCC, contributing to rapid cancer progression [67]. However, RGS1 knockdown can inhibit tumor growth, migration, and proliferation by promoting cancer cell apoptosis [67]. Therefore, targeting RGS1 has excellent potential for adjunct cancer immunotherapy for patients with CC. In addition, differential gene expression is associated with metabolic and hypoxic pathways and immune-cell activation and infiltration, altering CC risk and progression [71].
The E6 and E7 oncoproteins play a significant role in cancer progression caused by HPV. When absent, CC cells undergo apoptosis [72]. In addition, HPV16-E7 stimulates CCL5 and CCL6 chemokines, attracting mast cells to create an immunosuppressive microenvironment that supports tumor growth [73]. These viral oncoproteins contribute to six key characteristics of a cancerous environment, including resisting cell death, inducing angiogenesis, and triggering metastasis [74]. In addition, E6 and E7 inhibition induces aging among HPV+ CC cells and reactivates the tumor-suppressor gene p53 and other antiproliferative proteins [75]. Thus, E6 and E7 inhibitors are attractive therapeutic targets to explore.
LCs are potent myeloid innate immune cells in the skin and other epithelial environments, including stratified epithelia of the corneal, buccal, gingival, and genital mucosae [76,77,78]. HPV 16 virus-like particles (VLPs) called HPV16-L1 and L2 have been shown to avoid recognition by LCs and suppress their innate immune function to avoid the antiviral immune response (Figure 2) [79]. HPV 16 interacts with the annexin A2 hetero-tetramer (A2t) to infect the basal epithelial cells (BECs) and LCs. Blocking A2t with HPV16 L2 prevents HPV-mediated LC maturation suppression, as demonstrated by increased secretion of Th1-associated cytokines and surface expression of MHC class II on LC [79]. LCs in the cervix also serve as critical innate immune cells against HPV infection. They can present HPV antigens to induce an adaptive T-cell immune response when treated with different immunomodulators including poly I:C, 3M-002(TLR8 agonist) and resiquimod (TLR7/8 agonist) [80,81]. In addition, chimeric HPV VLPs (HPV cVLPs) can potentially activate LCs and DCs.
The activation of LCs and DCs is indicated by the upregulation of surface activation markers and an increase in the secretion of IL-12p70. This activation is critical for antiviral/antitumor NK and Th1 cells and increases IFN-γ [82,83,84]. The HPV-16 L1-VLPs recognition by DC surface antigens (human leukocyte antigen class 1, or HLA class I) and many cytokines/chemokines, particularly TNF-α, IL-6, and RANTES (regulated on activation normal T cell expressed and secreted), is mediated by DC-specific intercellular adhesion molecule-grabbing nonintegrin (DC-SIGN) [85]. HPV cVLPs have great potential for designing CC vaccines, but LC and DC stimulation with HPV without costimulation is insufficient to activate residential T cells [86]. Adjuvants added to HPV cVLPs can induce antiviral and antitumor T cell-mediated immunity against HR-HPVs, making it a novel immunotherapeutic approach for CC.
In CC, cellular metabolism plays a critical role in carcinogenic immunometabolic reprogramming. Patients with CIN and ICC due to HR-HPV infection show altered glucose transporter 1 (GLUT1), lactate dehydrogenase A (LDHA), and monocarboxylate transporter type 4 (MCT4, lactate transporter) [87]. LDHA is overexpressed in HPV16+ CIN patients, and GLUT1 expression is higher in CIN-stage I patients than in a control group, which further increases in CIN-II/III patients’ cancer cells. This increased expression indicates increased glucose utilization by glycolysis in CC cells, creating a glucose-deprived niche for immune cells. The overexpression of LDHA and MCT4 is more prevalent in later stages, indicating lactate accumulation in the TME or TIME (Figure 3) [87]. Lactate accumulation exerts immunosuppressive action by different mechanisms, including acidity and immune-cell apoptosis/death (Figure 3). Details of immunometabolic reprogramming responsible for the immunosuppressive TIME are discussed elsewhere by the authors [47].
An immunosuppressive TIME supports CC growth and metastasis. High levels of the immunosuppressive cytokines IL-10 and IL-13 are associated with cervical SCC (Figure 3). Increased levels of IFN-γ and IL-12p70 have antitumor action and are associated with decreased cervical SCC and grade incidence [88]. High IL-10 levels correlate with amplified arginase activity, decreasing L-arginine levels but increasing L-Arg metabolite levels, which is crucial to tumor growth [88,89,90,91]. HPV+ women, with either precancerous cervical lesions or CC, have elevated arginase levels, indicating persistent immunosuppression and a tumor-supportive immune environment [92]. Furthermore, indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO) are also critical players in CC immunopathogenesis [93,94]. TDO catabolizes tryptophan (Trp) to kynurenine (Kyn), a binding ligand for the aryl hydrocarbon receptor (AHR), which is involved in multiple bioregulatory processes contributing to CC progression [95]. Kyn metabolites are critical to generating an immunosuppressive microenvironment, including the generation of tolerogenic DCs (tDCs), which suppress potent antitumor T cell-mediated immunity (Figure 3). IFN-γ also promotes IDO expression. IFN-γ and Kyn induce autophagy in CC cells in vitro but this has not been replicated in vivo [96].
TDO expression in leukocytes surrounding intraepithelial or invasive CC lesions is critical in CC progression [93]. TDO is a critical prognostic oncolytic biomarker, as its overexpression correlates with poor overall and progression-free survival [97]. Mechanistically, TDO catabolizes Trp to Kyn, a binding ligand for the aryl hydrocarbon receptor (AHR), which is involved in multiple bioregulatory processes contributing to CC progression [98,99]. Details of the AHR pathway and its role in cancer progression are beyond the scope of this review but are extensively discussed elsewhere [100,101]. CAC has a lower TGF-β level and activity than CC SCC [102]. Hence, HPV escapes the host immune response (immunosurveillance) to establish the infection and grows inside basal/suprabasal keratinocytes without their transformation. However, its growth and multiplication in epithelial cells transform them into neoplastic cells and generate different oncoproteins. These oncoproteins also suppress antitumor immunity. The continuous growth of tumor cells develops a nutrient-depleted microenvironment to support an immunosuppressive TIME.

4. Factors Regulating CC Immunosuppressive TIME

Immunosuppressive TIME is key for tumor growth, progression, and metastasis [47]. The cancer and immune-cell metabolites and differentially released factors promote the development of the immunosuppressive TIME [47,103,104,105]. The CC immunosuppressive TIME is regulated by different regulating factors, including the HPV E6 and E7 proteins, as discussed earlier which helps in the maintenance of the malignant phenotype of infected cells and explains the absence of antigen loss in HPV-associated CC [106]. CC cells show increased aerobic glycolysis that causes lactate accumulation in the CC TIME, which blocks the antitumor activity of local and infiltrated immune cells, including the plasmacytoid DCs (pDCs) and central memory T cells [47,107].
Furthermore, tumor or immune-cell-derived regulating factors, including FoxP3 (forkhead box protein P3), CCL22/CCR4, OX40L/OX40 (tumor necrosis factor superfamily member 4/tumor necrosis factor receptor superfamily member 4), and SMAD3 (SMAD family member 3) also determine the maintenance of the immunosuppressive CC TIME (Figure 3) [108]. For example, FoxP3, CCL22, and CCR4 overexpression and SMAD3 downregulation in the CC support the immunosuppressive TIME. Furthermore, CXCR3 expression also determines the CC TIME characteristic, as a decreased CXCR3 expression is associated with low M1 macrophages, activated memory CD4+ T cells, and CD8+ T cells [109]. However, CD163 expressing M2 macrophages are elevated in the CC TIME, which suppress the programmed cell-death protein 1 (PD-1 or CD279)/programmed-death ligand 1 (PD-L1 or CD274) blockers efficacy [110,111]. It is noteworthy that CC patients with decreased CXCR3 levels die soon, as compared to patients with higher CXCR3 expression due to the lack of antitumor response of CXCR3-signaling CD8+ T cells in the TIME.
PD-1/PD-L1 overexpression (T cells, DCs, and macrophages) is well correlated with high-risk HPV infection and its progression to the CC with increased mortality by supporting the immunosuppressive TIME (Figure 3) [110,112,113,114,115,116,117]. Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4, another immune checkpoint) overexpression is also associated with CC immunosuppressive TIME (Figure 3) by increasing regulatory T cells’ (Tregs) and conventional T cells’ antitumor function by regulating CD28 signaling that impacts their interaction with CD80 (B7.1) and CD86 (B7.2) [118,119]. Increased CTLA4 levels also correlate with IL-1β expression CC as IL-1β increases the signal transduction of the CTLA4; therefore, targeting the IL-1β–CTLA4 axis may help to overcome the CC immunosuppressive TIME, as seen in colon cancer [118,120]. Cystatin 7 (CST7) in T cells is also downregulated in the CC patients which induces dysfunctional antitumor CD4+ and CD8+ T cell immunity [118,121]. These dysfunctional CD8+ T cells comprise the major T-cell population in the CC TIME. ERBB3 (Erb-B2 Receptor Tyrosine Kinase 3) or HER3 or human epidermal growth factor receptor 3 (EGF3) methylation in CC also supports immunosuppressive TIME by regulating different chemokines (CXCL9, CXCL5, CXCL13, CXCL11, CCL19, CCL18, CCL21, and CCL22) and tumor-immune lymphocyte (TIL) infiltration and expression of different immune checkpoints, including T-cell immunoreceptor with Ig and ITIM Domains (TIGIT), CTLA4, and lymphocyte activation gene 3 (LAG3) [122]. Thus, several regulatory factors support the development of immunosuppressive CC TIME, which is further maintained by the polarization of local and infiltrated immune cells to protumor immune cells.

5. Immune Cell Populations in the CC TIME

The immune response plays a crucial role in detecting and preventing the development of CC. However, factors such as age, prior or repeated human papillomavirus (HPV) infection, changes in the microbiota of the reproductive tract, and lifestyle choices can lead to immune dysregulation and increase the risk of CC [123,124,125,126]. In addition, the composition of immune cells also varies in precancerous lesions and stages of CC, highlighting the importance of the TIME in CC progression.
One of the critical components of the TIME is tumor-infiltrating lymphocytes (TILs), which are altered in CC and can contribute to tumor growth. Consistent with other cancers, TILs are altered in CC, thus contributing to tumor growth and advancement. Individuals with high immune expression have a more robust CC prognostic outlook than patients with less active CC TIME [71]. Notably, as many as 60% of TILs in tumors comprise tumor-specific T-cells with a high aptitude for tumor eradication [127]. In the following section, we will explore the involvement of different immune cells in the CC immunosuppressive TIME as understanding TIME plays a critical role in developing effective cancer immunotherapies [128,129].

5.1. Tumor-Associated Macrophages (TAMs)

Tumor-associated macrophages (TAMs) are another crucial component of the TIME, and their levels can influence cancer prognosis depending on their diversity as determined through single-cell omics [130,131]. For example, they help in neoangiogenesis which helps in tumor growth, survival, and metastasis by releasing angiogeninc factors, including vascular endothelial growth factor (VEGF) and creating a tumor-promoting immunosuppressive environment (Figure 3). There are two subgroups of macrophages, M1 and M2, with different roles in cancer progression [130,132]. M1 macrophages may be less detrimental to cancer prognosis, as they are more associated with phagocytosis and antitumor inflammation reactions, while M2 macrophages exhibit immunosuppressive, tumor-promoting activities by secreting different molecules, including IL-10, TGF-β, prostaglandin E2 (PGE2, due to the overactivation of cyclo-oxygenase II or COX-II enzyme) (Figure 3) [130,133]. These immunosuppressive molecules stimulate Th2 immune response and promote Treg polarization and function along with promoting the polarization and function of other immunosuppressive immune cells discussed later (Figure 3) [130] High-stage intraepithelial lesions are more likely to have M1 macrophages, while M2 macrophages are more common in tumors (Figure 1B) [134]. Elevated M2 levels in the TIME can create an immunosuppressive environment by supporting the generation and recruitment of myeloid-derived suppressor cells (MDSC) and regulatory T-cells (Tregs) generation and recruitment (Figure 1B and Figure 3) [130,135] while suppressing antitumor cytotoxic NK cells [136]. The increased CD163+M2 macrophages in the CC TIME are highly associated with overexpression of PD-L1 that supports immunosuppression through T-cell exhaustion and suppresses the PD-1/PD-L1 blockers’ efficacy, as discussed previously [137].
Understanding the CC TIME can help mitigate the detrimental effects of TAMs on prognosis. For instance, blocking the expression of transmembrane protein neuropilin-1 (NRP1) in M2 macrophages can prevent M2 polarization and recruitment [138]. Additionally, radiotherapy can repolarize tumor-promoting M2 macrophages to tumor-killing M1 macrophages [139]. Mitigating M2 prevalence in the TIME could be instrumental in stimulating immune responses to target CC [140]. Developing chimeric antigen receptor (CAR)-macrophages also has excellent potential in CC immunotherapy to overcome the immunosuppressive TIME, a significant hurdle in successful cancer clearance [141,142].

5.2. Neutrophils in the CC TIME

Neutrophils are immune cells that can either suppress or enhance tumor growth, depending on their function in the TIME [143,144,145]. In humans, neutrophils make up 50–70% of all circulating leukocytes, but this percentage may increase in tumors [146]. An increase in neutrophil count is associated with decreased overall survival for patients with CC. Overall, CC-patient survival decreases with the increase in absolute neutrophil count (≥6187/mm3) (Figure 1B) [147]. For example, an elevated CD66b+ tumor-associated neutrophil (TAN) count is also linked to shorter recurrence-free survival in CC due to increased neutrophil extracellular traps (NETs) [148], contributing to tumor proliferation and metastasis (Figure 3) [149]. The neutrophil-mediated IL-17 release in patients with squamous CC decreases their survival by creating a tumor-promoting TIME at early tumorigenesis [150].
The neutrophil-lymphocyte ratio (NLR) can be an independent prognostic factor in CC, with an NLR ≥ 3.6 indicating a poor overall response rate and survival [151]. NLR can also help predict the CC therapeutic response. For example, patients with an NLR of less than eight have a 57% chance of one-year survival following PD-1/PD-L1 inhibitors, while individuals with an NLR of eight or below have only a 27% chance of one-year survival [152]. A possible explanation of this trend is that neutrophils are associated with elevated cytokine secretions, thus enhancing tumor growth and metastasis [151]. Thus, an overaccumulation of tumor-supportive neutrophils further supports the immunosuppressive TIME. Therefore, therapeutic exploitation of TANs is critical in the successful tumor immunotherapy [153,154,155,156].

5.3. MDSCs in the CC TIME

Approaches targeting MDSCs in the CC TIME are critical to overcoming the immunosuppressive TIME and increasing the efficacy of existing immunotherapies [153,154,155,156]. Three types of MDSCs including monocytic, polymorphonuclear, and early-stage cells exert potent immunosuppressive action [157,158,159]. In addition, MDSCs secrete tumor-promoting arginase that generates tumor-promoting immunosuppressive polyamines in the CC TIME [157,160]. However, MDSCs activity in CC is governed by many aspects of the TIME, including immune and nonimmune cells and released factors. For example, growth factor granulocyte-colony stimulating factor (G-CSF) increases MDSCs’ number in CC (Figure 1B and Figure 3). Elevated G-CSF can induce tumor-related leukocytosis (TRL), a condition found in patients with advanced cancer [161]. Specifically, patients with CC with TRL are at greater risk for metastasis and are low responders to radiotherapy [162,163]. Given the connection and detrimental effect of MDSCs and G-CSF levels, future CC therapies should focus on alternative therapies to mitigate these levels. Novel immunotherapeutic approaches could focus on mitigating MDSCs and controlling G-CSF levels.
Furthermore, T cells also affect MDSCs’ function in TIME and vice-versa [164]. The combined activity of m-MDSCs and mucosal-associated invariant T (MAIT)-cells may be associated with CC progression (Figure 1B) [165]. All-trans retinoic acid (ATRA, a vitamin A-derivative) influences MDSCs maturation and eliminates their immunosuppressive activity. ATRA treatment decreases MDSC accumulation in BALB/C mice with CC and increases antitumor cytotoxic CD8+ T cells [166]. Combined ATRA and anti-PD-L1 therapies may be promising approaches to CC cancer immunotherapy, as this approach delays tumor growth and increases antitumor T-cells, IFN-γ, and TNF-α levels [166]. Thus, approaches targeting MDSCs in the CC TIME are critical to overcoming the immunosuppressive TIME and increasing the efficacy of existing immune checkpoint inhibitors (ICIs) and other immunotherapies.

5.4. MAIT Cells in the CC TIME

MAIT cells are unique T cells found in the body’s peripheral blood, liver, and mucosal surfaces, including the cervix [167]. They produce cytokines such as IFN-γ and IL-17, which are crucial in fighting pathogens such as bacteria, viruses, and fungi. They also regulate inflammatory responses and contribute to immune-mediated diseases [167,168,169,170]. MAIT cells are essential in different cancers and have potential in immunotherapy [171,172,173,174,175,176,177].
In patients with CC, there is a decrease in the number of MAIT cells in circulation, which is associated with poor progression-free survival [178]. The number of CD4-CD8-PD1+ MAIT (DN or double negative) cells in the peripheral circulation of patients with CC is directly related to disease severity [165]. The decrease in peripheral DN MAIT cells indicates that they might have migrated to the cancer tissue, further supporting the growth of the tumor [165]. Further investigation into the number and types of MAIT cells in CC biopsies and animal studies is necessary to understand this critical area of tumor immunology.
MAIT cells in the tumor microenvironment (TME) can either hinder or aid the antitumor activity of NK cells. While MAIT cell accumulation can contribute to tumor growth and spread, activating these cells can enhance their antitumor action by activating NK cells [179]. In cancer immunotherapy, inhibiting MHC class I-related protein 1 (MR-1) on cancer cells can be used to develop MAIT-cells-based treatments. The interaction with MR-1 expressed on tumor cells and TIME MAIT cells activate them to release IL-17A, suppressing cytotoxic T and NK cells. Thus, blocking MR-1 on cancer cells may help design MAIT-cells-based cancer immunotherapy. Additionally, combining a synthetic riboflavin synthesis pathway-derived antigen 5-OP-RU [5-(2-oxopropylideneamino)-6-D-ribitylaminouracil] and the CpG (a TLR9 agonist) can boost the antitumor immune response of TIME MAIT cells as indicated by the increased levels of CD69 expression, pronounced effector memory phenotype, and upregulation of effector molecules, including IFN-γ, granzyme B (GrB), and perforin [180]. Interestingly, 5-OP-RU and TLR9 agonist combination work independently of MHC class I related-1 molecule (MR1) expression in tumor cells. Reprogramming and redifferentiating TIME MAIT cells are also promising approaches for cancer immunotherapy. Therefore, exploring and targeting TIME MAIT cells in CC is a new and innovative cellular immunotherapy strategy.

5.5. Mast Cells in the CC TIME

Mast cells are critical innate immune cells with different immune and inflammation regulatory functions [181,182]. Initially, they were only associated with allergic reactions/diseases. However, recent advancements in immunology have established them as potent immunoregulatory cells that perform various immune functions, including maintaining immune homeostasis [181,182,183,184]. Mast cells play a critical role in the TIME of many cancers and their effect on promoting or inhibiting tumors depends on the type and stage of cancer [185,186]. Mast cells (tryptase-positive and tryptase/chymase-positive) are also found in the normal human cervix, and their number increases in benign inflammatory conditions [187,188,189]. A recent study reported a widespread distribution of mast cells in CC tissues and patients with low mast-cell density in their TIME had better overall survival rates [190]. Mast cells promote tumor growth by supporting neoangiogenesis and creating an immunosuppressive TIME. Specific mast-cell mediators, including histamine, TNF-α, and cannabinoids, also contribute to CC cell invasion and metastasis (Figure 3) [183,184,185,186,191]. In addition, mast-cell infiltration in the TIME increases the tumor’s resistance to anti-PD-1 immune checkpoint blockers [192,193]. Hence, targeting mast cells and their mediators in the CC TIME may inhibit CC growth and metastasis by inhibiting neoangiogenesis and immunosuppressive events that support CC metastasis.

5.6. Eosinophils in the CC TIME

Eosinophils are critical innate immune cells that are present in low numbers in blood but are present in higher numbers at mucosal surfaces [194,195,196]. They play a critical role in antimicrobial immunity, allergies, and tumor immunity [197,198,199]. For example, eosinophils work with DCs and T cells to produce inflammatory and adaptive immune responses [200]. A patient’s eosinophil count can predict the immune response in cervical SCC, and high levels of eosinophils can result in poor survival (Figure 1B) [201,202]. Hypoxic conditions influence the function of EOs during cancer progression. The prevalence of eosinophils increases with CC progression, and hypoxic conditions also influence their function [193,203]. Thymic stromal lymphopoietin (TSLP) stimulates CC growth [204] and regulates eosinophil activity in the hypoxic CC TME. The increased TSLP upregulates CCL17 production [193,204], which over-recruits eosinophils (Figure 3). In addition, TSLP promotes CC progression by promoting the immunosuppressive Th2 immune response (Figure 3). Eosinophils can create an immunosuppressive TIME by releasing Th2 cytokines and suppressing NK and T-cell functions in the CC (Figure 3) [205,206].

5.7. DCs in the CC TIME

DCs are an essential immune system component that can either enhance or suppress tumor response [200,201,202,203,204,205,206,207]. The different types of DCs, including conventional DCs (cDC1s and cDC2s), plasmacytoid DCs (pDCs), and mature DCs, express varying levels of costimulatory molecules and immune checkpoints, depending on the type and stage of the tumor [208,209]. For instance, cDC1s typically do not express PD-L1 and immunoglobulin-like transcript 2 (ILT2) under normal conditions. However, during tumor progression, they may express high T-cell immunoglobulin and mucin-domain containing-3 (TIM-3), a unique immune-checkpoint repertoire [209]. TIM-3 interacts with galectin-9, a C-type lectin, to stimulate antitumor function in innate immune cells, such as DCs, NK cells, and macrophages, by activating proinflammatory signaling pathways, including PI3K-mammalian target of rapamycin (mTOR) and hypoxia-inducible factor-1 (HIF-1) signaling without inducing apoptosis [203,204,205,206,207,208].
The decrease of galectin-9 in CC (CIN and SCC) patients increases its severity, whereas its increase is associated with a better prognosis regarding the overall survival [209,210]. However, severe CC cases (advanced stage IV) have an increased systemic galectin-9 level, indicating that circulating galectin-9 via TIM-3 interaction in systemic Th1 and CD8+ T cells induces their apoptosis and impairs their infiltration in the CC TME [211]. Furthermore, in HPV-associated patients with CC, circulating CD4+ and CD8+ T cells overexpress TIM-3, supporting that increased circulating galectin-9, including monocyte-specific galectin-9 in patients with CC, induces Th1 and CD8+ T cell apoptosis to suppress systemic T-cell-dependent antitumor immunity [212]. Thus, increased circulating TIM-3 expression of T cells and galectin-9 in patients with CC is associated with poor CC prognosis. In addition, increased TIM-3 expression in Tregs via galectin-9 interaction increases the immunosuppressive function (IL-10 and TGF-β release) in patients with CC [212,213]. Notably, the systemic galectin-9 level is independent of the local CC TME. Hence, local CC TME galectin-9 decreases the CC severity and improves overall patient survival, whereas systemic galectin-9 is associated with increased CC severity. Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM-1) is an adhesion molecule that also serves as a heterophilic ligand for TIM-3 expressed on T cells [214]. In severe or high-grade squamous intraepithelial lesions (SIL) of patients with CC, the increased CEACAM-1 and TIM-3 interaction further suppresses the antitumor activity of CD4+ and CD8+ T cells [215]. Furthermore, CC cells overexpress HMGB1 and serve as a prognostic indicator and a potential biomarker, suppressing antitumor T-cell function via interacting with TIM-3 [216,217,218,219,220].
cDC1s are critical for antitumor cytotoxic T cells and decrease Tregs via secreting the cGAS/STING signaling pathway-dependent type 1 IFNs [221]. The cDC1s’ decrease in the CAC TIME is associated with poor patient survival due to impaired T-cell-mediated antitumor immunity (Figure 1B and Figure 3) [222]. Although cDC1s highly express TIM-3, their decrease in severe CC cases indicates that they could not exert their antitumor action. Therefore, it should be interesting to investigate whether they die or transform to tumor-supportive tolerogenic DCs in the immunosuppressive CC TIME. The increased HMGB1 level in the CC TIME is associated with promoting pDCs to tDCs that further supports immunosuppressive TIME for the CC progression (Figure 2 and Figure 3) [223]. Furthermore, low cDC1 chemo-attractive chemokines in the CC TIME support the immunosuppressive niche. For example, NK cells in the TIME support cDC1 infiltration by releasing XCL1 and CCL5, and NK cells lose this function in the presence of PGE2 [224]. Overexpressed PGE2 in the CC TIME suppresses NK cell-mediated release of cDC1 chemokines to dysregulate the NK cell–cDC1–chemokine axis [225,226]. Thus, HMGB1 and PGE2 in the CC TIME suppress antitumor T cell, NK cell, and DC function, supporting CC growth and metastasis (Figure 3).
HPV-VLP vaccination stimulates DC and NK cell crosstalk to exert the antitumor activity in patients with CC, as indicated by the CD69 (an activation marker) and HLA-DR upregulation on DCs and increased NKCC and IFN-γ release [227,228,229]. DC-derived exosome vaccines also induce antitumor cytotoxic CD8+ T cell activity, proliferation, and IFN-γ secretion [230]. Immunotherapeutics work effectively under the right immunological conditions. For example, the synthetic dsRNA viral analog Poly I: C (polyinosinic: polycytidylic acid) vaccine is a promising CC vaccine. Poly I: C vaccine induces receptor-interacting protein kinase 3 (RIPK3) signaling for its direct cytotoxicity on tumor cells. The CC-cell necroptosis induces IL-1α release, which activates DC-mediated IL-12 production, critical for an antitumor immune response [231]. Thus, increasing cDC1s in CAC patients will increase their survival via increasing antitumor immunity. Further studies in this direction are critical to designing DC-specific vaccines for CC.

5.8. NK Cells in the CC TIME

NK cells are potent antitumor innate immune cells categorized as type 1 innate lymphoid cells (ILCs) [210]. Their role in antitumor immunity, including uterine cancer, has been discussed elsewhere [211,212]. However, in CC, the number and function of NK cells are reduced due to various cellular processes (Figure 1B). For example, CD3+CD56+ NK cell infiltration increases at early CC stages, which decreases as cancer progresses to advanced stages due to higher TGF-β1 in the tumor. TGF-β1 inhibits natural-killer group 2D (NKG2D), CD16, and Ki67 receptor function [213]. The decreased NK cell number in CC TIME is associated with HLA-I downregulation, potentially due to upregulated immunosuppressive cytokines, including IL-10, IL-13, and TGF-β [214], which inhibit NK-cell function. Patients with CC have HLA-E (a major histocompatibility (MHC) class I molecules involved in the NK-cell recognition pathway) overexpression, which is not well-associated with the prognostic outcome, potentially due to a high volume of exhausted or apoptotic CD8+ T cells [215]. However, elevated HLA-E expression in patients with CAC improves survival [216]. The decreased NK-cell number in the CC TIME further supports the decrease in potent antitumor cDC1s in patients with advanced CC. Therefore, the strategies to design NK-cell-based immunotherapies to target CC will be an exciting area to explore.

5.9. T Cells in the CC TIME

The type and concentration of T-cells in a person’s immune system can provide insight into their immune response and prognosis for CC. T cells infiltrate CC tumors, but the CD4+: CD8+ differs from that in the peripheral blood [217] and lower CD4/CD8 ratios are associated with faster tumor growth and lymph-node metastasis [218]. For example, healthy women have a CD4/CD8 ratio of 1.42 [219] but this number decreases to 0.6 and 1.17 in women with fatal and nonfatal CC, respectively [220]. These trends can be explained mechanistically, as CD4+ T cells activate the cytotoxic CD8+ T cells, and Tregs accumulate near advanced tumors, inhibiting antitumor immune activity. Th1 cells’ number and function also alter CC carcinogenesis (Figure 1B). For example, Th1 levels increase from low to high-grade squamous intraepithelial lesions but deplete from high-grade squamous intraepithelial lesions to SCC. In contrast, Th2 levels deplete from low- to high-grade squamous intraepithelial lesions [221]. Th2 and Th17 populations increase, and Th1 levels are depleted in CIN and CC, which supports that a shift in these cell populations starts prior to CC formation and, thus, contributes to CIN progressing into CC (Figure 1B) [222]. Th1 dominance is critical to antitumor immunity and contributes to immune memory, forming tumor-specific cytotoxic T-lymphocytes (CTLs) [223], further suggesting that Th1 depletion is crucial to CIN progression into CC.
HPV infections preceding CC development contribute to T-cell alterations. For example, CD4+ T-cells specific to HPV+ patients with CC can suppress T-cell proliferation and alter their function [223]. The ratio of cell types changes not only with cancer status but also with HPV status. For example, CD8+ T cells are more prevalent than CD4+ in the epithelial layer of an HPV+ normal cervix, but this becomes less prominent with an increasing CIN grade [225]. The CD4/CD8 ratio, as well as the quantity of CD4+ T cells, are indicative of CC survival in HPV+ individuals. Overall, a lower number of Tregs is detrimental to five-year survival but, more specifically, individuals with lower CD4/CD8 ratios have higher mortality rates than those with higher ratios [220]. Women undergoing neoadjuvant chemotherapy have more remarkable survival if they have higher CD4/CD8 ratios before their third round of treatment [226]. Furthermore, neoadjuvant chemotherapy increases CD4, CD8, CD20, and CD56 signals, most prominently in good responders, indicating the activation of antitumor Th1, cytotoxic T, B, and NK cells [227]. Therefore, the immunoactive TIME in good responders is crucial to support locoregional stimulation of antitumor immunity during neoadjuvant chemotherapy. Hence, neoadjuvant chemotherapy can be combined with ICIs in patients with CC to stimulate antitumor immunity.
Specific gene-expression profiles and ligands can significantly impact Tregs within the CC TIME. For example, Foxp3 and V-domain immunoglobulin suppressor of T-cell activation (VISTA) significantly correlate with CC prognosis, exhibiting higher expression in CC than in CIN or chronic cervicitis. Specifically, patients with double-negative (Foxp3 and VISTA) tumors show the best prognosis, while double-positive patients show the worst prognosis [228]. Foxp3 levels are also higher in patients with lymph node metastasis than those without metastasis [229]. Foxp3 levels are also higher in patients with CC with lymph-node metastasis than those without metastasis [229]. The increased FoxP3 expression in Th1 cells to transform them to Tregs occurs due to the intracellular STING activation in these T cells [230]. The intrinsic STING activation in T cells induces TANK binding kinase-1-interferon regulatory factor (TBK1-IRF3)-mediated mothers against decapentaplegic homolog 3 or SMAD3 and signal transducer and activator of transcription 5 (STAT5) phosphorylation independent of IFN-β to induce FoxP3 activation and their transformation to Tregs. In CC TIME, tumor-derived exosomes with TGF-β, cGAS, and 2′-3′-cGAMP activate STING signaling in tumor-infiltrated T cells to promote induced-Treg (iTreg) expansion [230].
Understanding T-cell-specific TIL activity changes is critical to designing better T-cell-based immunotherapeutics specific to CC type. Patients with CC have better effector T-cell infiltration than adenocarcinoma patients, with elevated CD45+ and CD3+ levels, Tregs, and PD-1 and TIM-3 immune checkpoints. These changes are prognostically significant and may indicate immunotherapeutic responses, as increases in CD3+ densities can decrease the death and relapse risk [231]. Therefore, immunotherapeutics that work well for SCC may not work as well in adenocarcinoma. Nevertheless, tumor-specific T cells are ideal candidates for personalized, adaptive immunotherapy. TILs from individual patients are primed for specific tumors and are immediately ready to return after infusion [127].
Under conditions that support oxidative phosphorylation (OXPHOS), Th17 cells have increased persistence and can decrease tumor growth in vivo [232]. In squamous CC TIME, an increased presence of Th17 cells has been associated with improved patient survival due to their anticancer effects [150,233]. The decreased presence of galectin-9 in the CC TIME promotes the development of Th17 cells, as galectin-9 interaction with TIM-3 induces apoptosis in mature Th17 cells [234]. However, Th17 cells in CAC are detrimental to the patient and can increase tumor growth and severity and contribute to CAC relapse after tumor removal [235]. The infiltration of Th17 cells in the CC TIME is facilitated by CCL20, which binds to overexpressed CCR6 [236]. Therefore, it would be interesting to explore the immunometabolic reprogramming of Th17 cells that govern their anti- and protumor functions in squamous and adenocarcinoma patients with CC to develop immunometabolism-based Th17 cell-directed immunotherapies.

5.10. B Cells in the CC TIME

B-cells play a crucial role in regulating the immune system by producing antibodies and releasing cytokines [237]. Studies on mice have shown that reducing the B-cell count can boost the body’s antitumor response by lowering IL-10 production and increasing IFN-γ levels from CD8+ T-cells and NK cells [238]. However, a subset of B-cells called Bregs can hinder cytokine secretion and counteract the antitumor response of other immune cells [239]. Understanding the role of B-cells in cancer immunity is critical, especially for HPV-associated cancers, which have shown conflicting results in human patients. For example, B-cells exert a vital antitumor role in HPV+ patients with CC [240]. Therefore, a deeper understanding of a patient’s cancer type is required in treatment.
Researchers are exploring the potential of B-cell-targeted immunotherapy in cancer treatment. For instance, PD-1 blockade and radiotherapy have proven effective in increasing memory B-cells, antigen-specific B-cells, and plasma cells in HPV-associated cancers [240]. However, Bregs have been found to inhibit CD8+ T-cell cytotoxicity in CC, leading to lower prognostic outcomes in individuals with low CD4/CD8 ratios [241]. There is still much to learn about the role of B-cells and Bregs in CC and their modulation of immunotherapy.

6. Targeting the CC TIME

Currently, different preventive and therapeutic approaches (which also have severe adverse reactions) are available for CC (Table 1), but we do not have effective immunotherapeutics to treat or target CC [242,243,244,245]. Targeting the TIME is a promising approach for cancer-specific immunotherapy, including CC. Researchers are studying the immunoregulatory factors of TIME to develop effective CC immunotherapies. For example, treating cervical epithelial cells with poly (dA:dT, a synthetic dsDNA analog) has shown potential in activating antiviral immunity and increasing CD8+ T-cell and DC populations to clear the tumor. This treatment induces the expression of different IFNs and associated IFN-stimulate genes (ISGs, including ISG-15, ISG-56), 2′-5′-oligoadenylate synthetase 1 (OAS1), OAS2, myxovirus resistance protein A (MxA), MxB, virus inhibitory protein, endoplasmic reticulum-associated, IFN-inducible (VIPERIN), and guanylate-binding protein 1 (GBP5)-dependent antiviral immunity by the activating retinoic acid-inducible gene I (RIG-1)-like receptor (RLR) signaling pathway [246,247]. This experimental approach has great potential to target HR-HPV-associated CC. CEC-specific cGAS/STING signaling activation by ADU-S100 (S100) promotes its antiviral and antitumor activity by acutely releasing type 1 IFNs and NF-κB-dependent proinflammatory cytokines, such as IL-6 and TNF-α [248]. In addition, ADU-S100 increases the cytotoxic CD8+ T cell and CD103+DC population to clear the tumor in vivo [248,249]. CD103+DCs also increase the efficacy of ICIs in TIME [250,251]. Therefore, HR-HPV targeted therapeutic vaccines could prove advantageous for CC. For example, NK cell-based therapy against the upregulation of HPV-VLPs in CC could be an exciting avenue to explore, as they appear to stimulate FRT NK cells [252]. Thus, HPV-targeted therapeutic vaccines and NK cell-based therapy against HPV-VLPs upregulation in CC are exciting avenues to explore.
Numerous ICIs are currently undergoing clinical trials, and some have shown promising results in combination with existing treatments [263,264,265,266,267,268,269,270,271,272,273,274,275,276,277,278,279,280,281,282]. For example, pembrolizumab increases the efficacy of existing chemotherapies in treating PD-L1-positive chemotherapy-resistant metastatic CC [264]. Cotreatment with pembrolizumab and the GX-188E vaccine is safe for HR-HPV infections and associated advanced CC [265]. Furthermore, HPV nanovaccine combination with laser therapy inhibits the CC progression by activating T-cells and inducing DC maturation [266]. However, translating experimental findings to humans requires a better understanding of the TIME in CC. For instance, radiotherapy may decrease the immune-cell population and their cytotoxic potential while increasing PD-1 capacity in CD4+ T cells. This trial has also uncovered further immune dysregulation due to elevated monocyte and MDSC levels [267], which can be detrimental to cancer immunotherapy.
Adoptive cell therapies (ACTs, including tumor-infiltrating T lymphocytes (TILs) and CAR-T cells) comprise a form of cancer immunotherapy, including ICIs and vaccines [268,269,270]. The details of engineered T cells with a potential to use in cancer immunotherapy have been discussed elsewhere [271]. Autologous TILs are ex vivo expanded tumor infiltrating T cells rescued from the tumor tissue and are transplanted back to the patient following lymphodepletion that has been mostly evaluated in metastatic skin melanoma and recurrent/metastatic CC of the cervix successfully as compared to other solid cancers [272,273]. Hence, autologous TILs (LN-145) offer CC patients a safe and viable therapeutic approach warranting further investigations [273]. TILs and their use in different solid tumors are discussed in detail elsewhere [274,275,276]. Further advances in cancer immunology have led to the development of CAR-T cells’-based cancer immunotherapies. CAR-T cell therapies have shown effectiveness against certain types of cancer, such as leukemia, but have limited efficacy against solid tumors [277]. However, researchers are currently investigating modified therapies, such as Mesothelin (MESO) CAR-T cell therapy, which has shown promising results in detecting MESO in SiHa cell lines. This approach has the potential to increase antitumor cytokine production and improve outcomes [278].
One challenge to CAR-T cell therapy in solid tumors is immune checkpoints, including the PD-1–PD-L1 axis, which can decrease efficacy [277]. However, the genetically modified CAR-T-PD1 approach has shown increased efficacy in experimental studies with DCs. This approach increases antitumor cytotoxic activity, suppresses tumor growth, and elevates IL-2, IFN-γ, and TNF-α secretions, ultimately improving survival [277]. Another approach under investigation is CAR-T cell therapy using NKG2D, which has shown excellent tumor clearance with low toxicity in CC cells [279]. These studies suggest CAR-T cell therapy can effectively treat CC when engineered to consider the unique CC TIME. Furthermore, T-cell receptor (TCR)-engineered/modified effector T (TCR-T) cells are another class of adoptive T-cell therapy, which have naturally occurring or minimally modified TCRs to develop T-cell-based immunotherapy for cancers [280]. These TCR-T cells recognize tumor-specific epitopes presented by major-histocompatibility complex (MHC) molecules expressed on cancer cells which gives them the advantage of potentially broader application. This is because of the presence of larger tumor-specific sequences within a cell and presented in the MHC than tumor-specific proteins on the surface [280]. TCR-T cells may have a higher potency than CAR-T cells for solid cancers, including CC, due to the MHC presentation of intracellular antigens specifically present in cancer cells. The details of TCR-T for solid tumors are discussed elsewhere [280,281]. Additionally, adeno-associated virus (AAV)-based gene therapy, including the delivery of oncolytic agents/drugs, has shown potential for personalized medicine or immunotherapy based on the HR-HPV type and patient genetic and immune status [282].

7. Future Perspective and Conclusions

Over the past 10–20 years, tumor immunology has made significant progress thanks to advancements in immunology, which have helped understand the growth, development, and spread of cancer cells. This progress has led to the discovery of ICIs as cancer immunotherapy by James P. Allison and Tasuku Honjo, which was awarded the Nobel Prize in Physiology and Medicine in 2018. In addition, researchers have identified that HR-HPVs are primarily responsible for CC in humans [283,284,285]. Understanding the immunosuppressive environment of CC is crucial for developing novel immunotherapies to advance treatment. A thorough understanding of the genetics and immune cells involved in CC can help predict therapeutic responses and allow for patient-specific approaches. Various immune genes have been used to predict changes in the immune microenvironment, which serve as indicators for immunotherapeutic response and survival [286]. For example, patients with high levels of CD8+ T cells in their tissues have shown more robust immunotherapy response rates [287]. Different immune cells, including LCs and MAIT cells, have great potential for developing cancer-specific immunotherapy [177,288,289]. In addition, HR-HPV CCs expressing viral L1 protein (major capsid protein) can be targeted by effective L1/tumor-specific CD4+ and CD8+ T cells or combined E7/L1 DC-based vaccines [290]. Furthermore, galectin-9 modifying/targeting strategies also have an excellent potential for modulating immunosuppressive CC TIME and developing novel immunotherapies [291,292,293]. Tumor-resident Mast cells are also emerging as novel immunotherapeutic targets for targeting the immunosuppressive TIME, which can be implied in patients with severe CC [294,295]. In conclusion, CC is a growing concern worldwide and it is crucial to understand its environment for future personalized immunotherapeutics to advance and to improve currently available therapies.

Author Contributions

Conceptualization, V.K.; Writing, review, and editing, V.K.; Writing first draft, C.B.; Final editing and proofreading, J.H.S.IV. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AAVAdeno-associated virus
ACAdenocarcinoma
ACTSAdoptive T-cell therapies
AHRAryl hydrocarbon receptor
AIFApoptosis inducible factor
AIM-2Absent in melanoma-2
AMPAntimicrobial peptides
APCAntigen-presenting cell
BECsBasal epithelial cells
CACCervical adenocarcinoma
CAR-T cellChimeric antigen receptor T cell
CC Cervical cancer
CECsCervical epithelial cells
cGAMPCyclic GMP-AMP
cGAScGMP-AMP synthase
CINCervical intraepithelial neoplasia
CLRC-type lectin receptors
CMICell-mediated immunity
CTLCytotoxic T lymphocyte
DNAM-1DNAX accessory molecule-1
DGEDifferential gene expression
FoxP3Forkhead box protein P3
FRTFemale reproductive tract
HIFHypoxia-inducible factor
HR-HPVHigh-risk HPV
hTERTHuman telomeres reverse transcriptase
ICIsImmune checkpoint inhibitors
IDOIndolamine dioxygenase
LCsLangerhans cells
MAITMucosal-associated invariant T
MIPMacrophage inflammatory protein
MUCMucin
NETNeutrophil extracellular trap
NKCCNK-cell-mediated toxicity
NKG2DNatural killer group 2D
NLRNeutrophil lymphocyte ratio
NODNucleotide-binding oligomerization domain
PRRPattern-recognition receptor
RGSRegulator of G protein signaling
RICKRIP-like interacting CLARP kinase
SCCSquamous cell carcinoma
SILSquamous intraepithelial lesion
STINGStimulating interferon genes
TCRT-cell receptor
TCR-T cellsTCR-engineered/modified effector T cells
TDOTryptophan dioxygenase
TIMETumor immune microenvironment
TMETumor microenvironment
TGF-βTransforming growth factor beta
TSLPThymic stromal lymphopoietin
TZTransformational Zone
VISTAV-domain immunoglobulin suppressor of T cell activation

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Brey, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. Tantengco, O.A.G.; Menon, R. Contractile function of the cervix plays a role in normal and pathological pregnancy and parturition. Med. Hypotheses 2020, 145, 110336. [Google Scholar] [CrossRef] [PubMed]
  3. Francoeur, A.A.; Liao, C.-I.; Casear, M.A.; Chan, A.; Kapp, D.S.; Cohen, J.G.; Salani, R.; Chan, J.K. The increasing incidence of stage IV cervical cancer in the USA: What factors are related? Int. J. Gynecol. Cancer 2022, 33, 136. [Google Scholar] [CrossRef] [PubMed]
  4. Gustafsson, J.K.; Johansson, M.E.V. The role of goblet cells and mucus in intestinal homeostasis. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 785–803. [Google Scholar] [CrossRef]
  5. Hasnain, S.Z.; Gallagher, A.L.; Grencis, R.K.; Thornton, D.J. A new role for mucins in immunity: Insights from gastrointestinal nematode infection. Int. J. Biochem. Cell Biol. 2013, 45, 364–374. [Google Scholar] [CrossRef] [PubMed]
  6. Johansson, M.E.; Hansson, G.C. Immunological aspects of intestinal mucus and mucins. Nat. Rev. Immunol. 2016, 16, 639–649. [Google Scholar] [CrossRef]
  7. Lee, S.K.; Kim, C.J.; Kim, D.-J.; Kang, J.-H. Immune Cells in the Female Reproductive Tract. Immune Netw. 2015, 15, 16–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Han, L.; Taub, R.; Jensen, J.T. Cervical mucus and contraception: What we know and what we don’t. Contraception 2017, 96, 310–321. [Google Scholar] [CrossRef]
  9. Gipson, I.K. Mucins of the human endocervix. Front. Biosci. 2001, 6, D1245–D1255. [Google Scholar] [CrossRef]
  10. Gipson, I.K.; Spurr-Michaud, S.; Moccia, R.; Zhan, Q.; Toribara, N.; Ho, S.B.; Gargiulo, A.R.; Hill, J.A., 3rd. MUC4 and MUC5B transcripts are the prevalent mucin messenger ribonucleic acids of the human endocervix. Biol. Reprod. 1999, 60, 58–64. [Google Scholar] [CrossRef] [Green Version]
  11. Linden, S.K.; Sutton, P.; Karlsson, N.G.; Korolik, V.; McGuckin, M.A. Mucins in the mucosal barrier to infection. Mucosal Immunol. 2008, 1, 183–197. [Google Scholar] [CrossRef] [Green Version]
  12. Sheng, Y.H.; Hasnain, S.Z. Mucus and Mucins: The Underappreciated Host Defence System. Front. Cell. Infect. Microbiol. 2022, 12, 856962. [Google Scholar] [CrossRef]
  13. Nasu, K.; Narahara, H. Pattern Recognition via the Toll-Like Receptor System in the Human Female Genital Tract. Mediat. Inflamm. 2010, 2010, 976024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Hart, K.M.; Murphy, A.J.; Barrett, K.T.; Wira, C.R.; Guyre, P.M.; Pioli, P.A. Functional expression of pattern recognition receptors in tissues of the human female reproductive tract. J. Reprod. Immunol. 2009, 80, 33–40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. De Tomasi, J.B.; Opata, M.M.; Mowa, C.N. Immunity in the Cervix: Interphase between Immune and Cervical Epithelial Cells. J. Immunol. Res. 2019, 2019, 7693183. [Google Scholar] [CrossRef] [Green Version]
  16. Farage, M.A.; Miller, K.W.; Gerberick, G.F.; Saito, F.; Ledger, W.; Witkin, S. Innate immunity in the lower female mucosal tract. J. Steroids Horm. Sci. 2011, 2, 106. [Google Scholar] [CrossRef]
  17. Quayle, A.J. The innate and early immune response to pathogen challenge in the female genital tract and the pivotal role of epithelial cells. J. Reprod. Immunol. 2002, 57, 61–79. [Google Scholar] [CrossRef] [PubMed]
  18. Fichorova, R.N.; Anderson, D.J. Differential expression of immunobiological mediators by immortalized human cervical and vaginal epithelial cells. Biol. Reprod. 1999, 60, 508–514. [Google Scholar] [CrossRef] [PubMed]
  19. Wira, C.R.; Grant-Tschudy, K.S.; Crane-Godreau, M.A. Epithelial cells in the female reproductive tract: A central role as sentinels of immune protection. Am. J. Reprod. Immunol. 2005, 53, 65–76. [Google Scholar] [CrossRef] [PubMed]
  20. Inki, P. Expression of syndecan-1 in female reproductive tract tissues and cultured keratinocytes. Mol. Hum. Reprod. 1997, 3, 299–305. [Google Scholar] [CrossRef] [Green Version]
  21. Gallo, R.L.; Nakatsuji, T. Microbial Symbiosis with the Innate Immune Defense System of the Skin. J. Investig. Dermatol. 2011, 131, 1974–1980. [Google Scholar] [CrossRef] [Green Version]
  22. Nelson, A.M.; Reddy, S.K.; Ratliff, T.S.; Hossain, M.Z.; Katseff, A.S.; Zhu, A.S.; Chang, E.; Resnik, S.R.; Page, C.; Kim, D.; et al. dsRNA Released by Tissue Damage Activates TLR3 to Drive Skin Regeneration. Cell Stem Cell 2015, 17, 139–151. [Google Scholar] [CrossRef] [Green Version]
  23. Wang, J.-N.; Li, M. The Immune Function of Keratinocytes in Anti-Pathogen Infection in the Skin. Int. J. Dermatol. Venereol. 2020, 3, 231–238. [Google Scholar] [CrossRef]
  24. Chieosilapatham, P.; Kiatsurayanon, C.; Umehara, Y.; Trujillo-Paez, J.V.; Peng, G.; Yue, H.; Nguyen, L.T.H.; Niyonsaba, F. Keratinocytes: Innate immune cells in atopic dermatitis. Clin. Exp. Immunol. 2021, 204, 296–309. [Google Scholar] [CrossRef] [PubMed]
  25. Jiang, Y.; Tsoi, L.C.; Billi, A.C.; Ward, N.L.; Harms, P.W.; Zeng, C.; Maverakis, E.; Kahlenberg, J.M.; Gudjonsson, J.E. Cytokinocytes: The diverse contribution of keratinocytes to immune responses in skin. JCI Insight 2020, 5, e142067. [Google Scholar] [CrossRef]
  26. Bernfield, M.; Götte, M.; Park, P.W.; Reizes, O.; Fitzgerald, M.L.; Lincecum, J.; Zako, M. Functions of cell surface heparan sulfate proteoglycans. Annu. Rev. Biochem. 1999, 68, 729–777. [Google Scholar] [CrossRef]
  27. Szatmári, T.; Mundt, F.; Kumar-Singh, A.; Möbus, L.; Ötvös, R.; Hjerpe, A.; Dobra, K. Molecular targets and signaling pathways regulated by nuclear translocation of syndecan-1. BMC Cell Biol. 2017, 18, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Rodriguez Garcia, M.; Patel, M.V.; Shen, Z.; Fahey, J.V.; Biswas, N.; Mestecky, J.; Wira, C.R. Chapter 108—Mucosal Immunity in the Human Female Reproductive Tract. In Mucosal Immunology, 4th ed.; Mestecky, J., Strober, W., Russell, M.W., Kelsall, B.L., Cheroutre, H., Lambrecht, B.N., Eds.; Academic Press: Boston, MA, USA, 2015; pp. 2097–2124. [Google Scholar]
  29. White, H.D.; Crassi, K.; Wira, C. Cytolytic functional activities of NK cells and cytotoxic T lymphocytes (CTL) are coordinately regulated in the human female reproductive tract. In Mucosal Solutions: Advances in Mucosal Immunology; The University of Sydney: Sydney, Australia, 1997; pp. 385–391. [Google Scholar]
  30. Trifonova, R.T.; Lieberman, J.; van Baarle, D. Distribution of Immune Cells in the Human Cervix and Implications for HIV Transmission. Am. J. Reprod. Immunol. 2014, 71, 252–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Pudney, J.; Quayle, A.J.; Anderson, D.J. Immunological Microenvironments in the Human Vagina and Cervix: Mediators of Cellular Immunity Are Concentrated in the Cervical Transformation Zone1. Biol. Reprod. 2005, 73, 1253–1263. [Google Scholar] [CrossRef] [Green Version]
  32. Givan, A.L.; White, H.D.; Stern, J.E.; Colby, E.; Guyre, P.M.; Wira, C.R.; Gosselin, E.J. Flow Cytometric Analysis of Leukocytes in the Human Female Reproductive Tract: Comparison of Fallopian Tube, Uterus, Cervix, and Vagina. Am. J. Reprod. Immunol. 1997, 38, 350–359. [Google Scholar] [CrossRef]
  33. Kutteh, W.H.; Mestecky, J.; Wira, C. Mucosal Immunity in the Human Female Reproductive Tract; Academic Press: New York, NY, USA, 2005. [Google Scholar]
  34. Sakamoto, Y.; Moran, P.; Bulmer, J.N.; Searle, R.F.; Robson, S.C. Macrophages and not granulocytes are involved in cervical ripening. J. Reprod. Immunol. 2005, 66, 161–173. [Google Scholar] [CrossRef]
  35. Monin, L.; Whettlock, E.M.; Male, V. Immune responses in the human female reproductive tract. Immunology 2020, 160, 106–115. [Google Scholar] [CrossRef] [Green Version]
  36. Wira, C.R.; Fahey, J.V.; Rodriguez-Garcia, M.; Shen, Z.; Patel, M.V. Regulation of Mucosal Immunity in the Female Reproductive Tract: The Role of Sex Hormones in Immune Protection against Sexually Transmitted Pathogens. Am. J. Reprod. Immunol. 2014, 72, 236–258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Maucort-Boulch, D.; Franceschi, S.; Plummer, M.; Group IHPSS. International correlation between human papillomavirus prevalence and cervical cancer incidence. Cancer Epidemiol. Biomark. Prev. 2008, 17, 717–720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Demarco, M.; Hyun, N.; Carter-Pokras, O.; Raine-Bennett, T.R.; Cheung, L.; Chen, X.; Hammer, A.; Campos, N.; Kinney, W.; Gage, J.C.; et al. A study of type-specific HPV natural history and implications for contemporary cervical cancer screening programs. eClinicalMedicine 2020, 22, 100293. [Google Scholar] [CrossRef] [PubMed]
  39. Zhang, J.; Jin, S.; Li, X.; Liu, L.; Xi, L.; Wang, F.; Zhang, S. Human Papillomavirus Type 16 Disables the Increased Natural Killer Cells in Early Lesions of the Cervix. J. Immunol. Res. 2019, 2019, 9182979. [Google Scholar] [CrossRef] [Green Version]
  40. Bosch, F.X.; Burchell, A.N.; Schiffman, M.; Giuliano, A.R.; de Sanjose, S.; Bruni, L.; Tortolero-Luna, G.; Kjaer, S.K.; Muñoz, N. Epidemiology and Natural History of Human Papillomavirus Infections and Type-Specific Implications in Cervical Neoplasia. Vaccine 2008, 26, K1–K16. [Google Scholar] [CrossRef]
  41. Bruno, M.T.; Scalia, G.; Cassaro, N.; Boemi, S. Multiple HPV 16 infection with two strains: A possible marker of neoplastic progression. BMC Cancer 2020, 20, 444. [Google Scholar] [CrossRef]
  42. Chen, W.; Zhang, X.; Molijn, A.; Jenkins, D.; Shi, J.-F.; Quint, W.; Schmidt, J.E.; Wang, P.; Liu, Y.-L.; Li, L.-K.; et al. Human papillomavirus type-distribution in cervical cancer in China: The importance of HPV 16 and 18. Cancer Causes Control 2009, 20, 1705–1713. [Google Scholar] [CrossRef]
  43. Haghshenas, M.; Golini-Moghaddam, T.; Rafiei, A.; Emadeian, O.; Shykhpour, A.; Ashrafi, G.H. Prevalence and type distribution of high-risk human papillomavirus in patients with cervical cancer: A population-based study. Infect. Agents Cancer 2013, 8, 20. [Google Scholar] [CrossRef] [Green Version]
  44. de Freitas, A.C.; de Oliveira, T.H.A.; Barros, M.R.; Venuti, A. hrHPV E5 oncoprotein: Immune evasion and related immunotherapies. J. Exp. Clin. Cancer Res. 2017, 36, 71. [Google Scholar] [CrossRef] [Green Version]
  45. Graham, S.V. The human papillomavirus replication cycle, and its links to cancer progression: A comprehensive review. Clin. Sci. 2017, 131, 2201–2221. [Google Scholar] [CrossRef] [Green Version]
  46. Basukala, O.; Banks, L. The Not-So-Good, the Bad and the Ugly: HPV E5, E6 and E7 Oncoproteins in the Orchestration of Carcinogenesis. Viruses 2021, 13, 1892. [Google Scholar] [CrossRef]
  47. Kumar, V.; Stewart, J.H. Immunometabolic reprogramming, another cancer hallmark. Front. Immunol. 2023, 14, 1125874. [Google Scholar] [CrossRef] [PubMed]
  48. Zhang, R.; Shen, C.; Zhao, L.; Wang, J.; McCrae, M.; Chen, X.; Lu, F. Dysregulation of host cellular genes targeted by human papillomavirus (HPV) integration contributes to HPV-related cervical carcinogenesis. Int. J. Cancer 2016, 138, 1163–1174. [Google Scholar] [CrossRef] [PubMed]
  49. Moody, C.A.; Laimins, L.A. Human papillomavirus oncoproteins: Pathways to transformation. Nat. Rev. Cancer 2010, 10, 550–560. [Google Scholar] [CrossRef] [PubMed]
  50. Stanley Margaret, A. Epithelial Cell Responses to Infection with Human Papillomavirus. Clin. Microbiol. Rev. 2012, 25, 215–222. [Google Scholar] [CrossRef] [Green Version]
  51. Fritz, J.H.; Le Bourhis, L.; Magalhaes, J.G.; Philpott, D.J. Innate immune recognition at the epithelial barrier drives adaptive immunity: APCs take the back seat. Trends Immunol. 2008, 29, 41–49. [Google Scholar] [CrossRef]
  52. Tindle, R.W. Immune evasion in human papillomavirus-associated cervical cancer. Nat. Rev. Cancer 2002, 2, 59–64. [Google Scholar] [CrossRef]
  53. Uhlorn, B.L.; Jackson, R.; Li, S.; Bratton, S.M.; Van Doorslaer, K.; Campos, S.K. Vesicular trafficking permits evasion of cGAS/STING surveillance during initial human papillomavirus infection. PLoS Pathog. 2020, 16, e1009028. [Google Scholar] [CrossRef]
  54. Berger, K.L.; Barriga, F.; Lace, M.J.; Turek, L.P.; Zamba, G.J.; Domann, F.E.; Lee, J.H.; Klingelhutz, A.J. Cervical keratinocytes containing stably replicating extrachromosomal HPV-16 are refractory to transformation by oncogenic H-Ras. Virology 2006, 356, 68–78. [Google Scholar] [CrossRef] [Green Version]
  55. Pett, M.R.; Herdman, M.T.; Palmer, R.D.; Yeo, G.S.H.; Shivji, M.K.; Stanley, M.A.; Coleman, N. Selection of cervical keratinocytes containing integrated HPV16 associates with episome loss and an endogenous antiviral response. Proc. Natl. Acad. Sci. USA 2006, 103, 3822–3827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. De Leo, A.; Calderon, A.; Lieberman, P.M. Control of Viral Latency by Episome Maintenance Proteins. Trends Microbiol. 2020, 28, 150–162. [Google Scholar] [CrossRef] [PubMed]
  57. Nees, M.; Geoghegan, J.M.; Hyman, T.; Frank, S.; Miller, L.; Woodworth, C.D. Papillomavirus type 16 oncogenes downregulate expression of interferon-responsive genes and upregulate proliferation-associated and NF-kappaB-responsive genes in cervical keratinocytes. J. Virol. 2001, 75, 4283–4296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Lau, L.; Gray, E.E.; Brunette, R.L.; Stetson, D.B. DNA tumor virus oncogenes antagonize the cGAS-STING DNA-sensing pathway. Science 2015, 350, 568–571. [Google Scholar] [CrossRef] [Green Version]
  59. Lou, M.; Huang, D.; Zhou, Z.; Shi, X.; Wu, M.; Rui, Y.; Su, J.; Zheng, W.; Yu, X.-F. DNA virus oncoprotein HPV18 E7 selectively antagonizes cGAS-STING-triggered innate immune activation. J. Med. Virol. 2023, 95, e28310. [Google Scholar] [CrossRef]
  60. Xiao, D.; Huang, W.; Ou, M.; Guo, C.; Ye, X.; Liu, Y.; Wang, M.; Zhang, B.; Zhang, N.; Huang, S.; et al. Interaction between susceptibility loci in cGAS-STING pathway, MHC gene and HPV infection on the risk of cervical precancerous lesions in Chinese population. Oncotarget 2016, 7, 84228–84238. [Google Scholar] [CrossRef] [Green Version]
  61. Berti, F.C.B.; Pereira, A.P.L.; Cebinelli, G.C.M.; Trugilo, K.P.; Brajão de Oliveira, K. The role of interleukin 10 in human papilloma virus infection and progression to cervical carcinoma. Cytokine Growth Factor. Rev. 2017, 34, 1–13. [Google Scholar] [CrossRef]
  62. Nees, M.; Geoghegan, J.M.; Munson, P.; Prabhu, V.; Liu, Y.; Androphy, E.; Woodworth, C.D. Human papillomavirus type 16 E6 and E7 proteins inhibit differentiation-dependent expression of transforming growth factor-beta2 in cervical keratinocytes. Cancer Res. 2000, 60, 4289–4298. [Google Scholar]
  63. Iglesias, M.; Yen, K.; Gaiotti, D.; Hildesheim, A.; Stoler, M.H.; Woodworth, C.D. Human papillomavirus type 16 E7 protein sensitizes cervical keratinocytes to apoptosis and release of interleukin-1alpha. Oncogene 1998, 17, 1195–1205. [Google Scholar] [CrossRef] [Green Version]
  64. Shimada, M.; Yamashita, A.; Saito, M.; Ichino, M.; Kinjo, T.; Mizuki, N.; Klinman, D.M.; Okuda, K. The human papillomavirus E6 protein targets apoptosis-inducing factor (AIF) for degradation. Sci. Rep. 2020, 10, 14195. [Google Scholar] [CrossRef] [PubMed]
  65. Guo, L.; Hua, K.A.-O. Cervical Cancer: Emerging Immune Landscape and Treatment. OncoTargets Ther. 2020, 13, 8037–8047. [Google Scholar] [CrossRef] [PubMed]
  66. Ghosh, M.; Saha, S.; Li, J.; Montrose, D.C.; Martinez, L.A. p53 engages the cGAS/STING cytosolic DNA sensing pathway for tumor suppression. Mol. Cell 2023, 83, 266–280.e266. [Google Scholar] [CrossRef] [PubMed]
  67. Zhang, S.; Wang, H.; Liu, J.; Tao, T.; Zeng, Z.; Wang, M. RGS1 and related genes as potential targets for immunotherapy in cervical cancer: Computational biology and experimental validation. J. Transl. Med. 2022, 20, 334. [Google Scholar] [CrossRef]
  68. Agenès, F.; Bosco, N.; Mascarell, L.; Fritah, S.; Ceredig, R. Differential expression of regulator of G-protein signalling transcripts and in vivo migration of CD4+ naïve and regulatory T cells. Immunology 2005, 115, 179–188. [Google Scholar] [CrossRef]
  69. Shi, G.X.; Harrison, K.; Han, S.B.; Moratz, C.; Kehrl, J.H. Toll-like receptor signaling alters the expression of regulator of G protein signaling proteins in dendritic cells: Implications for G protein-coupled receptor signaling. J. Immunol. 2004, 172, 5175–5184. [Google Scholar] [CrossRef] [Green Version]
  70. Bai, Y.; Hu, M.; Chen, Z.; Wei, J.; Du, H. Single-Cell Transcriptome Analysis Reveals RGS1 as a New Marker and Promoting Factor for T-Cell Exhaustion in Multiple Cancers. Front. Immunol. 2021, 12, 767070. [Google Scholar] [CrossRef]
  71. Kang, Y.; Huang, J.; Liu, Y.; Zhang, N.; Cheng, Q.; Zhang, Y. Integrated Analysis of Immune Infiltration Features for Cervical Carcinoma and Their Associated Immunotherapeutic Responses. Front. Cell Dev. Biol. 2021, 9, 573497. [Google Scholar] [CrossRef]
  72. Yamato, K.; Yamada, T.; Kizaki, M.; Ui-Tei, K.; Natori, Y.; Fujino, M.; Nishihara, T.; Ikeda, Y.; Nasu, Y.; Saigo, K.; et al. New highly potent and specific E6 and E7 siRNAs for treatment of HPV16 positive cervical cancer. Cancer Gene Ther. 2008, 15, 140–153. [Google Scholar] [CrossRef] [Green Version]
  73. Bergot, A.S.; Ford, N.; Leggatt, G.R.; Wells, J.W.; Frazer, I.H.; Grimbaldeston, M.A. HPV16-E7 expression in squamous epithelium creates a local immune suppressive environment via CCL2- and CCL5- mediated recruitment of mast cells. PLoS Pathog. 2014, 10, e1004466. [Google Scholar] [CrossRef]
  74. Pal, A.; Kundu, R. Human Papillomavirus E6 and E7: The Cervical Cancer Hallmarks and Targets for Therapy. Front. Microbiol. 2020, 10, 3116. [Google Scholar] [CrossRef] [Green Version]
  75. Taghizadeh, E.; Jahangiri, S.; Rostami, D.; Taheri, F.; Renani, P.G.; Taghizadeh, H.; Gheibi Hayat, S.M. Roles of E6 and E7 Human Papillomavirus Proteins in Molecular Pathogenesis of Cervical Cancer. Curr. Protein Pept. Sci. 2019, 20, 926–934. [Google Scholar] [CrossRef]
  76. Romani, N.; Brunner, P.M.; Stingl, G. Changing Views of the Role of Langerhans Cells. J. Investig. Dermatol. 2012, 132, 872–881. [Google Scholar] [CrossRef] [Green Version]
  77. Doebel, T.; Voisin, B.; Nagao, K. Langerhans Cells—The Macrophage in Dendritic Cell Clothing. Trends Immunol. 2017, 38, 817–828. [Google Scholar] [CrossRef]
  78. Merad, M.; Ginhoux, F.; Collin, M. Origin, homeostasis and function of Langerhans cells and other langerin-expressing dendritic cells. Nat. Rev. Immunol. 2008, 8, 935–947. [Google Scholar] [CrossRef] [PubMed]
  79. Woodham, A.W.; Raff, A.B.; Raff, L.M.; Da Silva, D.M.; Yan, L.; Skeate, J.G.; Wong, M.K.; Lin, Y.G.; Kast, W.M. Inhibition of Langerhans cell maturation by human papillomavirus type 16: A novel role for the annexin A2 heterotetramer in immune suppression. J. Immunol. 2014, 192, 4748–4757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Da Silva, D.M.; Woodham, A.W.; Skeate, J.G.; Rijkee, L.K.; Taylor, J.R.; Brand, H.E.; Muderspach, L.I.; Roman, L.D.; Yessaian, A.A.; Pham, H.Q.; et al. Langerhans cells from women with cervical precancerous lesions become functionally responsive against human papillomavirus after activation with stabilized Poly-I:C. Clin. Immunol. 2015, 161, 197–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Fahey, L.M.; Raff, A.B.; Da Silva, D.M.; Kast, W.M. Reversal of human papillomavirus-specific T cell immune suppression through TLR agonist treatment of Langerhans cells exposed to human papillomavirus type 16. J. Immunol. 2009, 182, 2919–2928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Fausch, S.C.; Da Silva, D.M.; Kast, W.M. Heterologous papillomavirus virus-like particles and human papillomavirus virus-like particle immune complexes activate human Langerhans cells. Vaccine 2005, 23, 1720–1729. [Google Scholar] [CrossRef]
  83. Vignali, D.A.A.; Kuchroo, V.K. IL-12 family cytokines: Immunological playmakers. Nat. Immunol. 2012, 13, 722–728. [Google Scholar] [CrossRef] [Green Version]
  84. Gee, K.; Guzzo, C.; Che Mat, N.F.; Ma, W.; Kumar, A. The IL-12 family of cytokines in infection, inflammation and autoimmune disorders. Inflamm. Allergy Drug. Targets 2009, 8, 40–52. [Google Scholar] [CrossRef]
  85. García-Piñeres, A.J.; Hildesheim, A.; Trivett, M.; Williams, M.; Wu, L.; Kewalramani, V.N.; Pinto, L.A. Role of DC-SIGN in the activation of dendritic cells by HPV-16 L1 virus-like particle vaccine. Eur. J. Immunol. 2006, 36, 437–445. [Google Scholar] [CrossRef]
  86. Woodham, A.W.; Yan, L.; Skeate, J.G.; van der Veen, D.; Brand, H.H.; Wong, M.K.; Da Silva, D.M.; Kast, W.M. T cell ignorance is bliss: T cells are not tolerized by Langerhans cells presenting human papillomavirus antigens in the absence of costimulation. Papillomavirus Res. 2016, 2, 21–30. [Google Scholar] [CrossRef] [Green Version]
  87. Reyna-Hernández, M.A.; Alarcón-Romero, L.D.C.; Ortiz-Ortiz, J.; Illades-Aguiar, B.; Jiménez-López, M.A.; Ocampo-Bárcenas, A.; Morrugares-Ixtepan, M.O.; Torres-Rojas, F.I. GLUT1, LDHA, and MCT4 Expression Is Deregulated in Cervical Cancer and Precursor Lesions. J. Histochem. Cytochem. 2022, 70, 437–446. [Google Scholar] [CrossRef]
  88. Bedoya, A.M.; Tate, D.J.; Baena, A.; Córdoba, C.M.; Borrero, M.; Pareja, R.; Rojas, F.; Patterson, J.R.; Herrero, R.; Zea, A.H.; et al. Immunosuppression in cervical cancer with special reference to arginase activity. Gynecol. Oncol. 2014, 135, 74–80. [Google Scholar] [CrossRef]
  89. Caldwell, R.W.; Rodriguez, P.C.; Toque, H.A.; Narayanan, S.P.; Caldwell, R.B. Arginase: A Multifaceted Enzyme Important in Health and Disease. Physiol. Rev. 2018, 98, 641–665. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Casero, R.A., Jr.; Murray Stewart, T.; Pegg, A.E. Polyamine metabolism and cancer: Treatments, challenges and opportunities. Nat. Rev. Cancer 2018, 18, 681–695. [Google Scholar] [CrossRef] [PubMed]
  91. Gerner, E.W.; Meyskens, F.L., Jr. Polyamines and cancer: Old molecules, new understanding. Nat. Rev. Cancer 2004, 4, 781–792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Souid, M.; Ghedira, R.; Souissi, S.; Bouzgarrou, N.; Gabbouj, S.; Shini-Hadhri, S.; Rhim, M.-S.; Boukadida, A.; Toumi, D.; Faleh, R.; et al. Arginase is involved in cervical lesions progression and severity. Immunobiology 2022, 227, 152189. [Google Scholar] [CrossRef] [PubMed]
  93. Venancio, P.A.; Consolaro, M.E.L.; Derchain, S.F.; Boccardo, E.; Villa, L.L.; Maria-Engler, S.S.; Campa, A.; Discacciati, M.G. Indoleamine 2,3-dioxygenase and tryptophan 2,3-dioxygenase expression in HPV infection, SILs, and cervical cancer. Cancer Cytopathol. 2019, 127, 586–597. [Google Scholar] [CrossRef]
  94. Hascitha, J.; Priya, R.; Jayavelu, S.; Dhandapani, H.; Selvaluxmy, G.; Sunder Singh, S.; Rajkumar, T. Analysis of Kynurenine/Tryptophan ratio and expression of IDO1 and 2 mRNA in tumour tissue of cervical cancer patients. Clin. Biochem. 2016, 49, 919–924. [Google Scholar] [CrossRef]
  95. Ferns, D.M.; Kema, I.P.; Buist, M.R.; Nijman, H.W.; Kenter, G.G.; Jordanova, E.S. Indoleamine-2,3-dioxygenase (IDO) metabolic activity is detrimental for cervical cancer patient survival. OncoImmunology 2015, 4, e981457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Yang, S.L.; Tan, H.X.; Niu, T.T.; Liu, Y.K.; Gu, C.J.; Li, D.J.; Li, M.Q.; Wang, H.Y. The IFN-γ-IDO1-kynureine pathway-induced autophagy in cervical cancer cell promotes phagocytosis of macrophage. Int. J. Biol. Sci. 2021, 17, 339–352. [Google Scholar] [CrossRef] [PubMed]
  97. Hu, Y.; Liu, Z.; Tang, H. Tryptophan 2,3-dioxygenase may be a potential prognostic biomarker and immunotherapy target in cancer: A meta-analysis and bioinformatics analysis. Front. Oncol. 2022, 12, 977640. [Google Scholar] [CrossRef]
  98. Wang, J.; Mijiti, Y.; Chen, Y.; Liu, Z. Aryl hydrocarbon receptor is a prognostic biomarker and is correlated with immune responses in cervical cancer. Bioengineered 2021, 12, 11922–11935. [Google Scholar] [CrossRef] [PubMed]
  99. Opitz, C.A.; Litzenburger, U.M.; Sahm, F.; Ott, M.; Tritschler, I.; Trump, S.; Schumacher, T.; Jestaedt, L.; Schrenk, D.; Weller, M.; et al. An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature 2011, 478, 197–203. [Google Scholar] [CrossRef] [Green Version]
  100. Feng, S.; Cao, Z.; Wang, X. Role of aryl hydrocarbon receptor in cancer. Biochim. Biophys. Acta (BBA)-Rev. Cancer 2013, 1836, 197–210. [Google Scholar] [CrossRef]
  101. Gasiewicz, T.A.; Henry, E.C.; Collins, L.L. Expression and Activity of Aryl Hydrocarbon Receptors in Development and Cancer. Crit. Rev. Eukaryot. Gene Expr. 2008, 18, 279–321. [Google Scholar] [CrossRef]
  102. Marvin, D.L.; Spaans, V.M.; de Kroon, C.D.; Slieker, R.C.; Khelil, M.; Ten Dijke, P.; Ritsma, L.; Jordanova, E.S. Low Transforming Growth Factor-β Pathway Activity in Cervical Adenocarcinomas. Front. Oncol. 2022, 12, 797453. [Google Scholar] [CrossRef]
  103. Labani-Motlagh, A.; Ashja-Mahdavi, M.; Loskog, A. The Tumor Microenvironment: A Milieu Hindering and Obstructing Antitumor Immune Responses. Front. Immunol. 2020, 11, 940. [Google Scholar] [CrossRef]
  104. Falcomatà, C.; Bärthel, S.; Schneider, G.; Rad, R.; Schmidt-Supprian, M.; Saur, D. Context-Specific Determinants of the Immunosuppressive Tumor Microenvironment in Pancreatic Cancer. Cancer Discov. 2023, 13, 278–297. [Google Scholar] [CrossRef]
  105. Wang, Q.; Shao, X.; Zhang, Y.; Zhu, M.; Wang, F.X.C.; Mu, J.; Li, J.; Yao, H.; Chen, K. Role of tumor microenvironment in cancer progression and therapeutic strategy. Cancer Med. 2023, 12, 11149–11165. [Google Scholar] [CrossRef] [PubMed]
  106. Piersma, S.J. Immunosuppressive tumor microenvironment in cervical cancer patients. Cancer Microenviron. 2011, 4, 361–375. [Google Scholar] [CrossRef] [Green Version]
  107. Yang, X.; Zhang, W.; Zhu, W. Profiling of immune responses by lactate modulation in cervical cancer reveals key features driving clinical outcome. Heliyon 2023, 9, e14896. [Google Scholar] [CrossRef] [PubMed]
  108. Zhao, M.; Li, Y.; Wei, X.; Zhang, Q.; Jia, H.; Quan, S.; Cao, D.; Wang, L.; Yang, T.; Zhao, J.; et al. Negative immune factors might predominate local tumor immune status and promote carcinogenesis in cervical carcinoma. Virol. J. 2017, 14, 5. [Google Scholar] [CrossRef] [Green Version]
  109. Xu, J.; Huang, Z.; Wang, Y.; Xiang, Z.; Xiong, B. Identification of Novel Tumor Microenvironment Regulating Factor That Facilitates Tumor Immune Infiltration in Cervical Cancer. Front. Oncol. 2022, 12, 846786. [Google Scholar] [CrossRef]
  110. Miyata, Y.; Ogo, E.; Abe, T.; Hirata, H.; Tsuda, N.; Ushijima, K.; Kawahara, A.; Akiba, J.; Obara, H.; Kakuma, T. Dynamics in the expression of programmed death ligand 1 and cluster of differentiation 163 in the tumor microenvironment of uterine cervical cancer: A single-center retrospective study. Radiat. Oncol. 2023, 18, 40. [Google Scholar] [CrossRef] [PubMed]
  111. Pu, Y.; Ji, Q. Tumor-Associated Macrophages Regulate PD-1/PD-L1 Immunosuppression. Front. Immunol. 2022, 13, 874589. [Google Scholar] [CrossRef] [PubMed]
  112. Huang, W.; Liu, J.; Xu, K.; Chen, H.; Bian, C. PD-1/PD-L1 inhibitors for advanced or metastatic cervical cancer: From bench to bed. Front. Oncol. 2022, 12, 849352. [Google Scholar] [CrossRef]
  113. Mezache, L.; Paniccia, B.; Nyinawabera, A.; Nuovo, G.J. Enhanced expression of PD L1 in cervical intraepithelial neoplasia and cervical cancers. Mod. Pathol. 2015, 28, 1594–1602. [Google Scholar] [CrossRef] [Green Version]
  114. Liu, C.; Lu, J.; Tian, H.; Du, W.; Zhao, L.; Feng, J.; Yuan, D.; Li, Z. Increased expression of PD-L1 by the human papillomavirus 16 E7 oncoprotein inhibits anticancer immunity. Mol. Med. Rep. 2017, 15, 1063–1070. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Yang, W.; Lu, Y.P.; Yang, Y.Z.; Kang, J.R.; Jin, Y.D.; Wang, H.W. Expressions of programmed death (PD)-1 and PD-1 ligand (PD-L1) in cervical intraepithelial neoplasia and cervical squamous cell carcinomas are of prognostic value and associated with human papillomavirus status. J. Obs. Gynaecol. Res. 2017, 43, 1602–1612. [Google Scholar] [CrossRef] [PubMed]
  116. Meng, Y.; Liang, H.; Hu, J.; Liu, S.; Hao, X.; Wong, M.S.K.; Li, X.; Hu, L. PD-L1 Expression Correlates with Tumor Infiltrating Lymphocytes and Response to Neoadjuvant Chemotherapy in Cervical Cancer. J. Cancer 2018, 9, 2938–2945. [Google Scholar] [CrossRef]
  117. Yang-Chun, F.; Zhen-Zhen, C.; Yan-Chun, H.; Xiu-Min, M. Association between PD-L1 and HPV status and the prognostic value for HPV treatment in premalignant cervical lesion patients. Medicine 2017, 96, e7270. [Google Scholar] [CrossRef]
  118. Yao, S.; Zhao, L.; Chen, S.; Wang, H.; Gao, Y.; Shao, N.-Y.; Dai, M.; Cai, H. Cervical cancer immune infiltration microenvironment identification, construction of immune scores, assisting patient prognosis and immunotherapy. Front. Immunol. 2023, 14, 1135657. [Google Scholar] [CrossRef] [PubMed]
  119. Walker, L.S.K. Treg and CTLA-4: Two intertwining pathways to immune tolerance. J. Autoimmun. 2013, 45, 49–57. [Google Scholar] [CrossRef] [Green Version]
  120. Imazeki, H.; Ogiwara, Y.; Kawamura, M.; Boku, N.; Kudo-Saito, C. CD11b(+)CTLA4(+) myeloid cells are a key driver of tumor evasion in colorectal cancer. J. Immunother. Cancer 2021, 9, e002841. [Google Scholar] [CrossRef]
  121. Li, H.; van der Leun, A.M.; Yofe, I.; Lubling, Y.; Gelbard-Solodkin, D.; van Akkooi, A.C.J.; van den Braber, M.; Rozeman, E.A.; Haanen, J.; Blank, C.U.; et al. Dysfunctional CD8 T Cells Form a Proliferative, Dynamically Regulated Compartment within Human Melanoma. Cell 2019, 176, 775–789.e718. [Google Scholar] [CrossRef]
  122. Yang, X.; Chen, Y.; Li, M.; Zhu, W. ERBB3 methylation and immune infiltration in tumor microenvironment of cervical cancer. Sci. Rep. 2022, 12, 8112. [Google Scholar] [CrossRef]
  123. Audirac-Chalifour, A.; Torres-Poveda, K.; Bahena-Román, M.; Téllez-Sosa, J.; Martínez-Barnetche, J.; Cortina-Ceballos, B.; López-Estrada, G.; Delgado-Romero, K.; Burguete-García, A.I.; Cantú, D.; et al. Cervical Microbiome and Cytokine Profile at Various Stages of Cervical Cancer: A Pilot Study. PLoS ONE 2016, 11, e0153274. [Google Scholar] [CrossRef] [Green Version]
  124. Kyrgiou, M.; Moscicki, A.-B. Vaginal microbiome and cervical cancer. Semin. Cancer Biol. 2022, 86, 189–198. [Google Scholar] [CrossRef]
  125. Vaccarella, S.; Lortet-Tieulent, J.; Plummer, M.; Franceschi, S.; Bray, F. Worldwide trends in cervical cancer incidence: Impact of screening against changes in disease risk factors. Eur. J. Cancer 2013, 49, 3262–3273. [Google Scholar] [CrossRef]
  126. Castanheira, C.P.; Sallas, M.L.; Nunes, R.A.L.; Lorenzi, N.P.C.; Termini, L. Microbiome and Cervical Cancer. Pathobiology 2021, 88, 187–197. [Google Scholar] [CrossRef]
  127. Tang, Y.; Zhang, A.X.J.; Chen, G.; Wu, Y.; Gu, W. Prognostic and therapeutic TILs of cervical cancer—Current advances and future perspectives. Mol. Ther.-Oncolytics 2021, 22, 410–430. [Google Scholar] [CrossRef]
  128. Binnewies, M.; Roberts, E.W.; Kersten, K.; Chan, V.; Fearon, D.F.; Merad, M.; Coussens, L.M.; Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Hedrick, C.C.; et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 2018, 24, 541–550. [Google Scholar] [CrossRef] [PubMed]
  129. Frankel, T.; Lanfranca, M.P.; Zou, W. The Role of Tumor Microenvironment in Cancer Immunotherapy. Adv. Exp. Med. Biol. 2017, 1036, 51–64. [Google Scholar]
  130. Ostuni, R.; Kratochvill, F.; Murray, P.J.; Natoli, G. Macrophages and cancer: From mechanisms to therapeutic implications. Trends Immunol. 2015, 36, 229–239. [Google Scholar] [CrossRef]
  131. Ma, R.-Y.; Black, A.; Qian, B.-Z. Macrophage diversity in cancer revisited in the era of single-cell omics. Trends Immunol. 2022, 43, 546–563. [Google Scholar] [CrossRef]
  132. Kumar, V. Macrophages: The Potent Immunoregulatory Innate Immune Cells. In Macrophage Activation; Khalid Hussain, B., Ed.; IntechOpen: Rijeka, Croatia, 2019; Chapter 1. [Google Scholar]
  133. Liu, J.; Geng, X.; Hou, J.; Wu, G. New insights into M1/M2 macrophages: Key modulators in cancer progression. Cancer Cell Int. 2021, 21, 389. [Google Scholar] [CrossRef] [PubMed]
  134. Li, C.; Hua, K. Dissecting the Single-Cell Transcriptome Network of Immune Environment Underlying Cervical Premalignant Lesion, Cervical Cancer and Metastatic Lymph Nodes. Front. Immunol. 2022, 13, 897366. [Google Scholar] [CrossRef] [PubMed]
  135. Mantovani, A.; Allavena, P.; Marchesi, F.; Garlanda, C. Macrophages as tools and targets in cancer therapy. Nat. Rev. Drug. Discov. 2022, 21, 799–820. [Google Scholar] [CrossRef] [PubMed]
  136. Nuñez, S.Y.; Ziblat, A.; Secchiari, F.; Torres, N.I.; Sierra, J.M.; Raffo Iraolagoitia, X.L.; Araya, R.E.; Domaica, C.I.; Fuertes, M.B.; Zwirner, N.W. Human M2 Macrophages Limit NK Cell Effector Functions through Secretion of TGF-β and Engagement of CD85j. J. Immunol. 2018, 200, 1008–1015. [Google Scholar] [CrossRef] [Green Version]
  137. Guo, F.; Feng, Y.C.; Zhao, G.; Zhang, R.; Cheng, Z.Z.; Kong, W.N.; Wu, H.L.; Xu, B.; Lv, X.; Ma, X.M. Tumor-Associated CD163(+) M2 Macrophage Infiltration is Highly Associated with PD-L1 Expression in Cervical Cancer. Cancer Manag. Res. 2020, 12, 5831–5843. [Google Scholar] [CrossRef] [PubMed]
  138. Chen, X.-J.; Wu, S.; Yan, R.-M.; Fan, L.-S.; Yu, L.; Zhang, Y.-M.; Wei, W.-F.; Zhou, C.-F.; Wu, X.-G.; Zhong, M.; et al. The role of the hypoxia-Nrp-1 axis in the activation of M2-like tumor-associated macrophages in the tumor microenvironment of cervical cancer. Mol. Carcinog. 2019, 58, 388–397. [Google Scholar] [CrossRef]
  139. Ren, J.; Li, L.; Yu, B.; Xu, E.; Sun, N.; Li, X.; Xing, Z.; Han, X.; Cui, Y.; Wang, X.; et al. Extracellular vesicles mediated proinflammatory macrophage phenotype induced by radiotherapy in cervical cancer. BMC Cancer 2022, 22, 88. [Google Scholar] [CrossRef] [PubMed]
  140. Krneta, T.; Gillgrass, A.; Poznanski, S.; Chew, M.; Lee, A.J.; Kolb, M.; Ashkar, A.A. M2-polarized and tumor-associated macrophages alter NK cell phenotype and function in a contact-dependent manner. J. Leukoc. Biol. 2017, 101, 285–295. [Google Scholar] [CrossRef] [PubMed]
  141. Wang, S.; Yang, Y.; Ma, P.; Zha, Y.; Zhang, J.; Lei, A.; Li, N. CAR-macrophage: An extensive immune enhancer to fight cancer. eBioMedicine 2022, 76, 103873. [Google Scholar] [CrossRef]
  142. Klichinsky, M.; Ruella, M.; Shestova, O.; Lu, X.M.; Best, A.; Zeeman, M.; Schmierer, M.; Gabrusiewicz, K.; Anderson, N.R.; Petty, N.E.; et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat. Biotechnol. 2020, 38, 947–953. [Google Scholar] [CrossRef]
  143. Shaul, M.E.; Fridlender, Z.G. Tumour-associated neutrophils in patients with cancer. Nat. Rev. Clin. Oncol. 2019, 16, 601–620. [Google Scholar] [CrossRef]
  144. Hedrick, C.C.; Malanchi, I. Neutrophils in cancer: Heterogeneous and multifaceted. Nat. Rev. Immunol. 2022, 22, 173–187. [Google Scholar] [CrossRef]
  145. Jaillon, S.; Ponzetta, A.; Di Mitri, D.; Santoni, A.; Bonecchi, R.; Mantovani, A. Neutrophil diversity and plasticity in tumour progression and therapy. Nat. Rev. Cancer 2020, 20, 485–503. [Google Scholar] [CrossRef]
  146. Coffelt, S.B.; Wellenstein, M.D.; de Visser, K.E. Neutrophils in cancer: Neutral no more. Nat. Rev. Cancer 2016, 16, 431–446. [Google Scholar] [CrossRef] [Green Version]
  147. Nomelini, R.S.; Mota, S.D.S.; Murta, E.F.C. Absolute band neutrophils count is a predictor of overall survival in advanced uterine cervical cancer. Arch. Gynecol. Obstet. 2022, 306, 1697–1701. [Google Scholar] [CrossRef]
  148. Carus, A.; Ladekarl, M.; Hager, H.; Nedergaard, B.S.; Donskov, F. Tumour-associated CD66b+ neutrophil count is an independent prognostic factor for recurrence in localised cervical cancer. Br. J. Cancer 2013, 108, 2116–2122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Yan, B.; Dai, X.; Ma, Q.; Wu, X. Stromal Neutrophil Extracellular Trap Density Is an Independent Prognostic Factor for Cervical Cancer Recurrence. Front. Oncol. 2021, 11, 659445. [Google Scholar] [CrossRef] [PubMed]
  150. Punt, S.; Fleuren, G.J.; Kritikou, E.; Lubberts, E.; Trimbos, J.B.; Jordanova, E.S.; Gorter, A. Angels and demons: Th17 cells represent a beneficial response, while neutrophil IL-17 is associated with poor prognosis in squamous cervical cancer. Oncoimmunology 2015, 4, e984539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Ittiamornlert, P.; Ruengkhachorn, I. Neutrophil-lymphocyte ratio as a predictor of oncologic outcomes in stage IVB, persistent, or recurrent cervical cancer patients treated by chemotherapy. BMC Cancer 2019, 19, 51. [Google Scholar] [CrossRef] [PubMed]
  152. Calo, C.A.; Barrington, D.A.; Brown, M.; Gonzalez, L.; Baek, J.; Huffman, A.; Benedict, J.; Backes, F.; Chambers, L.; Cohn, D.; et al. High pre-treatment neutrophil-to-lymphocyte ratio as a prognostic marker for worse survival in patients with recurrent/metastatic cervical cancer treated with immune checkpoint inhibitors. Gynecol. Oncol. Rep. 2022, 42, 101040. [Google Scholar] [CrossRef]
  153. Gruijs, M.; Sewnath, C.A.N.; van Egmond, M. Therapeutic exploitation of neutrophils to fight cancer. Semin. Immunol. 2021, 57, 101581. [Google Scholar] [CrossRef]
  154. Carnevale, S.; Ghasemi, S.; Rigatelli, A.; Jaillon, S. The complexity of neutrophils in health and disease: Focus on cancer. Semin. Immunol. 2020, 48, 101409. [Google Scholar] [CrossRef]
  155. Geh, D.; Leslie, J.; Rumney, R.; Reeves, H.L.; Bird, T.G.; Mann, D.A. Neutrophils as potential therapeutic targets in hepatocellular carcinoma. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 257–273. [Google Scholar] [CrossRef] [PubMed]
  156. Németh, T.; Sperandio, M.; Mócsai, A. Neutrophils as emerging therapeutic targets. Nat. Rev. Drug Discov. 2020, 19, 253–275. [Google Scholar] [CrossRef] [PubMed]
  157. Groth, C.; Hu, X.; Weber, R.; Fleming, V.; Altevogt, P.; Utikal, J.; Umansky, V. Immunosuppression mediated by myeloid-derived suppressor cells (MDSCs) during tumour progression. Br. J. Cancer 2019, 120, 16–25. [Google Scholar] [CrossRef] [Green Version]
  158. Kumar, V.; Patel, S.; Tcyganov, E.; Gabrilovich, D.I. The Nature of Myeloid-Derived Suppressor Cells in the Tumor Microenvironment. Trends Immunol. 2016, 37, 208–220. [Google Scholar] [CrossRef] [Green Version]
  159. Raskov, H.; Orhan, A.; Gaggar, S.; Gögenur, I. Neutrophils and polymorphonuclear myeloid-derived suppressor cells: An emerging battleground in cancer therapy. Oncogenesis 2022, 11, 22. [Google Scholar] [CrossRef]
  160. Wesolowski, R.; Markowitz, J.; Carson, W.E. Myeloid derived suppressor cells—A new therapeutic target in the treatment of cancer. J. Immunotherapy Cancer 2013, 1, 10. [Google Scholar] [CrossRef] [Green Version]
  161. Kawano, M.; Mabuchi, S.; Matsumoto, Y.; Sasano, T.; Takahashi, R.; Kuroda, H.; Kozasa, K.; Hashimoto, K.; Isobe, A.; Sawada, K.; et al. The significance of G-CSF expression and myeloid-derived suppressor cells in the chemoresistance of uterine cervical cancer. Sci. Rep. 2015, 5, 18217. [Google Scholar] [CrossRef] [Green Version]
  162. Sasano, T.; Mabuchi, S.; Kozasa, K.; Kuroda, H.; Kawano, M.; Takahashi, R.; Komura, N.; Yokoi, E.; Matsumoto, Y.; Hashimoto, K.; et al. The Highly Metastatic Nature of Uterine Cervical/Endometrial Cancer Displaying Tumor-Related Leukocytosis: Clinical and Preclinical Investigations. Clin. Cancer Res. 2018, 24, 4018–4029. [Google Scholar] [CrossRef] [Green Version]
  163. Cho, Y.; Kim, K.H.; Yoon, H.I.; Kim, G.E.; Kim, Y.B. Tumor-related leukocytosis is associated with poor radiation response and clinical outcome in uterine cervical cancer patients. Ann. Oncol. 2016, 27, 2067–2074. [Google Scholar] [CrossRef]
  164. Nagaraj, S.; Youn, J.I.; Gabrilovich, D.I. Reciprocal relationship between myeloid-derived suppressor cells and T cells. J. Immunol. 2013, 191, 17–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Lu, Z.; Zhu, M.; Marley, J.L.; Bi, K.; Wang, K.; Zhai, M.; Hu, H.; Guo, P.; Li, C.; Xu, Y.; et al. The combined action of monocytic myeloid-derived suppressor cells and mucosal-associated invariant T cells promotes the progression of cervical cancer. Int. J. Cancer 2021, 148, 1499–1507. [Google Scholar] [CrossRef] [PubMed]
  166. Liang, Y.; Wang, W.; Zhu, X.; Yu, M.; Zhou, C. Inhibition of myeloid-derived suppressive cell function with all-trans retinoic acid enhanced anti-PD-L1 efficacy in cervical cancer. Sci. Rep. 2022, 12, 9619. [Google Scholar] [CrossRef] [PubMed]
  167. Kumar, V.; Ahmad, A. Role of MAIT cells in the immunopathogenesis of inflammatory diseases: New players in old game. Int. Rev. Immunol. 2018, 37, 90–110. [Google Scholar] [CrossRef]
  168. Ussher, J.E.; Willberg, C.B.; Klenerman, P. MAIT cells and viruses. Immunol. Cell Biol. 2018, 96, 630–641. [Google Scholar] [CrossRef] [PubMed]
  169. Rouxel, O.; Lehuen, A. Mucosal-associated invariant T cells in autoimmune and immune-mediated diseases. Immunol. Cell Biol. 2018, 96, 618–629. [Google Scholar] [CrossRef]
  170. Hinks, T.S.C.; van Wilgenburg, B.; Wang, H.; Loh, L.; Koutsakos, M.; Kedzierska, K.; Corbett, A.J.; Chen, Z. Study of MAIT Cell Activation in Viral Infections In Vivo. Methods Mol. Biol. 2020, 2098, 261–281. [Google Scholar]
  171. Zumwalde, N.A.; Gumperz, J.E. Mucosal-Associated Invariant T Cells in Tumors of Epithelial Origin. Adv. Exp. Med. Biol. 2020, 1224, 63–77. [Google Scholar]
  172. Berzins, S.P.; Wallace, M.E.; Kannourakis, G.; Kelly, J. A Role for MAIT Cells in Colorectal Cancer. Front. Immunol. 2020, 11, 949. [Google Scholar] [CrossRef]
  173. Shaler, C.R.; Tun-Abraham, M.E.; Skaro, A.I.; Khazaie, K.; Corbett, A.J.; Mele, T.; Hernandez-Alejandro, R.; Haeryfar, S.M.M. Mucosa-associated invariant T cells infiltrate hepatic metastases in patients with colorectal carcinoma but are rendered dysfunctional within and adjacent to tumor microenvironment. Cancer Immunol. Immunother. 2017, 66, 1563–1575. [Google Scholar] [CrossRef]
  174. Haeryfar, S.M.M.; Shaler, C.R.; Rudak, P.T. Mucosa-associated invariant T cells in malignancies: A faithful friend or formidable foe? Cancer Immunol. Immunother. 2018, 67, 1885–1896. [Google Scholar] [CrossRef]
  175. Melo, A.M.; O’Brien, A.M.; Phelan, J.J.; Kennedy, S.A.; Wood, N.A.W.; Veerapen, N.; Besra, G.S.; Clarke, N.E.; Foley, E.K.; Ravi, A.; et al. Mucosal-Associated Invariant T Cells Display Diminished Effector Capacity in Oesophageal Adenocarcinoma. Front. Immunol. 2019, 10, 1580. [Google Scholar] [CrossRef]
  176. O’Neill, C.; Cassidy, F.C.; O’Shea, D.; Hogan, A.E. Mucosal Associated Invariant T Cells in Cancer-Friend or Foe? Cancers 2021, 13, 1582. [Google Scholar] [CrossRef] [PubMed]
  177. Li, Y.-R.; Zhou, K.; Wilson, M.; Kramer, A.; Zhu, Y.; Dawson, N.; Yang, L. Mucosal-associated invariant T cells for cancer immunotherapy. Mol. Ther. 2023, 31, 631–646. [Google Scholar] [CrossRef] [PubMed]
  178. Huang, W.-C.; Hsiao, Y.-C.; Wu, C.-C.; Hsu, Y.-T.; Chang, C.-L. Less circulating mucosal-associated invariant T cells in patients with cervical cancer. Taiwan. J. Obstet. Gynecol. 2019, 58, 117–121. [Google Scholar] [CrossRef] [PubMed]
  179. Yan, J.; Allen, S.; McDonald, E.; Das, I.; Mak, J.Y.W.; Liu, L.; Fairlie, D.P.; Meehan, B.S.; Chen, Z.; Corbett, A.J.; et al. MAIT Cells Promote Tumor Initiation, Growth, and Metastases via Tumor MR1. Cancer Discov. 2020, 10, 124–141. [Google Scholar] [CrossRef]
  180. Ruf, B.; Catania, V.V.; Wabitsch, S.; Ma, C.; Diggs, L.P.; Zhang, Q.; Heinrich, B.; Subramanyam, V.; Cui, L.L.; Pouzolles, M.; et al. Activating Mucosal-Associated Invariant T Cells Induces a Broad Antitumor Response. Cancer Immunol. Res. 2021, 9, 1024–1034. [Google Scholar] [CrossRef]
  181. Kolkhir, P.; Elieh-Ali-Komi, D.; Metz, M.; Siebenhaar, F.; Maurer, M. Understanding human mast cells: Lesson from therapies for allergic and non-allergic diseases. Nat. Rev. Immunol. 2022, 22, 294–308. [Google Scholar] [CrossRef]
  182. Kumar, V.; Sharma, A. Mast cells: Emerging sentinel innate immune cells with diverse role in immunity. Mol. Immunol. 2010, 48, 14–25. [Google Scholar] [CrossRef]
  183. Abraham, S.N.; St John, A.L. Mast cell-orchestrated immunity to pathogens. Nat. Rev. Immunol. 2010, 10, 440–452. [Google Scholar] [CrossRef] [Green Version]
  184. Galli, S.J.; Nakae, S.; Tsai, M. Mast cells in the development of adaptive immune responses. Nat. Immunol. 2005, 6, 135–142. [Google Scholar] [CrossRef]
  185. Aponte-López, A.; Muñoz-Cruz, S. Mast Cells in the Tumor Microenvironment. Adv. Exp. Med. Biol. 2020, 1273, 159–173. [Google Scholar]
  186. Komi, D.E.A.; Redegeld, F.A. Role of Mast Cells in Shaping the Tumor Microenvironment. Clin. Rev. Allergy Immunol. 2020, 58, 313–325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Cabanillas-Saez, A.; Schalper, J.A.; Nicovani, S.M.; Rudolph, M.I. Characterization of mast cells according to their content of tryptase and chymase in normal and neoplastic human uterine cervix. Int. J. Gynecol. Cancer 2002, 12, 92–98. [Google Scholar] [CrossRef] [PubMed]
  188. Naik, R.; Pai, M.R.; Poornima Baliga, B.; Nayak, K.S.; Shankarnarayana; Dighe, P. Mast cell profile in uterine cervix. Indian J. Pathol. Microbiol. 2004, 47, 178–180. [Google Scholar] [PubMed]
  189. Kalyani, R.; Rajeshwari, G. Significance of mast cells in non-neoplastic and neoplastic lesions of uterine cervix. Biomed. Res. Ther. 2016, 3, 3. [Google Scholar] [CrossRef]
  190. Guo, F.; Kong, W.N.; Li, D.W.; Zhao, G.; Wu, H.L.; Anwar, M.; Shang, X.Q.; Sun, Q.N.; Ma, C.L.; Ma, X.M. Low Tumor Infiltrating Mast Cell Density Reveals Prognostic Benefit in Cervical Carcinoma. Technol. Cancer Res. Treat. 2022, 21, 15330338221106530. [Google Scholar] [CrossRef]
  191. Benítez-Bribiesca, L.; Wong, A.; Utrera, D.; Castellanos, E. The role of mast cell tryptase in neoangiogenesis of premalignant and malignant lesions of the uterine cervix. J. Histochem. Cytochem. 2001, 49, 1061–1062. [Google Scholar] [CrossRef] [Green Version]
  192. Somasundaram, R.; Connelly, T.; Choi, R.; Choi, H.; Samarkina, A.; Li, L.; Gregorio, E.; Chen, Y.; Thakur, R.; Abdel-Mohsen, M.; et al. Tumor-infiltrating mast cells are associated with resistance to anti-PD-1 therapy. Nat. Commun. 2021, 12, 346. [Google Scholar] [CrossRef]
  193. Li, J.; Peng, G.; Zhu, K.; Jie, X.; Xu, Y.; Rao, X.; Xu, Y.; Chen, Y.; Xing, B.; Wu, G.; et al. PD-1+ mast cell enhanced by PD-1 blocking therapy associated with resistance to immunotherapy. Cancer Immunol. Immunother. 2023, 72, 633–645. [Google Scholar] [CrossRef]
  194. Shamri, R.; Xenakis, J.J.; Spencer, L.A. Eosinophils in innate immunity: An evolving story. Cell Tissue Res. 2011, 343, 57–83. [Google Scholar] [CrossRef] [Green Version]
  195. Travers, J.; Rothenberg, M.E. Eosinophils in mucosal immune responses. Mucosal Immunol. 2015, 8, 464–475. [Google Scholar] [CrossRef] [Green Version]
  196. Rothenberg, M.E.; Hogan, S.P. The eosinophil. Annu. Rev. Immunol. 2006, 24, 147–174. [Google Scholar] [CrossRef]
  197. Kita, H. Eosinophils: Multifaceted biological properties and roles in health and disease. Immunol. Rev. 2011, 242, 161–177. [Google Scholar] [CrossRef]
  198. Wen, T.; Rothenberg, M.E. The Regulatory Function of Eosinophils. Microbiol. Spectr. 2016, 4. [Google Scholar] [CrossRef] [Green Version]
  199. Ravin, K.A.; Loy, M. The Eosinophil in Infection. Clin. Rev. Allergy Immunol. 2016, 50, 214–227. [Google Scholar] [CrossRef]
  200. Blanchard, C.; Rothenberg, M.E. Chapter 3 Biology of the Eosinophil. In Advances in Immunology; Academic Press: Cambridge, MA, USA, 2009; pp. 81–121. [Google Scholar]
  201. Kurose, N.; Mizuguchi, S.; Ohkanemasa, Y.; Yamashita, M.; Nakano, M.; Guo, X.; Aikawa, A.; Nakada, S.; Sasagawa, T.; Yamada, S. Adenosquamous carcinoma of the uterine cervix displaying tumor-associated tissue eosinophilia. SAGE Open Med. Case Rep. 2019, 7, 2050313X19828235. [Google Scholar] [CrossRef]
  202. van Driel, W.J.; Hogendoorn, P.C.W.; Jansen, F.-W.; Zwinderman, A.H.; Trimbos, J.B.; Fleuren, G.J. Tumor-associated eosinophilic infiltrate of cervical cancer is indicative for a less effective immune response. Hum. Pathol. 1996, 27, 904–911. [Google Scholar] [CrossRef]
  203. Höckel, M.; Schlenger, K.; Aral, B.; Mitze, M.; Schäffer, U.; Vaupel, P. Association between Tumor Hypoxia and Malignant Progression in Advanced Cancer of the Uterine Cervix1. Cancer Res. 1996, 56, 4509–4515. [Google Scholar]
  204. Xie, F.; Meng, Y.-H.; Liu, L.-B.; Chang, K.-K.; Li, H.; Li, M.-Q.; Li, D.-J. Cervical Carcinoma Cells Stimulate the Angiogenesis through TSLP Promoting Growth and Activation of Vascular Endothelial Cells. Am. J. Reprod. Immunol. 2013, 70, 69–79. [Google Scholar] [CrossRef]
  205. Grisaru-Tal, S.; Itan, M.; Klion, A.D.; Munitz, A. A new dawn for eosinophils in the tumour microenvironment. Nat. Rev. Cancer 2020, 20, 594–607. [Google Scholar] [CrossRef]
  206. Grisaru-Tal, S.; Rothenberg, M.E.; Munitz, A. Eosinophil–lymphocyte interactions in the tumor microenvironment and cancer immunotherapy. Nat. Immunol. 2022, 23, 1309–1316. [Google Scholar] [CrossRef]
  207. Steinman, R.M.; Banchereau, J. Taking dendritic cells into medicine. Nature 2007, 449, 419–426. [Google Scholar] [CrossRef]
  208. Sakref, C.; Bendriss-Vermare, N.; Valladeau-Guilemond, J. Phenotypes and Functions of Human Dendritic Cell Subsets in the Tumor Microenvironment. Methods Mol. Biol. 2023, 2618, 17–35. [Google Scholar]
  209. Carenza, C.; Calcaterra, F.; Oriolo, F.; Di Vito, C.; Ubezio, M.; Della Porta, M.G.; Mavilio, D.; Della Bella, S. Costimulatory Molecules and Immune Checkpoints Are Differentially Expressed on Different Subsets of Dendritic Cells. Front. Immunol. 2019, 10, 1325. [Google Scholar] [CrossRef]
  210. Spits, H.; Bernink, J.H.; Lanier, L. NK cells and type 1 innate lymphoid cells: Partners in host defense. Nat. Immunol. 2016, 17, 758–764. [Google Scholar] [CrossRef]
  211. Kumar, V.; Bauer, C.; Stewart, J.H. Chasing Uterine Cancer with NK Cell-Based Immunotherapies. Future Pharmacol. 2022, 2, 642–659. [Google Scholar] [CrossRef]
  212. Wolf, N.K.; Kissiov, D.U.; Raulet, D.H. Roles of natural killer cells in immunity to cancer, and applications to immunotherapy. Nat. Rev. Immunol. 2023, 23, 90–105. [Google Scholar] [CrossRef]
  213. Tu, Y.; Pan, M.; Song, S.; Hua, J.; Liu, R.; Li, L. CD3+CD56+ natural killer T cell infiltration is increased in cervical cancer and negatively correlated with tumour progression. Biotechnol. Biotechnol. Equip. 2019, 33, 1380–1391. [Google Scholar] [CrossRef] [Green Version]
  214. Céspedes, M.A.; Rodríguez, J.A.; Medina, M.; Bravo, M.; Cómbita, A.L. Analysis of NK Cells in Peripheral Blood and Tumor Infiltrating Lymphocytes in Cervical Cancer Patients. Rev. Colomb. Cancerol. 2012, 16, 16–26. [Google Scholar] [CrossRef]
  215. Gooden, M.; Lampen, M.; Jordanova, E.S.; Leffers, N.; Trimbos, J.B.; van der Burg, S.H.; Nijman, H.; van Hall, T. HLA-E expression by gynecological cancers restrains tumor-infiltrating CD8(+) T lymphocytes. Proc. Natl. Acad. Sci. USA 2011, 108, 10656–10661. [Google Scholar] [CrossRef] [Green Version]
  216. Spaans, V.M.; Peters, A.A.W.; Fleuren, G.J.; Jordanova, E.S. HLA-E expression in cervical adenocarcinomas: Association with improved long-term survival. J. Transl. Med. 2012, 10, 184. [Google Scholar] [CrossRef] [Green Version]
  217. Das, D.; Sarkar, B.; Mukhopadhyay, S.; Banerjee, C.; Mondal, S.B. An Altered Ratio of CD4+ And CD8+ T Lymphocytes in Cervical Cancer Tissues and Peripheral Blood—A Prognostic Clue? Asian Pac. J. Cancer Prev. 2018, 19, 471–478. [Google Scholar]
  218. Sheu, B.-C.; Hsu, S.-M.; Ho, H.-N.; Lin, R.-H.; Torng, P.-L.; Huang, S.-C. Reversed CD4/CD8 ratios of tumor-infiltrating lymphocytes are correlated with the progression of human cervical carcinoma. Cancer 1999, 86, 1537–1543. [Google Scholar] [CrossRef]
  219. Jiang, W.; Kang, L.; Lu, H.-Z.; Pan, X.; Lin, Q.; Pan, Q.; Xue, Y.; Weng, X.; Tang, Y.-W. Normal Values for CD4 and CD8 Lymphocyte Subsets in Healthy Chinese Adults from Shanghai. Clin. Vaccine Immunol. 2004, 11, 811–813. [Google Scholar] [CrossRef] [Green Version]
  220. Shah, W.; Yan, X.; Jing, L.; Zhou, Y.; Chen, H.; Wang, Y. A reversed CD4/CD8 ratio of tumor-infiltrating lymphocytes and a high percentage of CD4+FOXP3+ regulatory T cells are significantly associated with clinical outcome in squamous cell carcinoma of the cervix. Cell. Mol. Immunol. 2011, 8, 59–66. [Google Scholar] [CrossRef] [Green Version]
  221. Zhang, J.; Meng, S.; Zhang, X.; Shao, K.; Lin, C. Infiltration Patterns of Cervical Epithelial Microenvironment Cells During Carcinogenesis. Front. Immunol. 2022, 13, 888176. [Google Scholar] [CrossRef]
  222. Lin, W.; Niu, Z.; Zhang, H.; Kong, Y.; Wang, Z.; Yang, X.; Yuan, F. Imbalance of Th1/Th2 and Th17/Treg during the development of uterine cervical cancer. Int. J. Clin. Exp. Pathol. 2019, 12, 3604–3612. [Google Scholar]
  223. Nishimura, T.; Nakui, M.; Sato, M.; Iwakabe, K.; Kitamura, H.; Sekimoto, M.; Ohta, A.; Koda, T.; Nishimura, S. The critical role of Th1-dominant immunity in tumor immunology. Cancer Chemother. Pharmacol. 2000, 46, S52–S61. [Google Scholar] [CrossRef]
  224. van der Burg, S.H.; Piersma, S.J.; de Jong, A.; van der Hulst, J.M.; Kwappenberg, K.M.C.; van den Hende, M.; Welters, M.J.P.; Van Rood, J.J.; Fleuren, G.J.; Melief, C.J.M.; et al. Association of cervical cancer with the presence of CD4+ regulatory T cells specific for human papillomavirus antigens. Proc. Natl. Acad. Sci. USA 2007, 104, 12087–12092. [Google Scholar] [CrossRef] [Green Version]
  225. Maskey, N.; Thapa, N.; Maharjan, M.; Shrestha, G.; Maharjan, N.; Cai, H.; Liu, S. Infiltrating CD4 and CD8 lymphocytes in HPV infected uterine cervical milieu. Cancer Manag. Res. 2019, 11, 7647–7655. [Google Scholar] [CrossRef] [Green Version]
  226. Sanif, R.; Nurwany, R. Prognostic significance of CD4/CD8 ratio in patients with advanced cervical cancer. J. Phys. Conf. Ser. 2019, 1246, 012053. [Google Scholar] [CrossRef]
  227. Zhang, Y.; Yu, M.; Jing, Y.; Cheng, J.; Zhang, C.; Cheng, L.; Lu, H.; Cai, M.-C.; Wu, J.; Wang, W.; et al. Baseline immunity and impact of chemotherapy on immune microenvironment in cervical cancer. Br. J. Cancer 2021, 124, 414–424. [Google Scholar] [CrossRef]
  228. Li, L.; Xu, X.T.; Wang, L.L.; Qin, S.B.; Zhou, J.Y. Expression and clinicopathological significance of Foxp3 and VISTA in cervical cancer. Am. J. Transl. Res. 2021, 13, 10428–10438. [Google Scholar]
  229. Wu, M.-Y.; Kuo, T.-Y.; Ho, H.-N. Tumor-infiltrating lymphocytes contain a higher proportion of FOXP3+ T lymphocytes in cervical cancer. J. Formos. Med. Assoc. 2011, 110, 580–586. [Google Scholar] [CrossRef] [Green Version]
  230. Ni, H.; Zhang, H.; Li, L.; Huang, H.; Guo, H.; Zhang, L.; Li, C.; Xu, J.-X.; Nie, C.-P.; Li, K.; et al. T cell-intrinsic STING signaling promotes regulatory T cell induction and immunosuppression by upregulating FOXP3 transcription in cervical cancer. J. ImmunoTherapy Cancer 2022, 10, e005151. [Google Scholar] [CrossRef]
  231. Nedergaard, B.S.; Ladekarl, M.; Thomsen, H.F.; Nyengaard, J.R.; Nielsen, K. Low density of CD3+, CD4+ and CD8+ cells is associated with increased risk of relapse in squamous cell cervical cancer. Br. J. Cancer 2007, 97, 1135–1138. [Google Scholar] [CrossRef] [Green Version]
  232. Hong, H.S.; Mbah, N.E.; Shan, M.; Loesel, K.; Lin, L.; Sajjakulnukit, P.; Correa, L.O.; Andren, A.; Lin, J.; Hayashi, A.; et al. OXPHOS promotes apoptotic resistance and cellular persistence in TH17 cells in the periphery and tumor microenvironment. Sci. Immunol. 2022, 7, eabm8182. [Google Scholar] [CrossRef]
  233. Alves, J.J.P.; De Medeiros Fernandes, T.A.A.; De Araújo, J.M.G.; Cobucci, R.N.O.; Lanza, D.C.F.; Bezerra, F.L.; Andrade, V.S.; Fernandes, J.V. Th17 response in patients with cervical cancer. Oncol. Lett. 2018, 16, 6215–6227. [Google Scholar] [CrossRef] [Green Version]
  234. Oomizu, S.; Arikawa, T.; Niki, T.; Kadowaki, T.; Ueno, M.; Nishi, N.; Yamauchi, A.; Hirashima, M. Galectin-9 suppresses Th17 cell development in an IL-2-dependent but Tim-3-independent manner. Clin. Immunol. 2012, 143, 51–58. [Google Scholar] [CrossRef]
  235. Punt, S.; van Vliet, M.E.; Spaans, V.M.; de Kroon, C.D.; Fleuren, G.J.; Gorter, A.; Jordanova, E.S. FoxP3(+) and IL-17(+) cells are correlated with improved prognosis in cervical adenocarcinoma. Cancer Immunol. Immunother. 2015, 64, 745–753. [Google Scholar] [CrossRef] [Green Version]
  236. Yu, Q.; Lou, X.-M.; He, Y. Preferential Recruitment of Th17 Cells to Cervical Cancer via CCR6-CCL20 Pathway. PLoS ONE 2015, 10, e0120855. [Google Scholar] [CrossRef]
  237. Mauri, C.; Bosma, A. Immune Regulatory Function of B Cells. Annu. Rev. Immunol. 2012, 30, 221–241. [Google Scholar] [CrossRef]
  238. Inoue, S.; Leitner, W.W.; Golding, B.; Scott, D. Inhibitory Effects of B Cells on Antitumor Immunity. Cancer Res. 2006, 66, 7741–7747. [Google Scholar] [CrossRef] [Green Version]
  239. Sarvaria, A.; Madrigal, J.A.; Saudemont, A. B cell regulation in cancer and anti-tumor immunity. Cell. Mol. Immunol. 2017, 14, 662–674. [Google Scholar] [CrossRef] [Green Version]
  240. Kim, S.S.; Shen, S.; Miyauchi, S.; Sanders, P.D.; Franiak-Pietryga, I.; Mell, L.; Gutkind, J.S.; Cohen, E.E.W.; Califano, J.A.; Sharabi, A.B. B Cells Improve Overall Survival in HPV-Associated Squamous Cell Carcinomas and Are Activated by Radiation and PD-1 Blockade. Clin. Cancer Res. 2020, 26, 3345–3359. [Google Scholar] [CrossRef] [Green Version]
  241. Chen, Z.; Zhu, Y.; Du, R.; Pang, N.; Zhang, F.; Dong, D.; Ding, J.; Ding, Y. Role of Regulatory B Cells in the Progression of Cervical Cancer. Mediat. Inflamm. 2019, 2019, 6519427. [Google Scholar] [CrossRef]
  242. Vora, C.; Gupta, S. Targeted therapy in cervical cancer. ESMO Open. 2018, 3, e000462. [Google Scholar] [CrossRef] [Green Version]
  243. Harper, D.M.; Vierthaler, S.L.; Santee, J.A. Review of Gardasil. J. Vaccines Vaccin. 2010, 1, 1000107. [Google Scholar] [CrossRef] [Green Version]
  244. Monie, A.; Hung, C.F.; Roden, R.; Wu, T.C. Cervarix: A vaccine for the prevention of HPV 16, 18-associated cervical cancer. Biologics 2008, 2, 97–105. [Google Scholar]
  245. Grau-Bejar, J.F.; Garcia-Duran, C.; Garcia-Illescas, D.; Mirallas, O.; Oaknin, A. Advances in immunotherapy for cervical cancer. Ther. Adv. Med. Oncol. 2023, 15, 17588359231163836. [Google Scholar] [CrossRef]
  246. Shao, D.-D.; Meng, F.-Z.; Liu, Y.; Xu, X.-Q.; Wang, X.; Hu, W.-H.; Hou, W.; Ho, W.-Z. Poly(dA:dT) Suppresses HSV-2 Infection of Human Cervical Epithelial Cells Through RIG-I Activation. Front. Immunol. 2021, 11, 598884. [Google Scholar] [CrossRef]
  247. Rehwinkel, J.; Gack, M.U. RIG-I-like receptors: Their regulation and roles in RNA sensing. Nat. Rev. Immunol. 2020, 20, 537–551. [Google Scholar] [CrossRef]
  248. Shi, F.; Su, J.; Wang, J.; Liu, Z.; Wang, T. Activation of STING inhibits cervical cancer tumor growth through enhancing the anti-tumor immune response. Mol. Cell. Biochem. 2021, 476, 1015–1024. [Google Scholar] [CrossRef] [PubMed]
  249. Li, L.; Kim, S.; Herndon, J.M.; Goedegebuure, P.; Belt, B.A.; Satpathy, A.T.; Fleming, T.P.; Hansen, T.H.; Murphy, K.M.; Gillanders, W.E. Cross-dressed CD8α+/CD103+ dendritic cells prime CD8+ T cells following vaccination. Proc. Natl. Acad. Sci. USA 2012, 109, 12716–12721. [Google Scholar] [CrossRef] [Green Version]
  250. Williford, J.-M.; Ishihara, J.; Ishihara, A.; Mansurov, A.; Hosseinchi, P.; Marchell, T.M.; Potin, L.; Swartz, M.A.; Hubbell, J.A. Recruitment of CD103+ dendritic cells via tumor-targeted chemokine delivery enhances efficacy of checkpoint inhibitor immunotherapy. Sci. Adv. 2019, 5, eaay1357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  251. Salmon, H.; Idoyaga, J.; Rahman, A.; Leboeuf, M.; Remark, R.; Jordan, S.; Casanova-Acebes, M.; Khudoynazarova, M.; Agudo, J.; Tung, N.; et al. Expansion and Activation of CD103(+) Dendritic Cell Progenitors at the Tumor Site Enhances Tumor Responses to Therapeutic PD-L1 and BRAF Inhibition. Immunity 2016, 44, 924–938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Langers, I.; Renoux, V.; Reschner, A.; Touze, A.; Coursaget, P.; Boniver, J.; Koch, J.; Delvenne, P.; Jacobs, N. Natural killer and dendritic cells collaborate in the immune response induced by the vaccine against uterine cervical cancer. Eur. J. Immunol. 2014, 44, 3585–3595. [Google Scholar] [CrossRef] [Green Version]
  253. Soca Gallego, L.; Dominguez, A.; Parmar, M. Human Papilloma Virus Vaccine. In StatPearls; Statpearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  254. Chan, J.K.; Deng, W.; Higgins, R.V.; Tewari, K.S.; Bonebrake, A.J.; Hicks, M.; Gaillard, S.; Ramirez, P.T.; Chafe, W.; Monk, B.J.; et al. A phase II evaluation of brivanib in the treatment of persistent or recurrent carcinoma of the cervix: An NRG Oncology/Gynecologic Oncology Group study. Gynecol. Oncol. 2017, 146, 554–559. [Google Scholar] [CrossRef] [Green Version]
  255. Santin, A.D.; Deng, W.; Frumovitz, M.; Buza, N.; Bellone, S.; Huh, W.; Khleif, S.; Lankes, H.A.; Ratner, E.S.; O’Cearbhaill, R.E.; et al. Phase II evaluation of nivolumab in the treatment of persistent or recurrent cervical cancer (NCT02257528/NRG-GY002). Gynecol. Oncol. 2020, 157, 161–166. [Google Scholar] [CrossRef]
  256. Naumann, R.W.; Oaknin, A.; Meyer, T.; Lopez-Picazo, J.M.; Lao, C.; Bang, Y.J.; Boni, V.; Sharfman, W.H.; Park, J.C.; Devriese, L.A.; et al. Efficacy and safety of nivolumab (Nivo) + ipilimumab (Ipi) in patients (pts) with recurrent/metastatic (R/M) cervical cancer: Results from CheckMate 358. Ann. Oncol. 2019, 30, v898–v899. [Google Scholar] [CrossRef]
  257. Liu, K.; Zhu, Y.; Zhou, Y.; Zhu, H. Cemiplimab as Second-Line Therapy for Patients with Recurrent Cervical Cancer: A United States-based Cost-effectiveness Analysis. Adv. Ther. 2023, 40, 1838–1849. [Google Scholar] [CrossRef]
  258. Zheng, X.; Gu, H.; Cao, X.; Pan, B.; Xiang, H.; Ju, M.; Xu, S.; Zheng, M. Tislelizumab for cervical cancer: A retrospective study and analysis of correlative blood biomarkers. Front. Immunol. 2023, 14, 1113369. [Google Scholar] [CrossRef]
  259. Basu, P.; Mehta, A.; Jain, M.; Gupta, S.; Nagarkar, R.V.; John, S.; Petit, R. A Randomized Phase 2 Study of ADXS11-001 Listeria monocytogenes-Listeriolysin O Immunotherapy with or without Cisplatin in Treatment of Advanced Cervical Cancer. Int. J. Gynecol. Cancer 2018, 28, 764–772. [Google Scholar] [CrossRef] [Green Version]
  260. Galicia-Carmona, T.; Arango-Bravo, E.; Serrano-Olvera, J.A.; Flores-de La Torre, C.; Cruz-Esquivel, I.; Villalobos-Valencia, R.; Morán-Mendoza, A.; Castro-Eguiluz, D.; Cetina-Pérez, L. ADXS11-001 LM-LLO as specific immunotherapy in cervical cancer. Hum. Vaccin. Immunother. 2021, 17, 2617–2625. [Google Scholar] [CrossRef]
  261. Hong, D.S.; Concin, N.; Vergote, I.; de Bono, J.S.; Slomovitz, B.M.; Drew, Y.; Arkenau, H.T.; Machiels, J.P.; Spicer, J.F.; Jones, R.; et al. Tisotumab Vedotin in Previously Treated Recurrent or Metastatic Cervical Cancer. Clin. Cancer Res. 2020, 26, 1220–1228. [Google Scholar] [CrossRef] [Green Version]
  262. Song, X.; Li, R.; Wang, H.; Song, P.; Guo, W.; Chen, Z.S. Tisotumab vedotin for the treatment of cervical carcinoma. Drugs Today 2022, 58, 213–222. [Google Scholar] [CrossRef]
  263. Sherer, M.V.; Kotha, N.V.; Williamson, C.; Mayadev, J. Advances in immunotherapy for cervical cancer: Recent developments and future directions. Int. J. Gynecol. Cancer 2022, 32, 281–287. [Google Scholar] [CrossRef]
  264. Colombo, N.; Dubot, C.; Lorusso, D.; Caceres, M.V.; Hasegawa, K.; Shapira-Frommer, R.; Tewari, K.S.; Salman, P.; Hoyos Usta, E.; Yañez, E.; et al. Pembrolizumab for Persistent, Recurrent, or Metastatic Cervical Cancer. N. Engl. J. Med. 2021, 385, 1856–1867. [Google Scholar] [CrossRef]
  265. Youn, J.W.; Hur, S.Y.; Woo, J.W.; Kim, Y.M.; Lim, M.C.; Park, S.Y.; Seo, S.S.; No, J.H.; Kim, B.G.; Lee, J.K.; et al. Pembrolizumab plus GX-188E therapeutic DNA vaccine in patients with HPV-16-positive or HPV-18-positive advanced cervical cancer: Interim results of a single-arm, phase 2 trial. Lancet Oncol. 2020, 21, 1653–1660. [Google Scholar] [CrossRef]
  266. Zhang, L.; Wang, K.; Huang, Y.; Zhang, H.; Zhou, L.; Li, A.; Sun, Y. Photosensitizer-induced HPV16 E7 nanovaccines for cervical cancer immunotherapy. Biomaterials 2022, 282, 121411. [Google Scholar] [CrossRef]
  267. van Meir, H.; Nout, R.A.; Welters, M.J.P.; Loof, N.M.; de Kam, M.L.; van Ham, J.J.; Samuels, S.; Kenter, G.G.; Cohen, A.F.; Melief, C.J.M.; et al. Impact of (chemo)radiotherapy on immune cell composition and function in cervical cancer patients. OncoImmunology 2017, 6, e1267095. [Google Scholar] [CrossRef]
  268. June, C.H.; O’Connor, R.S.; Kawalekar, O.U.; Ghassemi, S.; Milone, M.C. CAR T cell immunotherapy for human cancer. Science 2018, 359, 1361–1365. [Google Scholar] [CrossRef] [Green Version]
  269. Bonini, C.; Mondino, A. Adoptive T-cell therapy for cancer: The era of engineered T cells. Eur. J. Immunol. 2015, 45, 2457–2469. [Google Scholar] [CrossRef] [Green Version]
  270. Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355. [Google Scholar] [CrossRef] [Green Version]
  271. Ellis, G.I.; Sheppard, N.C.; Riley, J.L. Genetic engineering of T cells for immunotherapy. Nat. Rev. Genet. 2021, 22, 427–447. [Google Scholar] [CrossRef]
  272. Etxeberria, I.; Olivera, I.; Bolaños, E.; Cirella, A.; Teijeira, Á.; Berraondo, P.; Melero, I. Engineering bionic T cells: Signal 1, signal 2, signal 3, reprogramming and the removal of inhibitory mechanisms. Cell. Mol. Immunol. 2020, 17, 576–586. [Google Scholar] [CrossRef]
  273. Jazaeri, A.A.; Zsiros, E.; Amaria, R.N.; Artz, A.S.; Edwards, R.P.; Wenham, R.M.; Slomovitz, B.M.; Walther, A.; Thomas, S.S.; Chesney, J.A.; et al. Safety and efficacy of adoptive cell transfer using autologous tumor infiltrating lymphocytes (LN-145) for treatment of recurrent, metastatic, or persistent cervical carcinoma. J. Clin. Oncol. 2019, 37, 2538. [Google Scholar] [CrossRef]
  274. Radvanyi, L.G. Tumor-Infiltrating Lymphocyte Therapy: Addressing Prevailing Questions. Cancer J. 2015, 21, 450–464. [Google Scholar] [CrossRef]
  275. Kazemi, M.H.; Sadri, M.; Najafi, A.; Rahimi, A.; Baghernejadan, Z.; Khorramdelazad, H.; Falak, R. Tumor-infiltrating lymphocytes for treatment of solid tumors: It takes two to tango? Front. Immunol. 2022, 13, 1018962. [Google Scholar] [CrossRef]
  276. Li, B. Why do tumor-infiltrating lymphocytes have variable efficacy in the treatment of solid tumors? Front. Immunol. 2022, 13, 973881. [Google Scholar] [CrossRef]
  277. Zheng, J.; Huang, J.; Ma, W.; Yang, W.; Hu, B.A.-O. The Antitumor Activity of CAR-T-PD1 Cells Enhanced by HPV16mE7-Pulsed and SOCS1-Silenced DCs in Cervical Cancer Models. Cancer Manag. Res. 2021, 13, 6045–6053. [Google Scholar] [CrossRef] [PubMed]
  278. He, Y.; Li, X.-M.; Yin, C.-H.; Wu, Y.-M. Killing cervical cancer cells by specific chimeric antigen receptor-modified T cells. J. Reprod. Immunol. 2020, 139, 103115. [Google Scholar] [CrossRef] [PubMed]
  279. Zhang, Y.; Li, X.; Zhang, J.; Mao, L. Novel cellular immunotherapy using NKG2D CAR-T for the treatment of cervical cancer. Biomed. Pharmacother. 2020, 131, 110562. [Google Scholar] [CrossRef] [PubMed]
  280. Tsimberidou, A.-M.; Van Morris, K.; Vo, H.H.; Eck, S.; Lin, Y.-F.; Rivas, J.M.; Andersson, B.S. T-cell receptor-based therapy: An innovative therapeutic approach for solid tumors. J. Hematol. Oncol. 2021, 14, 102. [Google Scholar] [CrossRef]
  281. Li, D.; Li, X.; Zhou, W.-L.; Huang, Y.; Liang, X.; Jiang, L.; Yang, X.; Sun, J.; Li, Z.; Han, W.-D.; et al. Genetically engineered T cells for cancer immunotherapy. Signal. Transduct. Target. Ther. 2019, 4, 35. [Google Scholar] [CrossRef] [Green Version]
  282. Ghanaat, M.; Goradel, N.H.; Arashkia, A.; Ebrahimi, N.; Ghorghanlu, S.; Malekshahi, Z.V.; Fattahi, E.; Negahdari, B.; Kaboosi, H. Virus against virus: Strategies for using adenovirus vectors in the treatment of HPV-induced cervical cancer. Acta Pharmacol. Sin. 2021, 42, 1981–1990. [Google Scholar] [CrossRef]
  283. Petry, K.U. HPV and cervical cancer. Scand. J. Clin. Lab. Investig. 2014, 74, 59–62. [Google Scholar] [CrossRef]
  284. Castellsagué, X. Natural history and epidemiology of HPV infection and cervical cancer. Gynecol. Oncol. 2008, 110, S4–S7. [Google Scholar] [CrossRef]
  285. Bhattacharjee, R.; Das, S.S.; Biswal, S.S.; Nath, A.; Das, D.; Basu, A.; Malik, S.; Kumar, L.; Kar, S.; Singh, S.K.; et al. Mechanistic role of HPV-associated early proteins in cervical cancer: Molecular pathways and targeted therapeutic strategies. Crit. Rev. Oncol./Hematol. 2022, 174, 103675. [Google Scholar] [CrossRef]
  286. Yu, S.; Li, X.; Zhang, J.; Wu, S. Development of a Novel Immune Infiltration-Based Gene Signature to Predict Prognosis and Immunotherapy Response of Patients With Cervical Cancer. Front. Immunol. 2021, 12, 709493. [Google Scholar] [CrossRef]
  287. Li, F.; Li, C.; Cai, X.; Xie, Z.; Zhou, L.; Cheng, B.; Zhong, R.; Xiong, S.; Li, J.; Chen, Z.; et al. The association between CD8+ tumor-infiltrating lymphocytes and the clinical outcome of cancer immunotherapy: A systematic review and meta-analysis. eClinicalMedicine 2021, 41, 101134. [Google Scholar] [CrossRef] [PubMed]
  288. Sugimoto, C.; Murakami, Y.; Ishii, E.; Fujita, H.; Wakao, H. Reprogramming and redifferentiation of mucosal-associated invariant T cells reveal tumor inhibitory activity. eLife 2022, 11, e70848. [Google Scholar] [CrossRef] [PubMed]
  289. Stoitzner, P.; Sparber, F.; Tripp, C.H. Langerhans cells as targets for immunotherapy against skin cancer. Immunol. Cell Biol. 2010, 88, 431–437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  290. Bellone, S.; El-Sahwi, K.; Cocco, E.; Casagrande, F.; Cargnelutti, M.; Palmieri, M.; Bignotti, E.; Romani, C.; Silasi, D.A.; Azodi, M.; et al. Human papillomavirus type 16 (HPV-16) virus-like particle L1-specific CD8+ cytotoxic T lymphocytes (CTLs) are equally effective as E7-specific CD8+ CTLs in killing autologous HPV-16-positive tumor cells in cervical cancer patients: Implications for L1 dendritic cell-based therapeutic vaccines. J. Virol. 2009, 83, 6779–6789. [Google Scholar]
  291. Lv, Y.; Ma, X.; Ma, Y.; Du, Y.; Feng, J. A new emerging target in cancer immunotherapy: Galectin-9 (LGALS9). Genes. Dis. 2022. [Google Scholar] [CrossRef]
  292. Wang, L.; Zhao, Y.; Wang, Y.; Wu, X. The Role of Galectins in Cervical Cancer Biology and Progression. BioMed. Res. Int. 2018, 2018, 2175927. [Google Scholar] [CrossRef] [Green Version]
  293. Kandel, S.; Adhikary, P.; Li, G.; Cheng, K. The TIM3/Gal9 signaling pathway: An emerging target for cancer immunotherapy. Cancer Lett. 2021, 510, 67–78. [Google Scholar] [CrossRef]
  294. Kaesler, S.; Wölbing, F.; Kempf, W.E.; Skabytska, Y.; Köberle, M.; Volz, T.; Sinnberg, T.; Amaral, T.; Möckel, S.; Yazdi, A.; et al. Targeting tumor-resident mast cells for effective anti-melanoma immune responses. JCI Insight 2019, 4, e125057. [Google Scholar] [CrossRef] [Green Version]
  295. Oldford, S.A.; Marshall, J.S. Mast cells as targets for immunotherapy of solid tumors. Mol. Immunol. 2015, 63, 113–124. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the immune-cell population in the normal adult cervix and CC TIME. (A) Immune-cell population in the normal adult human cervix. The cervix is divided into ecto- and endocervix. Endocervix is rich in mucus and glandular goblet cells. Endocervix opens in the uterus. The zone connecting the endo- and ectocervix is called the transformation zone (TZ). Ectocervix is rich in innate immune cell population (Keratinocytes, LC, DC, macrophages, and NK cells) due to its increased chances of exposure to potential pathogens. However, the T-cell population does not vary in the endo- and ectocervix. (B). Immune-cell population in the CC TIME. CC TIME supporting tumor growth and metastasis becomes immunosuppressive and TANs, TAMs (M2 macrophages), MDSCs, tDCs, and Tregs predominate it. On the other antitumor Th1 and cytotoxic CD8+ T and NK cells decrease in number. Details are mentioned in the text.
Figure 1. Schematic representation of the immune-cell population in the normal adult cervix and CC TIME. (A) Immune-cell population in the normal adult human cervix. The cervix is divided into ecto- and endocervix. Endocervix is rich in mucus and glandular goblet cells. Endocervix opens in the uterus. The zone connecting the endo- and ectocervix is called the transformation zone (TZ). Ectocervix is rich in innate immune cell population (Keratinocytes, LC, DC, macrophages, and NK cells) due to its increased chances of exposure to potential pathogens. However, the T-cell population does not vary in the endo- and ectocervix. (B). Immune-cell population in the CC TIME. CC TIME supporting tumor growth and metastasis becomes immunosuppressive and TANs, TAMs (M2 macrophages), MDSCs, tDCs, and Tregs predominate it. On the other antitumor Th1 and cytotoxic CD8+ T and NK cells decrease in number. Details are mentioned in the text.
Biology 12 00941 g001
Figure 2. HR-HPV-mediated CC immunopathogenesis. HR-HPVs infect basal epithelial cells, keratinocytes, and fibroblasts. HR-HPV infection transforms epithelial cells into neoplastic cells and later into cancer. The other keratinocytes escape from tumorigenic transformation and undergo apoptosis to support the immunosuppressive environment for tumor growth. In epithelial cells, HR-HPV infection prevents their apoptosis by inducing p53 mutation that also inhibits the cGAS/STING-mediated antitumor immune response. In addition, other proinflammatory innate immune functions of epithelial cells are also blocked, creating a chronic tumor-supportive immunosuppressive niche. Notably, in keratinocytes, only cGAS/STING-mediated NF-κB-mediated release of proinflammatory cytokines is inhibited leaving the type 1 IFN generation intact. This leads to the chronic type 1 IFN generation, which supports tumor growth. Furthermore, HR-HPV escapes phagocytosis by antigen-presenting cells (APCs, macrophages, LCs, and DCs) and antigen presentation to T and B cells. Metabolites released in the TIME by cancer cells further suppress cDC1s and NK cells. For example, DCs repolarize to tDCs in the presence of tryptophan and its metabolism by IDO. Details are discussed in the text.
Figure 2. HR-HPV-mediated CC immunopathogenesis. HR-HPVs infect basal epithelial cells, keratinocytes, and fibroblasts. HR-HPV infection transforms epithelial cells into neoplastic cells and later into cancer. The other keratinocytes escape from tumorigenic transformation and undergo apoptosis to support the immunosuppressive environment for tumor growth. In epithelial cells, HR-HPV infection prevents their apoptosis by inducing p53 mutation that also inhibits the cGAS/STING-mediated antitumor immune response. In addition, other proinflammatory innate immune functions of epithelial cells are also blocked, creating a chronic tumor-supportive immunosuppressive niche. Notably, in keratinocytes, only cGAS/STING-mediated NF-κB-mediated release of proinflammatory cytokines is inhibited leaving the type 1 IFN generation intact. This leads to the chronic type 1 IFN generation, which supports tumor growth. Furthermore, HR-HPV escapes phagocytosis by antigen-presenting cells (APCs, macrophages, LCs, and DCs) and antigen presentation to T and B cells. Metabolites released in the TIME by cancer cells further suppress cDC1s and NK cells. For example, DCs repolarize to tDCs in the presence of tryptophan and its metabolism by IDO. Details are discussed in the text.
Biology 12 00941 g002
Figure 3. Cervical cancer and Immune cells crosstalk to create and support the immunosuppressive CC TIME. Cancerous CECs with increased aerobic glycolysis (due to increased energy demand to maintain their rapid growth and proliferation) overproduce lactate. This lactate is released into the TME or TIME to create an acidic microenvironment. The acidic TME induces apoptotic cell death of different antitumor immune cells and reprograms their immunometabolism to polarize them to tumor-supportive immunosuppressive immune cells (M1 to M2 macrophages, increase in MDSCs, TANs, Tregs, and tDCs). These immunosuppressive immune cells synthesize, express, and release different tumor-promoting immunosuppressive molecules for their growth and proliferation along with supporting the cancer growth, proliferation, and metastasis. These immune cells also release angiogenic factors to support neoangiogenesis for cancer survival and metastasis. Eosinophils and mast cells release several factors (TSLP, TNF-α, histamine, and catecholamines) to support immunosuppressive CC TIME and cancer-cell proliferation and metastasis. Details are mentioned in the text.
Figure 3. Cervical cancer and Immune cells crosstalk to create and support the immunosuppressive CC TIME. Cancerous CECs with increased aerobic glycolysis (due to increased energy demand to maintain their rapid growth and proliferation) overproduce lactate. This lactate is released into the TME or TIME to create an acidic microenvironment. The acidic TME induces apoptotic cell death of different antitumor immune cells and reprograms their immunometabolism to polarize them to tumor-supportive immunosuppressive immune cells (M1 to M2 macrophages, increase in MDSCs, TANs, Tregs, and tDCs). These immunosuppressive immune cells synthesize, express, and release different tumor-promoting immunosuppressive molecules for their growth and proliferation along with supporting the cancer growth, proliferation, and metastasis. These immune cells also release angiogenic factors to support neoangiogenesis for cancer survival and metastasis. Eosinophils and mast cells release several factors (TSLP, TNF-α, histamine, and catecholamines) to support immunosuppressive CC TIME and cancer-cell proliferation and metastasis. Details are mentioned in the text.
Biology 12 00941 g003
Table 1. Currently available CC preventive vaccines and therapeutics, including monoclonal antibodies, therapeutic vaccines, and antibody–drug conjugates (ADCs).
Table 1. Currently available CC preventive vaccines and therapeutics, including monoclonal antibodies, therapeutic vaccines, and antibody–drug conjugates (ADCs).
CC Prevention StrategiesTarget
  • Cervarix (Recombinant HPV bivalent vaccine, comprising HPV16 and HPV18 L1 virus-like particles (VLPs) formulated in ASO4 (alum combined with a TLR4 ligand, MPL (3-O-desacyl-4′-monophosphoryl lipid A) adjuvant) [244]. It is used in females aged between 10–25 years and is not available in the USA.
Prevents HPV-16 and -18-associated CC via inducing immunity, including anti-HPV-16 and -18 antibodies (IgG1)
2.
Gardasil (Recombinant HPV quadrivalent vaccine, no longer available in the USA) [243] and Gardasil 9 (Recombinant HPV nonavalent vaccine, available in the USA, age of administration 9–45 years) [253]
Gardasil protects against low-risk HPV-6 and -11, which cause most genital warts, and against HR-HPV-16 and -18 for at least five years [243].
Gardasil 9 protects against infection with low-risk HPV types 6 and 11, which cause most genital warts, and against HR-HPV types 16, 18, 31, 33, 45, 52, and 58, responsible for different cancers by inducing the humoral antiviral immunity [253].
Targeted drug therapies for CC
  • Bevacizumab (Avastin, Alymsys, Avastin, Mvasi, or Zirabev) is USFDA approved and used in combination with chemotherapies, including paclitaxel and either cisplatin or topotecan hydrochloride [242]
Targets human vascular endothelial growth factor (VEGF) to inhibit angiogenesis or neoangiogenesis in CC
2.
Brivanib, currently under evaluation (phase II trial) to target advanced CC [242,254]
Targets VEGF and fibroblast growth factor receptor (FGFR)
3.
Pembrolizumab (Keytruda), a USFDA-approved immune checkpoint inhibitor (ICI) [242]
Targets PD-1 on T cells to prevent their exhaustion that increases anticancer immunity to clear CC cells
4.
Nivolumab, under phase II clinical trial. It has low antitumor activity and an acceptable safety profile in patients with persistent/recurrent CC [255]
Targets human PD-1
5.
Ipilumab and Nivolumab combination in recurrent/metastatic CC patients [256]
Blocks PD-1–PD-L1 interaction to enhance the antitumor immunity
6.
Cemiplimab, a second-line therapy for patients with persistent/recurrent CC [257]
Targets human PD-1 to block PD-1–PD-L1 interaction to prevent immune exhaustion
7.
Tislelizumab, approved by China’s National Medical Products Administration and under USFDA review for different solid cancers. It has also shown beneficial antitumor activity and tolerable toxicity in patients with recurrent/metastatic CC [258]
Targets human PD-1 to inhibit PD-1–PD-L1 interaction and minimizes binding to Fcγ receptors to serve as an ICI.
8.
Axalimogene filolisbac (ADXS11-001), a live, attenuated Listeria monocytogenes bacterial vector secreting HPV-16 E7 fused to listeriolysin O (LLO), a therapeutic vaccine in patients with recurrent/refractory CC patients [259,260]
Raises anti-HPV-16 cellular immunity, including cytotoxic T cell-mediated immune response
9.
Tisotumab vedotin-tftv (Tivdak), An USFDA approved antibody–drug conjugate (ADC) for recurrent/metastatic CC [261,262]
This ADC delivers cytotoxic agent monomethyl auristatin E (MMAE) directly into tumor cells to target highly expressed tissue factor (TF) or conjugation factor III in CC
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kumar, V.; Bauer, C.; Stewart, J.H., IV. TIME Is Ticking for Cervical Cancer. Biology 2023, 12, 941. https://doi.org/10.3390/biology12070941

AMA Style

Kumar V, Bauer C, Stewart JH IV. TIME Is Ticking for Cervical Cancer. Biology. 2023; 12(7):941. https://doi.org/10.3390/biology12070941

Chicago/Turabian Style

Kumar, Vijay, Caitlin Bauer, and John H. Stewart, IV. 2023. "TIME Is Ticking for Cervical Cancer" Biology 12, no. 7: 941. https://doi.org/10.3390/biology12070941

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