Open access peer-reviewed chapter

Molecular and Cell Biology of Cervical Cancer

Written By

Natalia Garcia-Becerra, Carlos A. Garcia-Becerra, Leonardo Fernandez-Avila, Jose Roberto Cruz-Lozano, Veronica Soltero-Molinar, Isabel Arias-Gallardo, Sofia Briseida Leyva-Delgado, Angel E. Chávez-Torres, Dalia I. Murillo-Geraldo and Jesús E. Juarez-Garcia

Submitted: 29 June 2023 Reviewed: 16 July 2023 Published: 21 December 2023

DOI: 10.5772/intechopen.1002395

From the Edited Volume

Cervical Cancer - Recent Advances and New Perspectives

Michael Friedrich

Chapter metrics overview

58 Chapter Downloads

View Full Metrics

Abstract

The molecular and cell biology of cervical cancer will be covered in detail in this chapter, particularly emphasizing the disease’s etiology, brief epidemiology, risk factors, cervical cancer hallmarks, and the main signaling pathways involved. The chapter will go in-depth about the characteristics of cancer, such as changes in cell cycle regulation, apoptosis, and cell differentiation, as well as the tumoral microenvironment. Signal pathways like the PI3K/AKT/mTOR pathway and the Wnt/beta-catenin pathway will be highlighted for their significance in the development of cervical cancer. The chapter will thoroughly explain the molecular and cell biology underlying this terrible illness.

Keywords

  • cervical cancer
  • molecular biology
  • cancer biology
  • cancer cells
  • tumor niche
  • tumor microenvironment
  • cancer signaling
  • hpv
  • cancer hallmarks

1. Introduction

Cervical cancer (CC) is a cellular alteration that originates in the cervix due to the persistent infection of oncogenic genotypes of the human papillomavirus (HPV) and initially manifests itself through precancerous lesions of slow and progressive evolution [1].

CC progression takes around 20 years to generate an invasive carcinoma; it begins with the precancerous lesions called cervical intraepithelial neoplasias (CIN) [2]. It is estimated that around 30% of women with advanced CIN premalignant lesions who do not receive treatment can progress to CC [3].

CC is recognized as a significant health burden in low- and middle-income countries, where it is ranked as the fourth most common type of female cancer [4, 5] and the second leading cause of cancer death in women worldwide [6]. In 2020, there were an estimated 604,000 new cases of cervical cancer and 324,000 deaths worldwide, with almost 90% of these cases occurring in low- and middle-income countries [6]. CC prevention is derived from vaccination schemes against HPV, the use of condoms, and timely detection of precancerous lesions through cervical cytology and HPV detection tests [7].

In October 2019, the World Health Organization (WHO) reported that 124 countries already had a timely and free vaccination program for 10-year-old girls. However, this is still insufficient to significantly reduce CC cases, given that the distribution of vaccines barely covers 30% of the world population, and screening schemes are not usually applied in all health sectors in low- and medium-income countries [8]. Because of this situation, in 2020, the WHO committee supported the Global Strategy towards the Elimination of Cervical Cancer, and it marked a significant milestone as it became the first-ever elimination strategy for a cancer in the history of the WHO.

The strategy sets forth three specific global targets aimed at preventing and treating CC; by 2030, 90% of girls should receive complete HPV vaccination before reaching 15 years of age; screening using a high-performance test should be conducted on 70% of women by age 35 and again by age 45; and 90% of identified CC patients should receive the suitable treatment [9]. These targets serve as measurable indicators to gauge progress in preventing and managing cervical cancer globally.

Known risk factors for developing CC are HPV, low socioeconomic status, smoking, young age at first intercourse, unprotected sexual intercourse, polygamy, long-term use of hormonal contraceptives, and multiple births [7, 10, 11, 12, 13]. However, it has been shown that the most critical risk factor for CC development is persistent infection by high-risk HPV genotypes, which is essential for cell transformation and can be detected in 99.7% of CC cases [14, 15].

HPVs belong to the Papillomaviridae family, which are small, non-enveloped, double-stranded DNA viruses of about 50–60 nm diameter [16]. HPVs possess tropism for mucosal and cutaneous keratinocytes, and according to Papillomavirus Episteme, there are over two hundred genotypes that can be subclassified according to their oncogenic potential as high-risk and low-risk.

Low-risk HPVs, such as genotypes 6, 11, 42, 43, and 44, are classified as such because they commonly cause only benign epithelial lesions, such as warts and papillomas. On the other hand, high-risk genotypes, including 16, 18, 31, 33, 34, 39, 45, 52, 53, 58, 68, and 70, are strongly linked to the development of CC [17, 18]. Genotypes 16 and 18 are considered of the most significant clinical relevance due to their association with approximately 70% of the CC cases worldwide, in addition to their great oncogenic potential [19, 20].

The oncogenic nature of high-risk HPV is attributed to the activity of E6 and E7 oncoproteins, which are the only ones that remain active even when the transformation process of cervical cells has already begun [21]. Continuous expression of the E6 and E7 oncoproteins is essential to initiate and sustain the transformation of infected cells since they activate immunological and carcinogenic pathways to favor tumorigenesis and modulate local immunity [22, 23].

Advertisement

2. Oncogenes, tumor suppressor-genes, and mutations

Cancer develops due to a series of events at the molecular level triggered by genetic and epigenetic changes, which in turn modify the normal cellular biological behavior [24, 25, 26]. The molecular events are associated with the altered function of specific genes classified as proto-oncogenes and tumor suppressor genes (TSG); these maintain a balance among the normal cell life cycle, growth, and proliferation with programmed cell death (apoptosis) [26].

The transition from proto-oncogenes (genes with normal biological functions in cell homeostasis) to oncogenes (mutated proto-oncogenes with a gain in their normal biological function) is considered one of the first molecular events that trigger subsequent carcinogenesis [24, 25]. Changes in function from proto-oncogenes to oncogenes allow the appearance of key signs of cancer in a cell population, such as loss of apoptosis, uncontrolled cell multiplication, angiogenesis, and metastasis to distant organs [26].

Another fundamental molecular transformation for carcinogenesis is the loss of function of TSG, which is the primary antagonistic mechanism of carcinogenesis by maintaining cell homeostasis, promoting the repair of cell damage, and inducing apoptosis when this repair fails to occur [24, 26]; it has been hypothesized that the loss of function of both alleles of these genes is necessary to cause a phenotypic change; this is known as Knudson’s phenomenon or the “two-hit theory” [27].

2.1 Cervical cancer and tumor suppressor genes

A sequence of steps in carcinogenesis onset related to high-risk HPV has been hypothesized in CC, presumably beginning with the decrease in the activity of tumor suppressor genes [26].

HPV infects the basal cells of the cervical epithelium, using the normal cycle of epithelial cells, which replicate in the basal region (where the virus replicates minimally), migrate to the apical region, and specialize (where viral replication increases) [28, 29, 30]. The process of carcinogenesis secondary to infection entails a process of several decades of persistent or repetitive infection, in which, also accidentally, the viral genome integrates into the host genome [26, 28, 30, 31], which is not part of the normal life cycle of the virus [30].

Upon integration into the genome, various molecular processes take place. These processes have two outcomes: Firstly, they interfere with the E2 gene from inhibiting the E6 and E7 viral oncogenes [32, 33], and secondly, they contribute to an increase in genomic instability of neighboring sections of the host DNA. This instability is caused by mechanisms such as insertion into fragile regions, rearrangement or duplication of contiguous genes, and the specific way viral genes are inserted [30, 31, 34]. Regarding TSG, E6 induces ubiquitin-mediated degradation of p53 protein, so its protective action against genetic damage is inhibited [22, 28]; E7 interacts with the LXCXE motif segment of the amino-terminal end of the Rb family of proteins (Rb, p110, p130, and p107 mainly), suppressing them and allowing the unopposed expression of E2F transcription factor, progressing from G1 to S phase in the cell cycle (Figure 1) [22, 28].

Figure 1.

Schematic representation of the main action of E6 and E7 viral oncoproteins on tumor suppressor genes. E6 blocking the function of the tp53 protein and inducing its degradation. E7 blocking the function of the RB protein and inducing its degradation.

2.2 Proto-oncogenes and oncogenes

Once genomic instability and cell immortalization are established by mechanisms that will be described further, it is considered that oncogenesis continues with the transition from proto-oncogenes to oncogenes. One of the main pathways described in more than 90% of cells with CC is mediated by PI3K/Akt [35], which promotes mechanisms of cellular proliferation; the activation of this pathway has been described secondary to the suppression of the PTEN gene [35] or to the overexpression of PI3KCA [25].

Other important oncogenes related to the proliferation of CC are the c-myc transcription factor, the ERB2 tyrosine kinase receptor, and the “HaRAS” oncogene [35].

Advertisement

3. Cell survival and immortality in cervical cancer

In CC, cancer cells survive by avoiding apoptosis and acquiring the capacity to replicate indefinitely. These characteristics and other hallmarks of cancer allow its uncontrolled proliferation and the formation of tumor masses in the cervix [36]. Apoptosis is a crucial physiological mechanism that limits the expansion of the cell population, to maintain tissue homeostasis by eliminating aged cells or cells that have outlived their useful life and allowing cell renewal or by eliminating potentially malignant cells that are experiencing irreparable damage caused by internal or external factors [37].

It is thus a well-recognized fact that resistance to apoptosis is a hallmark of cancer in general, a requirement for the persistence of transformed cells that, through various strategies, manage to evade it [36].

Nevertheless, what allows CC cells to overcome apoptosis? Aforesaid, HPV infection is a necessary and predisposing factor for developing and progressing CC. Interestingly, HPV E6 and E7 oncoproteins have been shown to enable cancer cells to escape apoptosis by disrupting or modifying pathways that precipitate this mechanism and that are finely regulated by pro- and anti-apoptotic proteins [38]. The ubiquitin-mediated degradation of p53 by E6 oncoprotein prevents cell cycle arrest and, ultimately, the apoptotic machinery, mediated in part by transcriptional activation of BAX and PUMA [39]. Given this fact, it is worth mentioning that the presence of E6 leads to a significant increase in the activity of the survivin promoter, a protein that prevents apoptosis. Survivin expression is negatively regulated by p53, and since E6 negatively regulates p53, it is likely that it also influences the regulation of survivintranscription. This suggests that the survivin gene is relevant to the anti-apoptotic function of E6. In addition, E6 can also bind directly to BAK protein, causing its degradation, thus blocking the intrinsic or mitochondrial pathway of apoptosis. The binding capacity of E6 to this protein is similar in HPVs considered high or low risk, suggesting that the inactivation of BAK is essential for the virus replication cycle [40]. E6 also prevents an extrinsic receptor-mediated response to apoptosis. In vitro studies have shown that E6 protein protects cells from TNF-mediated death through a p53-independent mechanism. It has been observed that E6 binds to a specific part of the TNF-R1 receptor, preventing apoptotic signal transduction [41]. Likewise, it has been pointed out that E6 can bind to the DED domains of the adaptor protein FADD and pro-caspase 8, critical elements of the extrinsic pathway, stimulating their degradation [42]. These findings show that E6 promotes survival by exerting multiple effects that inhibit apoptosis.

Regarding E7 oncoprotein, its role in cell cycle has been previously described; however, concerning apoptosis, it plays a dual role; it can induce or inhibit this process depending on its interaction with different proteins and mechanisms and significantly depending on the HPV present [43].

Moreover, considering that the E5 oncoprotein does not usually receive much attention due to its elimination at later stages of infection, it is essential to mention that its presence in early stages is crucial for survival and propagation of the virus in the cervical epithelium [44]. It modulates the epidermal growth factor signaling pathway, which induces the degradation of BAX, which is essential for triggering the intrinsic apoptosis pathway [45]. A relationship has also been found between its presence and the downregulation of death receptors such as CD95 in cervical tumors leading to impaired apoptosis [46].

Given that CC cells have effectively evaded the apoptosis mechanism, it is essential to mention that this enables them to achieve immortality, that is, an unlimited replication potential, thus contributing to the development and progression of cancer. Usually, as cells replicate, the ends of the chromosomes that protect the DNA during cell division, called telomeres, shorten with each successive cycle. This is part of cellular aging. However, in cancer cells, it is necessary to prevent the telomeres from shortening so the tumor can continue growing [47].

Telomerase is an enzyme responsible for telomere replication and is overexpressed in cancer cells but inactive in healthy cells [48]. In this regard, it has already been reported that HPV E6 and E7 persistently drive the expression of a catalytic unit in telomerase called hTERT, which gives these cells unlimited replicative capacity [49]. This is possible because E6 and E7 activate the hTERT promoter with the help of proteins such as c-myc and Sp1 that act as positive regulators and NFX1. NFX1 normally represses hTERT expression but is degraded by E6/E6AP, which activates the hTERT promoter [50].

Advertisement

4. Main signaling pathways (JAK/STAT, Ras/MEK/Erk, PI3K/Akt/mTOR, WNT/b-catenin)

Giving continuity to the critical regulation of external and internal factors on cell function, space is given to understand the signaling that makes this possible at the molecular level. Below are four signaling pathways relevant to the development of CC since their inappropriate regulation allows the cell to experience anabolic, proliferative, growth-promoting, and cell-survival effects.

4.1 Janus Kinase/Signal Transducer and Transcription Activator (JAK/STAT) signaling pathway

The Janus Kinase/Signal Transducer and Transcription Activator (JAK/STAT) signaling pathway is characterized by its physiological contribution to cell proliferation, differentiation, and death by causing a final gene transcription reaction in the cell nucleus. Its dysfunction can cause alterations in immune regulation and tumor processes [51].

The sequence of events begins with the interaction of cytokines and growth factors with their respective receptors, types I and II, on the cell membrane. The intracellular portion of the receptor interacts with the inactive JAK [52], which undergoes dimerization and oligomerization [53]. In both cases, a conformational change is induced in the cytoplasmic domain [52]. The described effect is the juxtaposition of JAKs for their phosphorylation and transphosphorylation by other JAKs or other families of tyrosine kinases.

Activation of JAKs phosphorylates the cytoplasmic domain, creating a binding site for other signaling molecules such as STAT proteins [51]. Cytoplasmic STAT binds to phosphorylated receptors, thus becoming substrates to be phosphorylated by JAKs. After phosphorylation, STATs form homodimers or heterodimers capable of translocating to the nucleus and activating gene transcription [23, 51]. Regulators of this pathway have been identified as suppressor signaling cytokines (SOCS), activated protein inhibitors of STAT (PIAS), and protein tyrosine phosphatases (PTP) [54].

4.2 Mitogen-activated protein kinase (MAPK) signaling pathway

Similarly, the dysfunction and incorrect regulation of the mitogen-activated protein kinase (MAPK) signaling cascade has been linked to carcinogenic events since it is also involved in cell proliferation, survival, differentiation, and migration [5556]. This cascade begins with stimuli, including growth factors, tumor-promoting substances, and differentiation factors. There is a stimulation of Ras-GDP to convert to Ras-GTP, resulting in its activation resulting in its activation and subsequent Raf phosphorylation. In turn, Raf is responsible for the phosphorylation of MEK1. MEK1 phosphorylates ERK, an extracellular receptor kinase responsible for regulating cytosolic proteins, transcription factors, and metastasis [55, 56].

4.3 PI3K/AKT/mTOR signaling cascade

The PI3K/AKT/mTOR signaling cascade also affects energy metabolism. The negative effects related to CC lie in the contribution to the creation and proliferation of malignant cell phenotypes [55, 56]. The cascade begins with a lipid kinase, PI3K, activated by the effect of extracellular stimuli recognized by receptors associated with G proteins (GPCRs) or receptor tyrosine kinases (RTKs), also using Ras-GTPases [55, 56]. The stimuli in question are given by growth factors or cytokines, which cause receptor dimerization and transphosphorylation. Once the receptor is phosphorylated, it is ready to bind to and activate proteins, in this case, PI3K. Activated PI3K produces PIP3, from PIP2, and PI(3,4)P2. PIP3, in turn, attracts the AKT molecule to the cell membrane, which is then phosphorylated and activated by PDK1 and mTORC2 [57]. AKT is responsible for activating mTORC1, the molecule responsible for the anabolic effects in the cell. This signaling cascade is important in regulating cell functions such as cell growth, motility, survival, metabolism, and angiogenesis [55, 56]. Dysfunction of this cascade, particularly its overactivation, is implicated in carcinogenesis.

4.4 WNT/beta-catenin signaling pathway

Lastly, the fourth relevant signaling pathway in the pathogenesis of CC is Wnt/beta-catenin, also called canonical. Wnt is proteins incorporated into exosomes for transport and have target-specific genes that can be transcribed through beta-catenin stabilization [58]. Beta-catenin in its inactive state is bound to the so-called destruction complex consisting of Axin, CKI, GSK3, APC, Dvl (or Dsh), and Beta-TrCP (Figure 2), which regulates it by degradation [59]. Beta-catenin phosphorylation serves as a stimulus for beta-TrCP to ubiquitinate it, resulting in its proteasomal. The effects of beta-catenin can be evidenced only by avoiding its degradation when its levels in the cytosol are high.

Figure 2.

Schematic representation of the molecular components taking part in the Wnt/beta-catenin pathway.

The beginning of the cascade occurs when Wnt activates its receptor complex formed by the Frizzled receptor and co-receptor LRP5/6; this induces the phosphorylation of LRP6 by CK1 and GSK3, allowing the translocation of the destruction complex from the cytosol to the membrane [58]. This process activates the Dvl (Dsh) component, which in its active state induces the sequestration or degradation of Axin in such a way that it inhibits the destruction complex by stabilizing cytosolic beta-catenin [59]. Wnt has target genes within the DNA in the nucleus and requires a molecule called TCF for its transcription. In the inactive Wnt state, TCF in the nucleus is inhibited by Groucho, preventing it from binding to DNA and starting gene transcription [60, 61]. In its active state, Wnt triggers the steps mentioned above, culminating in the increase in intracellular beta-catenin. As beta-catenin accumulates in the cytosol, it enters the nucleus, removes Groucho from its inhibitory position, and binds to TCF [60, 61]. This accumulation leads to the transcription of Wnt target genes. The effects of Wnt/beta-catenin signaling in CC promote epithelial-mesenchymal transition, migration, growth, and cell proliferation [62].

The beginning of the cascade occurs when Wnt activates its receptor complex formed by the Frizzled receptor and co-receptor LRP5/6; this induces the phosphorylation of LRP6 by CK1 and GSK3, allowing the translocation of the destruction complex from the cytosol to the membrane [58]. This process activates the Dvl (Dsh) component, which in its active state induces the sequestration or degradation of Axin in such a way that it inhibits the destruction complex by stabilizing cytosolic beta-catenin [59]. Wnt has target genes within the DNA in the nucleus and requires a molecule called TCF for its transcription. In the inactive Wnt state, TCF in the nucleus is inhibited by Groucho, preventing it from binding to DNA and starting gene transcription [60, 61]. In its active state, Wnt triggers the steps mentioned above, culminating in the increase in intracellular beta-catenin. As beta-catenin accumulates in the cytosol, it enters the nucleus, removes Groucho from its inhibitory position, and binds to TCF [60, 61].

This accumulation leads to the transcription of Wnt target genes. The effects of Wnt/beta-catenin signaling in CC promote epithelial-mesenchymal transition, migration, growth, and cell proliferation [62].

Advertisement

5. Cell cycle alterations and genomic instability

Viral infections are related to about 9.9% of cancers worldwide as they contribute to the oncogenesis of host cells [63]. The transformative process of the HPV-infected cell could be classified as a multifactorial process in its virulence mechanisms, which ensure the survival and proliferation of the virus at the expense of the integrity of the human genome, implying a loss of genomic stability through a mutation or an oncogenic exposure, thus being the center point of cancer [36].

The intrinsic HPV proteins that are especially important in the transformative process of the cell are initially the E1 and E2 proteins and the E6 and E7 [64]. The process begins with the integration of the viral genome through the E1 helicase and the E2 binding protein; likewise, E1 facilitates the recruitment of DNA damage response (DDR) mechanisms, a set of proteins that signal the locations of damage to its repair and the signaling response by the ATM, which occurs in the event of a break in the double chain [65, 66]. Added to the expression of E1 and E2, it causes the formation of free radicals that damage DNA, causing changes in guanine to tyrosine kinase [67].

Finally, once the viral genome integrates with the host’s DNA, the hybrid gene presents alterations such as duplications, deletions, translocations, and inversions. Likewise, the integration of the viral genome itself can theoretically act as a de novo mutation and can promote recombination errors [64, 68]. The integration of the viral genome in a cell previously altered by HPV infection causes the E2 protein to be truncated or removed, and this causes overexpression of E6 and, therefore, of its genes, which derives into p53 degradation, as mentioned previously [64]. The E6 and E7 oncoproteins target p53 and Rb, respectively, predisposing the cell to remain in the proliferative amplification stage without the possibility of exiting the cell cycle, which leads to the accumulation of mutations. However, E7 individually can transform a cell into a cancer cell [69].

Ubiquitin-mediated degradation of p53 by E6 promotes genomic instability and the subsequent birth of cancer [64], since the absence of p53 activity allows the cell to proceed to the S phase of the cell cycle without having repaired the damaged areas of the DNA [21].

The progression from G1 to S without previous DNA revision and repair results in the accumulation of errors in the chromosomes [64]. E7 is the primary mediator for DDR activation, which is localized to the junction foci of the viral genome with the host genome. It is theorized that this DDR hijacking prevents the repair of further DNA damage [70]. In response to DDR, targeted transduction of the viral genome mediated by homologous recombination reduces the capacity to repair DNA double-strand breaks by 50% [71].

Other pro-carcinogenic activities of the E6 and E7 oncoproteins are the association with centrosome amplification and shortening, increasing cell cycle defects [7172]. This process drives the proliferation of infected cells from infected keratinocytes [2273]. The effect of E6 and E7 on telomeres causes lengthening and sometimes shortening capable of driving arrangements such as bridging in anaphase and “breakage-fusion-bridge” cycles or replication indefinitely [64, 74]; in CC, 64% of the cases show a high expression of alteration in telomeres [64, 75].

Advertisement

6. Metabolic switch

The “metabolic switch” or deregulation of cellular metabolism is considered one of the hallmarks of cancer and refers to the metabolic mutation that tumor cells present with an affinity for “aerobic” glycolysis to obtain energy and favor the synthesis of macromolecules for the cellular replacement [76, 77, 78], which will be explained in detail later.

Under normal conditions, healthy body cells in the presence of oxygen obtain energy through aerobic metabolism, that is, the citric acid cycle or Krebs cycle, where carbohydrates, amino acids, and lipids are metabolized [77]. Due to the oxidations in this cycle, three molecules of nicotinamide adenine dinucleotide (NAD) and one of flavin adenine dinucleotide (FAD) are produced (generating 2.5 ATP each NADH and 1.5 ATP each FADH) [77]. Those molecules (NADH and FADH) subsequently enter the respiratory chain and oxidative phosphorylation, resulting in a total of up to 36 ATP molecules through these aerobic routes [77].

Additionally, there is glycolysis, a metabolic pathway that occurs in normal cells without oxygen, where glucose is catabolized, obtaining 2 ATP molecules and two pyruvate molecules for each glucose molecule [79]. Both routes end with the same goal: to satisfy the energy needs of the tissues.

Most malignant cells alter these metabolic pathways to obtain energy (Figure 3), presenting greater glucose uptake compared to normal cells and metabolizing it through glycolysis, most of which is converted into lactate even in the presence of a high oxygen level. This metabolic deregulation is known as the Warburg Effect, where Otto Warburg postulated that malignant cells obtained their energy from “aerobic” glycolysis [76]. This restructuring favors and fosters an environment suitable for the use of cancer cells [76, 78]. Although glycolysis is less efficient to produce enough ATP molecules, cancer cells alter metabolic pathways using glycolysis as the main pathway and oxidative phosphorylation to a lesser extent, resulting in a fast production of ATP to meet the exuberant energy demands of cancer cells during their proliferation [80].

Figure 3.

Comparison of cancer cell metabolism vs. normal cell metabolism, both in the presence of oxygen.

In glycolysis, there is a leak of substrates useful for other metabolic routes with intermediate products that will serve for the subsequent synthesis of proteins, nucleic acids, and lipids that are fundamental for the development and division of malignant cells [81, 82]. In turn, this dysregulation prompts cellular adaptation to metabolic stress and protects cancer cells from reactive oxygen species (ROS)-related damage [82].

The specific role of HPV occurs in the microenvironment after the infection by this virus helps to evade the immune response and promote local immunosuppression [80]; in addition, it contributes to the restructuring of the metabolism of infected cells in the immune microenvironment. There is evidence of metabolism changes related to glutamine, taurine, and lysine in positive cases of HPV, which are related to the interaction of this pathogen and normal cellular metabolism [83]. Also, E6 and E7 may favor the Warburg effect by contributing to chemoresistance [29, 84].

When lactate accumulates in abnormal cells, the acid environment facilitates the deregulation of normal metabolism and signaling pathways of immune cells such as dendritic cells, macrophages, and T lymphocytes, producing the immune response. While all these changes are favorable to continue viral propagation and the persistence of the malignant cellular process, T lymphocytes are forced to change by such a high lactate environment, decreasing their functionality [80].

Advertisement

7. Cervical cancer and immune system

CC has two essential elements for its establishment and progression: the parenchyma, comprised of cancer cells, and the stroma, which includes connective tissue, blood vessels, extracellular matrix (ECM), nutrients, and the immune system [77]. These elements contribute through three situations: immune escaping, angiogenesis activation, and tumor progression (proliferation, invasion, and metastasis) [85].

The tumor microenvironment (TME) is defined as a complex ecosystem surrounding a tumor inside the body; the TME is in a constant battle between a tumor suppression response and a tumor-promoting response [86].

Cancer cells represent the core of the TME, and they are responsible for manipulating the cell and non-cell components through signaling mechanisms to take advantage of the non-tumoral cells to promote carcinogenesis and metastasis [87].

Intercellular communication is generated by the synthesis of cytokines, chemokines, growth factors, and inflammatory mediators, among others, but novel mechanisms recently reported include cell-free DNA, exosomes, circulating tumor cells, and apoptotic bodies [88, 89, 90, 91].

Near the tumoral cell core, cancer-associated fibroblasts (CAFs) are located; these populations play a crucial role in carcinogenesis as they promote the proliferation, migration, and survival of cancer cells [91]; however, it is essential to acknowledge that as a heterogeneous cell population, some are related to antitumoral activities. In the tumoral niche, some CAFs promote the recruitment of immunosuppressive cells through the synthesis of ECM proteins [92], and others promote angiogenesis by producing fibroblast growth factor 2 (FGF2) and vascular endothelial growth factor A (VEGFA); they also offer nutrients to tumoral cells such as ketone and cytokines necessary for mitochondrial biogenesis and autophagy [93].

Among the non-malignant cells located at TME are the tumor-infiltrating lymphocytes (TILs), which comprise one of the primary mediators of the dynamic yet ambiguous immune response. This group comprises T cell subsets such as CD4+, CD8+, regulatory T cells (Tregs), and Natural Killer (NK) cells [94]. Even though most of these cell populations are specialized in antitumoral functions, the overall action is insufficient to eradicate the tumor effectively due to the low levels of antitumoral cells, and the TME promotes an immuno-suppressor stimulus that induces a senescent state among those cells. In CC, the presence of cytotoxic CD8+ T cells activated by tumoral antigens is recognized as an excellent prognostic marker due to their killing activities and suppression of angiogenesis through IL-2 and IFN-γ secretion. On the other hand, CD4+ T cells coordinate an ambiguous immune response; Th1 cells synthesize potent modulators of cell-mediated immune responses [95]. However, recent studies have demonstrated that CIN or CC patients had a lower proportion of Th1 subtype and a higher proportion of Th2, Th17, and Treg cells when compared against healthy controls, and this imbalance aggravates along with the progression of the disease [96].

Finally, it is essential to mention the role of macrophages since they represent one of the significant components of tumor infiltrates and are responsible for producing high amounts of inflammatory molecules, IL-1𝛽, IL-6, IL-23, and TNF-𝛼, ROS, hypoxia-induced factor (HIF), necessary for inflammatory processes [97]. At the TME, macrophages are identified as Tumor-Associated Macrophages (TAMs) and are often associated with tumor progression and worsening of CC patients. TAMs can polarize into two phenotypes, M1 macrophages that release inflammatory factors, promote immune responses, and inhibit the CC occurrence [98], and the M2 phenotype, which is correlated with poor prognosis, chemoresistance, and diminished patient survival [99].

The growing evidence demonstrates the relevance of the TME in CC progression since the interaction between tumoral cells and their surroundings promotes their proliferation, resistance to apoptosis, chemoresistance, aggressiveness, and immune evasion. Thus, TME is crucial in determining CC patients’ therapeutic response and clinical outlook.

Advertisement

8. Angiogenesis

Angiogenesis is the formation of new blood vessels as an essential mechanism for the growth, survival, and spread of solid tumors [100]. This process not only occurs in neoplasias but also is involved in the growth of the endometrium and fetal development, among other processes [101, 102].

For the formation of new vessels, in tumor development, there is an imbalance between stimulating and inhibitory factors, where there is an increase in angiogenic factors such as vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), platelet-derived growth factor (PDGF), and angiopoietins (Ang), which are produced by the same tumor cells, macrophages, and lymphocytes that may be attached to the tumor [103, 104]. In addition, a decrease in antiangiogenic agents such as Thrombospodin-1 (TSP-1) is detected, allowing the transition from an avascular phase to a vascular phase, also called “angiogenic switch” [105, 106, 107].

This imbalance begins in the tumor cells, where there are areas with little irrigation and, therefore, low oxygenation; this tumoral tissue starts producing HIF1, which is a regulator of the recruitment of endothelial progenitor cells (EPC), pericyte progenitor cells (PPC), and monocytes, and thus carries out vascular restructuring; it also increases VEGF activity [108, 109, 110].

VEGF, the major angiogenic factor, consists of a family of proteins, VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-E, and the placental growth factor. These proteins interact with three tyrosine kinase receptors, with VEGFR1 and VEGFR2, to trigger the signaling cascade promoting migration, proliferation, and survival of endothelial cells, also increasing the permeability of existing blood vessels, and this allows the leakage of multiple plasma proteins and the formation of new vessels (Figure 4). VEGF also inhibits apoptosis of newly formed blood vessels [111, 112].

Figure 4.

Schematic representation of the intracellular response to VEGF stimulation, in tumor angiogenesis.

FGF-1 and FGF-2 are also important angiogenic factors; FGF-2 increases the expression of other angiogenic agents, such as VEGF, and regulates the balance of Ang, leading to a predominance of Ang-2 [113, 114, 115]. On the other hand, Ang-2 is secreted by endothelial cells at sites of active vascular remodeling and engages in tumor initiation. Multiple studies indicate that the imbalance of Ang-2 and Ang-1 is associated with vascular instability, a key point in the initiation of angiogenesis in tumors [115, 116].

In recent years, efforts have been made to inhibit the signaling cascade in CC as a possible therapeutic target. There are several drugs in use, such as bevacizumab, which is a drug that inhibits the formation of new vessels by neutralizing VEGF activity, among others. However, these drugs must be in conjunction with chemotherapy or radiation therapy [117, 118, 119, 120].

Advertisement

9. Epithelial-mesenchymal transition & metastasis

As the fourth most common cancer among females worldwide, lymph node metastasis (LNM) is a key prognostic factor and a leading cause of death in patients with CC [121]. In metastasis, the ability to undergo reversible cellular and phenotypic changes is crucial for disseminating cancer cells to adapt to the changing microenvironments and stress during this pathological process. One main form of cellular plasticity is the epithelial-mesenchymal transition (EMT); in this process, epithelial cells lose tight cell-cell connections and polarity, which confers migratory and invasive properties (Figure 5); in its reverse process, mesenchymal-epithelial transition (MET) cells loose migratory freedom; they begin expressing junction complexes and adopt apicobasal polarity. This form of cellular plasticity may occur in physiological processes like human embryonic development (e.g., neural crest formation) and pathological conditions such as organ fibrosis, cancer progression, and metastasis [122, 123].

Figure 5.

Stepwise epithelial-mesenchymal transitions. 1. Tumor cells acquire the ability to dissociate themselves from the primary tumor mass through EMT; these epithelial cells lose their cell-cell junctions to become motile. 2. Now these cells can migrate and invade through the ECM. 3. Intravasation into blood vessels or lymphatic vessels occur; in this phase, tumor cells pass through the endothelial lamina and enter systemic circulation. 4. During extravasation, tumor cells extravasate through the capillary endothelium of distant organs into its parenchyma. 5. Tumor cells establish themselves and proliferate, forming micrometastases. 6. Colonization of distant organs and re-activation of epithelial properties occur at the secondary site via the MET where these cells become malignant secondary tumors; EMT facilitates cancer cells to invade, intravasate, and survive in circulation; cancer cells need to undergo MET to colonize efficiently at distant organs.

9.1 Metastasis cascade

Tumor cells that have undergone EMT exhibit stem-cell-like properties, including the ability to self-renew, tumor-initiation properties, and resistance to chemotherapy and radiotherapy, which explains why metastasis is the primary cause of death in cancer patients and why advances in tumor expression markers can help identify and prevent metastasis from happening [123, 124].

There are different markers to identify the state in which a tumor cell can be found; in the case of tumor cells in the epithelial state, researchers look out for the expression of E-cadherin, EpCAM, claudins, occludins, and cytokeratins; in the case of cells in the mesenchymal state, they can look for expression of vimentin (VIM), fibronectin, and α-SMA [125].

While EMT does not require changes in DNA sequence and can be reversible [124], the same cannot be said about other pathways; for example, due to high energy demand during EMT, several morphological and metabolic changes are made by 5’AMP-activated kinase (AMPK), which is a cellular energy homeostasis sensor that controls the balance between energy intake and demand; it modulates processes such as carbohydrate and lipid metabolism, biosynthesis, autophagy, and cell cycle.

Specifically, in the case of CC metastasis, a study by Konieczny et al. demonstrated that AMPK expression is related to malignant behavior in CC cells [126]. Another example is the case of protein tyrosine phosphatase receptor type M (PTPRM); Liu et al. demonstrated that PTPRM was upregulated in CC with LNM; as a result, it promoted tumor cell proliferation, migration, and lymphangiogenesis (a critical early metastasis event important in LNM and a prognosis factor in patients with cervical cancer), as well as EMT via the activation of Src-AKT signaling pathway and induced lymphangiogenesis in a VEGF-C-dependent manner [121]. Research on the EMT and MET has also uncovered numerous novel signaling pathways, including TGF-β, Wnt, Notch, Hedgehog, and PI3K pathways, that facilitate EMT in tumor cells [121].

Advertisement

10. Conclusion

In the previous chapter, the currently known mechanisms that promote CC oncogenesis were explained. It is essential to emphasize the evident relationship between infection by high-risk HPV serotypes and the development of CC. There are well-defined molecular mechanisms by years of research since the relationship was discovered by Zur Hausen et al. regarding CC and HPV. Although there are still other mechanisms pending elucidation, what is clear is that the more its etiopathogenesis and the complexity of its molecular mechanics are known, the more are the possibilities of developing effective treatments.

Appendices and nomenclature

ADC

adenocarcinoma

ADSC

adenosquamous carcinoma

AMPK

5’AMP-activated kinase

Ang

angiopoietins

CC

cervical cancer

CIN

cervical intraepithelial neoplasia

DDR

DNA damage response

EMC

extracellular matrix

EMT

epithelial-mesenchymal transition

EPC

endothelial progenitor cells

FAD

flavin adenine dinucleotide

GFGF

fibroblast growth factor

GPCR

receptors associated with G protein.

HIF-1

hypoxia-induced factor-1

HPV

human papillomavirus

hTERT

human telomerase reverse transcriptase

JAK/STAT

janus kinase/ signal transducer and transcription activator

LMN

lymph node metastasis

MAPK

mitogen activated protein kinase

MET

mesenchymal-epithelial transition

NAD

nicotinamide adenine dinucleotide

PDGF

platelet-derived growth factor

PIAS

activated protein inhibitors of stat.

PPC

pericyte progenitor cells

PTP

protein tyrosine phosphatase

PTPRM

protein tyrosine phosphatase receptor type M

ROS

reactive oxygen species

RTKs

receptor tyrosine kinase

SCC

squamous cell carcinoma

SOCS

suppressor signaling cytokines.

TAM

tumor-associated macrophages

TILs

tumor-infiltrating lymphocytes

TME

tumor microenvironment

Tregs

regulatory T cells

TSG

tumor suppressor genes

TSP-1

Thrombospondin-1

VEGF

vascular endothelial growth factor

VIM

vimentin

WHO

World Health Organization

References

  1. 1. Sanchez Gaitan E. Tamizaje y tratamiento de las lesiones precancerosas para la prevención del cáncer cervicouterino. Revista Medica Sinergia. 2019;4:e300. DOI: 10.31434/rms.v4i11.300
  2. 2. Prendiville W, Sankaranarayanan R. Colposcopy and Treatment of Cervical Precancer. Vol. 45. Lyon, France: World Health Organization; 2017. p. 178
  3. 3. Eun T, Perkins J. Screening for cervical cancer. Medical Clinics of North America. 2020;104:15. DOI: 10.1016/j.mcna.2020.08.006
  4. 4. Buskwofie A, David-West G, Clare CA. A review of cervical cancer: Incidence and disparities. Journal of the National Medical Association. 2020;112:229-232. DOI: 10.1016/j.jnma.2020.03.002
  5. 5. Arbyn M, Weiderpass E, Bruni L, de Sanjosé S, Saraiya M, Ferlay J, et al. Estimates of incidence and mortality of cervical cancer in 2018: A worldwide analysis. The Lancet Global Health. 2020;8:e191-e203. DOI: 10.1016/s2214-109x(19)30482-6
  6. 6. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer Journal for Clinicians. 2021;71:209-249. DOI: 10.3322/caac.21660
  7. 7. Zhang S, Huifang X, Zhang L, Qiao Y. Cervical cancer: Epidemiology, risk factors and screening. Chinese Journal of Cancer Research. 2020;32:8
  8. 8. Organization, W.H. Major Milestone Reached as 100 Countries have Introduced HPV Vaccine into National Schedule. Available from: https://www.who.int/news/item/31-10-2019-major-milestone-reached-as-100-countries-have-introduced-hpv-vaccine-into-national-schedule
  9. 9. Organization PAH. Pan American Health Organization. Plan of Action for Cervical Cancer Prevention and Control 2018-2030. Washington, D.C.: PAHO; 2018. pp. 1-44
  10. 10. Kuhn L, Denny L, Pollack A, Lorincz A, Richart R, Wright T. Human papillomavirus DNA testing for cervical cancer screening in low-resource settings. Journal of the National Center Institute. 2000;92:7. DOI: 10.1093/jnci/92.10.818
  11. 11. Hellberg D. Sex steroids and cervical cancer. Anticancer Research. 2012;32:9
  12. 12. Fonseca-Moutinho J. Smoking and cervical cancer. ISRN Obstetrics and Gynecology. 2011;2011:1-6
  13. 13. Jensen KE, Schmiedel S, Norrild B, Frederiksen K, Iftner T, Kjaer SK. Parity as a cofactor for high-grade cervical disease among women with persistent human papillomavirus infection: A 13-year follow-up. British Journal of Cancer. 2013;108:234-239. DOI: 10.1038/bjc.2012.513
  14. 14. Kashyap N, Krishnan N, Kaur S, Ghai S. Risk factors of cervical cancer: A case-control study. Asia-Pacific Journal of Oncology Nursing. 2019;6:308-314. DOI: 10.4103/apjon.apjon_73_18
  15. 15. Ngoma M, Autier P. Cancer prevention: Cervical cancer. Ecancermedicalscience. 2019;13:952. DOI: 10.3332/ecancer.2019.952
  16. 16. Harden ME, Munger K. Human papillomavirus molecular biology. Mutation Research, Reviews in Mutation Research. 2017;772:3-12. DOI: 10.1016/j.mrrev.2016.07.002
  17. 17. Leite KRM, Pimenta R, Canavez J, Canavez F, de Souza FR, Vara L, et al. HPV genotype prevalence and success of vaccination to prevent cervical cancer. Acta Cytologica. 2020;64:420-424. DOI: 10.1159/000506725
  18. 18. Venetianer R, Clarke MA, van der Marel J, Tota J, Schiffman M, Dunn ST, et al. Identification of HPV genotypes causing cervical precancer using tissue-based genotyping. International Journal of Cancer. 2020;146:2836-2844. DOI: 10.1002/ijc.32919
  19. 19. Manini I, Montomoli E. Epidemiology and prevention of human papillomavirus. Annali di igiene: Medicina preventiva e di comunita. 2018;30:28-32. DOI: 10.7416/ai.2018.2231
  20. 20. Chan CK, Aimagambetova G, Ukybassova T, Kongrtay K, Azizan A. Human papillomavirus infection and cervical cancer: Epidemiology, screening, and vaccination-review of current perspectives. Journal of Oncology. 2019;2019:3257939. DOI: 10.1155/2019/3257939
  21. 21. Hoppe-Seyler K, Bossler F, Braun JA, Herrmann AL, Hoppe-Seyler F. The HPV E6/E7 oncogenes: Key factors for viral carcinogenesis and therapeutic targets. Trends in Microbiology. 2018;26:158-168. DOI: 10.1016/j.tim.2017.07.007
  22. 22. Estevao D, Costa NR, Gil da Costa RM, Medeiros R. Hallmarks of HPV carcinogenesis: The role of E6, E7 and E5 oncoproteins in cellular malignancy. Biochim Biophys Acta Gene Regul Mech. 2019;1862:153-162. DOI: 10.1016/j.bbagrm.2019.01.001
  23. 23. Gutiérrez-Hoya A, Soto-Cruz I. Role of the JAK/STAT pathway in cervical cancer: Its relationship with HPV E6/E7 oncoproteins. Cell. 2020;9:2297
  24. 24. Kontomanolis EN, Koutras A, Syllaios A, Schizas D, Mastoraki A, Garmpis N, et al. Role of oncogenes and tumor-suppressor genes in carcinogenesis: A review. Anticancer Research. 2020;40:6009-6015. DOI: 10.21873/anticanres.14622
  25. 25. Kontomanolis EN, Koutras A, Fasoulakis Z, Syllaios A, Diakosavvas M, Angelou K, et al. A brief overview of oncogenes and signal transduction pathways in gynecological cancer. Cancer Diagnosis & Prognosis. 2022;2:134-143. DOI: 10.21873/cdp.10087
  26. 26. Oxford Textbook of Cancer Biology. Oxford, United Kingdom: Oxford University Press; 2019
  27. 27. Chernoff J. The two-hit theory hits 50. Molecular Biology of the Cell. 2021;32:rt1. DOI: 10.1091/mbc.E21-08-0407
  28. 28. Moody CA, Laimins LA. Human papillomavirus oncoproteins: Pathways to transformation. Nature Reviews. Cancer. 2010;10:550-560. DOI: 10.1038/nrc2886
  29. 29. Egawa N, Egawa K, Griffin H, Doorbar J. Human papillomaviruses; epithelial tropisms, and the development of neoplasia. Viruses. 2015;7:3863-3890. DOI: 10.3390/v7072802
  30. 30. McBride AA, Warburton A. The role of integration in oncogenic progression of HPV-associated cancers. PLoS Pathogens. 2017;13:e1006211. DOI: 10.1371/journal.ppat.1006211
  31. 31. Shen-Gunther J, Cai H, Wang Y. HPV integration site mapping: A rapid method of viral integration site (VIS) analysis and visualization using automated workflows in CLC microbial genomics. International Journal of Molecular Sciences. 2022;23:8132
  32. 32. Dong G, Broker TR, Chow LT. Human papillomavirus type 11 E2 proteins repress the homologous E6 promoter by interfering with the binding of host transcription factors to adjacent elements. Journal of Virology. 1994;68:1115-1127. DOI: 10.1128/jvi.68.2.1115-1127.1994
  33. 33. Tan SH, Leong LE, Walker PA, Bernard HU. The human papillomavirus type 16 E2 transcription factor binds with low cooperativity to two flanking sites and represses the E6 promoter through displacement of Sp1 and TFIID. Journal of Virology. 1994;68:6411-6420. DOI: 10.1128/jvi.68.10.6411-6420.1994
  34. 34. Warburton A, Della Fera AN, McBride AA. Dangerous liaisons: Long-term replication with an extrachromosomal HPV genome. Viruses. 2021;1846:13
  35. 35. Yang YK, Xi WY, Xi RX, Li JY, Li Q , Gao YE. MicroRNA-494 promotes cervical cancer proliferation through the regulation of PTEN. Oncology Reports. 2015;33:2393-2401. DOI: 10.3892/or.2015.3821
  36. 36. Hanahan D. Hallmarks of cancer: New dimensions. Cancer Discovery. 2022;12:31-46. DOI: 10.1158/2159-8290.Cd-21-1059
  37. 37. Morana O, Wood W, Gregory CD. The apoptosis paradox in cancer. International Journal of Molecular Sciences. 2022;23:1-19. DOI: 10.3390/ijms23031328
  38. 38. Pal A, Kundu R. Human papillomavirus E6 and E7: The cervical cancer hallmarks and targets for therapy. Frontiers in Microbiology. 2019;10:3116. DOI: 10.3389/fmicb.2019.03116
  39. 39. Bernard X, Robinson P, Nominé Y, Masson M, Charbonnier S, Ramirez-Ramos JR, et al. Proteasomal degradation of p53 by human papillomavirus E6 oncoprotein relies on the structural integrity of p53 core domain. PLoS One. 2011;6:e25981. DOI: 10.1371/journal.pone.0025981
  40. 40. Thomas M, Banks L. Human papillomavirus (HPV) E6 interactions with Bak are conserved amongst E6 proteins from high and low risk HPV types. The Journal of General Virology. 1999;80(Pt 6):1513-1517. DOI: 10.1099/0022-1317-80-6-1513
  41. 41. Filippova M, Song H, Connolly JL, Dermody TS, Duerksen-Hughes PJ. The human papillomavirus 16 E6 protein binds to tumor necrosis factor (TNF) R1 and protects cells from TNF-induced apoptosis. The Journal of Biological Chemistry. 2002;277:21730-21739. DOI: 10.1074/jbc.M200113200
  42. 42. Garnett TO, Filippova M, Duerksen-Hughes PJ. Accelerated degradation of FADD and procaspase 8 in cells expressing human papilloma virus 16 E6 impairs TRAIL-mediated apoptosis. Cell Death and Differentiation. 2006;13:1915-1926. DOI: 10.1038/sj.cdd.4401886
  43. 43. Aguilar-Lemarroy A, Gariglio P, Whitaker NJ, Eichhorst ST, zur Hausen H, Krammer PH, et al. Restoration of p53 expression sensitizes human papillomavirus type 16 immortalized human keratinocytes to CD95-mediated apoptosis. Oncogene. 2002;21:165-175. DOI: 10.1038/sj.onc.1204979
  44. 44. Stöppler MC, Straight SW, Tsao G, Schlegel R, McCance DJ. The E5 gene of HPV-16 enhances keratinocyte immortalization by full-length DNA. Virology. 1996;223:251-254. DOI: 10.1006/viro.1996.0475
  45. 45. Oh JM, Kim SH, Cho EA, Song YS, Kim WH, Juhnn YS. Human papillomavirus type 16 E5 protein inhibits hydrogen-peroxide-induced apoptosis by stimulating ubiquitin-proteasome-mediated degradation of Bax in human cervical cancer cells. Carcinogenesis. 2010;31:402-410. DOI: 10.1093/carcin/bgp318
  46. 46. Kabsch K, Mossadegh N, Kohl A, Komposch G, Schenkel J, Alonso A, et al. The HPV-16 E5 protein inhibits TRAIL- and FasL-mediated apoptosis in human keratinocyte raft cultures. Intervirology. 2004;47:48-56. DOI: 10.1159/000076642
  47. 47. Bailey SM. Editorial: Hallmark of cancer: Replicative immortality. Frontiers in Oncology. 2023;13:1204094. DOI: 10.3389/fonc.2023.1204094
  48. 48. Souiden Y, Bouraoui A, Chaieb K, Mahdouani K. Telomeres and telomerase as targeted therapies in cancer treatment. Bulletin du Cancer. 2010;97:1087-1104. DOI: 10.1684/bdc.2010.1155
  49. 49. Belgiovine C, Chiodi I, Mondello C. Telomerase: Cellular immortalization and neoplastic transformation. Multiple functions of a multifaceted complex. Cytogenetic and Genome Research. 2008;122:255-262. DOI: 10.1159/000167811
  50. 50. Liu X, Dakic A, Zhang Y, Dai Y, Chen R, Schlegel R. HPV E6 protein interacts physically and functionally with the cellular telomerase complex. Proceedings of the National Academy of Sciences. 2009;106:18780-18785. DOI: 10.1073/pnas.0906357106
  51. 51. Hu X, Li J, Fu M, Zhao X, Wang W. The JAK/STAT signaling pathway: From bench to clinic. Signal transduction and targeted. Therapy. 2021;6:402. DOI: 10.1038/s41392-021-00791-1
  52. 52. O'Shea JJ, Schwartz DM, Villarino AV, Gadina M, McInnes IB, Laurence A. The JAK-STAT pathway: Impact on human disease and therapeutic intervention. Annual Review of Medicine. 2015;66:311-328. DOI: 10.1146/annurev-med-051113-024537
  53. 53. Murray PJ. The JAK-STAT signaling pathway: Input and output integration. Journal of Immunology. 2007;178:2623-2629. DOI: 10.4049/jimmunol.178.5.2623
  54. 54. Xue C, Yao Q , Gu X, Shi Q , Yuan X, Chu Q , et al. Evolving cognition of the JAK-STAT signaling pathway: Autoimmune disorders and cancer. Signal Transduction and Targeted Therapy. 2023;8:204. DOI: 10.1038/s41392-023-01468-7
  55. 55. Li Q , Li Z, Luo T, Shi H. Targeting the PI3K/AKT/mTOR and RAF/MEK/ERK pathways for cancer therapy. Molecular Biomedicine. 2022;3:47. DOI: 10.1186/s43556-022-00110-2
  56. 56. Steelman LS, Chappell WH, Abrams SL, Kempf RC, Long J, Laidler P, et al. Roles of the Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR pathways in controlling growth and sensitivity to therapy-implications for cancer and aging. Aging (Albany NY). 2011;3:192-222. DOI: 10.18632/aging.100296
  57. 57. Bossler F, Hoppe-Seyler K, Hoppe-Seyler F. PI3K/AKT/mTOR signaling regulates the virus/host cell crosstalk in HPV-positive cervical cancer cells. International Journal of Molecular Sciences. 2019;20:2188
  58. 58. MacDonald BT, Tamai K, He X. Wnt/beta-catenin signaling: Components, mechanisms, and diseases. Developmental Cell. 2009;17:9-26. DOI: 10.1016/j.devcel.2009.06.016
  59. 59. Stamos JL, Weis WI. The β-catenin destruction complex. Cold Spring Harbor Perspectives in Biology. 2013;5:a007898. DOI: 10.1101/cshperspect.a007898
  60. 60. Komiya Y, Habas R. Wnt signal transduction pathways. Organogenesis. 2008;4:68-75. DOI: 10.4161/org.4.2.5851
  61. 61. Zhan T, Rindtorff N, Boutros M. Wnt signaling in cancer. Oncogene. 2017;36:1461-1473. DOI: 10.1038/onc.2016.304
  62. 62. Liu J, Xiao Q , Xiao J, Niu C, Li Y, Zhang X, et al. Wnt/β-catenin signalling: Function, biological mechanisms, and therapeutic opportunities. Signal Transduction and Targeted Therapy. 2022;7:3. DOI: 10.1038/s41392-021-00762-6
  63. 63. Zapatka M, Borozan I, Brewer DS, Iskar M, Grundhoff A, Alawi M, et al. The landscape of viral associations in human cancers. Nature Genetics. 2020;52:320-330. DOI: 10.1038/s41588-019-0558-9
  64. 64. Porter VL, Marra MA. The drivers, mechanisms, and consequences of genome instability in HPV-driven cancers. Cancers. 2022;14:4623
  65. 65. Parish JL, Bean AM, Park RB, Androphy EJ. ChlR1 is required for loading papillomavirus E2 onto mitotic chromosomes and viral genome maintenance. Molecular Cell. 2006;24:867-876. DOI: 10.1016/j.molcel.2006.11.005
  66. 66. Sakakibara N, Mitra R, McBride AA. The papillomavirus E1 helicase activates a cellular DNA damage response in viral replication foci. Journal of Virology. 2011;85:8981-8995. DOI: 10.1128/jvi.00541-11
  67. 67. Cruz-Gregorio A, Manzo-Merino J, Gonzaléz-García MC, Pedraza-Chaverri J, Medina-Campos ON, Valverde M, et al. Human papillomavirus types 16 and 18 early-expressed proteins differentially modulate the cellular redox state and DNA damage. International Journal of Biological Sciences. 2018;14:21-35. DOI: 10.7150/ijbs.21547
  68. 68. Peter M, Stransky N, Couturier J, Hupé P, Barillot E, de Cremoux P, et al. Frequent genomic structural alterations at HPV insertion sites in cervical carcinoma. The Journal of Pathology. 2010;221:320-330. DOI: 10.1002/path.2713
  69. 69. Halbert CL, Demers GW, Galloway DA. The E7 gene of human papillomavirus type 16 is sufficient for immortalization of human epithelial cells. Journal of Virology. 1991;65:473-478. DOI: 10.1128/jvi.65.1.473-478.1991
  70. 70. Sitz J, Blanchet SA, Gameiro SF, Biquand E, Morgan TM, Galloy M, et al. Human papillomavirus E7 oncoprotein targets RNF168 to hijack the host DNA damage response. Proceedings of the National Academy of Sciences. 2019;116:19552-19562. DOI: 10.1073/pnas.1906102116
  71. 71. Wallace NA, Khanal S, Robinson KL, Wendel SO, Messer JJ, Galloway DA. High-risk Alphapapillomavirus oncogenes impair the homologous recombination pathway. Journal of Virology. 2017;91:1-22. DOI: 10.1128/jvi.01084-01017
  72. 72. Duensing S, Duensing A, Flores ER, Do A, Lambert PF, Münger K. Centrosome abnormalities and genomic instability by episomal expression of human papillomavirus type 16 in raft cultures of human keratinocytes. Journal of Virology. 2001;75:7712-7716. DOI: 10.1128/jvi.75.16.7712-7716.2001
  73. 73. Klingelhutz AJ, Roman A. Cellular transformation by human papillomaviruses: Lessons learned by comparing high- and low-risk viruses. Virology. 2012;424:77-98. DOI: 10.1016/j.virol.2011.12.018
  74. 74. Cheung AL, Deng W. Telomere dysfunction, genome instability and cancer. Frontiers in Bioscience. 2008;13:2075-2090. DOI: 10.2741/2825
  75. 75. Widschwendter A, Müller HM, Hubalek MM, Wiedemair A, Fiegl H, Goebel G, et al. Methylation status and expression of human telomerase reverse transcriptase in ovarian and cervical cancer. Gynecologic Oncology. 2004;93:407-416. DOI: 10.1016/j.ygyno.2004.01.036
  76. 76. Warburg O. On the origin of cancer cells. Science. 1956;123:309-314. DOI: 10.1126/science.123.3191.309
  77. 77. Vinay Kumar AKA, Aster JC. Robbins Basic Pathology. 10th ed. Philadelphia, USA: Elsevier—Health Sciences Division; 2023
  78. 78. Bast RC, Kufe DW, Pollock RE, Weichselbaum RR, Holland JF, Frei E. Holland-Frei Cancer Medicine. 10th ed. Hoboken, New Jersey: John Wiley and Sons; 2023
  79. 79. Kennelly PJ, Botham KM, McGuinness OP, Rodwell VW, Weil P. Harper's Illustrated Biochemistry. 32nd ed. New York: McGraw Hill; 2023
  80. 80. Yuan Y, Cai X, Shen F, Ma F. HPV post-infection microenvironment and cervical cancer. Cancer Letters. 2021;497:243-254. DOI: 10.1016/j.canlet.2020.10.034
  81. 81. Brand KA, Hermfisse U. Aerobic glycolysis by proliferating cells: A protective strategy against reactive oxygen species1. The FASEB Journal. 1997;11:388-395. DOI: 10.1096/fasebj.11.5.9141507
  82. 82. Torresano L, Nuevo-Tapioles C, Santacatterina F, Cuezva JM. Metabolic reprogramming and disease progression in cancer patients. Biochimica et Biophysica Acta (BBA)—Molecular Basis of Disease. 1866;2020:165721. DOI: 10.1016/j.bbadis.2020.165721
  83. 83. Ilhan ZE, Łaniewski P, Thomas N, Roe DJ, Chase DM, Herbst-Kralovetz MM. Deciphering the complex interplay between microbiota, HPV, inflammation and cancer through cervicovaginal metabolic profiling. eBioMedicine. 2019;44:675-690. DOI: 10.1016/j.ebiom.2019.04.028
  84. 84. Zhang R, Su J, Xue SL, Yang H, Ju LL, Ji Y, et al. HPV E6/p53 mediated down-regulation of miR-34a inhibits Warburg effect through targeting LDHA in cervical cancer. American Journal of Cancer Research. 2016;6:312-320
  85. 85. Henke E, Nandigama R, Ergün S. Extracellular matrix in the tumor microenvironment and its impact on cancer therapy. Frontiers in Molecular Biosciences. 2019;6:160. DOI: 10.3389/fmolb.2019.00160
  86. 86. Anderson NM, Simon MC. The tumor microenvironment. Current Biology. 2020;30:R921-R925. DOI: 10.1016/j.cub.2020.06.081
  87. 87. Hanahan D, Coussens LM. Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012;21:309-322. DOI: 10.1016/j.ccr.2012.02.022
  88. 88. Balkwill FR, Capasso M, Hagemann T. The tumor microenvironment at a glance. Journal of Cell Science. 2012;125:5591-5596. DOI: 10.1242/jcs.116392
  89. 89. Cisneros-Villanueva M, Hidalgo-Pérez L, Rios-Romero M, Cedro-Tanda A, Ruiz-Villavicencio CA, Page K, et al. Cell-free DNA analysis in current cancer clinical trials: A review. British Journal of Cancer. 2022;126:391-400. DOI: 10.1038/s41416-021-01696-0
  90. 90. Lin D, Shen L, Luo M, Zhang K, Li J, Yang Q , et al. Circulating tumor cells: Biology and clinical significance. Signal Transduction and Targeted Therapy. 2021;6:404. DOI: 10.1038/s41392-021-00817-8
  91. 91. Li I, Nabet BY. Exosomes in the tumor microenvironment as mediators of cancer therapy resistance. Molecular Cancer. 2019;18:32. DOI: 10.1186/s12943-019-0975-5
  92. 92. Monteran L, Erez N. The dark side of fibroblasts: Cancer-associated fibroblasts as mediators of immunosuppression in the tumor microenvironment. Frontiers in Immunology. 1835;2019:10. DOI: 10.3389/fimmu.2019.01835
  93. 93. Lisanti MP, Martinez-Outschoorn UE, Chiavarina B, Pavlides S, Whitaker-Menezes D, Tsirigos A, et al. Understanding the "lethal" drivers of tumor-stroma co-evolution. Cancer Biology & Therapy. 2010;10:537-542. DOI: 10.4161/cbt.10.6.13370
  94. 94. Tang Y, Zhang AXJ, Chen G, Wu Y, Gu W. Prognostic and therapeutic TILs of cervical cancer—Current advances and future perspectives. Molecular Therapy - Oncolytics. 2021;22:410-430. DOI: 10.1016/j.omto.2021.07.006
  95. 95. Ma W, Wang K, Du J, Luan J, Lou G. Multi-dose parecoxib provides an immunoprotective effect by balancing T helper 1 (Th1), Th2, Th17 and regulatory T cytokines following laparoscopy in patients with cervical cancer. Molecular Medicine Reports. 2015;11:2999-3008. DOI: 10.3892/mmr.2014.3003
  96. 96. Lin W, Niu Z, Zhang H, Kong Y, Wang Z, Yang X, et al. Imbalance of Th1/Th2 and Th17/Treg during the development of uterine cervical cancer. International Journal of Clinical and Experimental Pathology. 2019;12:3604-3612
  97. 97. Balkwill F. Cancer and the chemokine network. Nature Reviews. Cancer. 2004;4:540-550. DOI: 10.1038/nrc1388
  98. 98. Wang S, Liu R, Yu Q , Dong L, Bi Y, Liu G. Metabolic reprogramming of macrophages during infections and cancer. Cancer Letters. 2019;452:14-22. DOI: 10.1016/j.canlet.2019.03.015
  99. 99. Petrillo M, Zannoni GF, Martinelli E, Pedone Anchora L, Ferrandina G, Tropeano G, et al. Polarisation of tumor-associated macrophages toward M2 phenotype correlates with poor response to chemoradiation and reduced survival in patients with locally advanced cervical Cancer. PLoS One. 2015;10:e0136654. DOI: 10.1371/journal.pone.0136654
  100. 100. Hillen F, Griffioen AW. Tumour vascularization: Sprouting angiogenesis and beyond. Cancer Metastasis Reviews. 2007;26:489-502. DOI: 10.1007/s10555-007-9094-7
  101. 101. Folkman J, D'Amore PA. Blood vessel formation: What is its molecular basis? Cell. 1996;87:1153-1155. DOI: 10.1016/S0092-8674(00)81810-3
  102. 102. Albrecht ED, Babischkin JS, Lidor Y, Anderson LD, Udoff LC, Pepe GJ. Effect of estrogen on angiogenesis in co-cultures of human endometrial cells and microvascular endothelial cells. Human Reproduction. 2003;18:2039-2047. DOI: 10.1093/humrep/deg415
  103. 103. Appelmann I, Liersch R, Kessler T, Mesters RM, Berdel WE. Angiogenesis inhibition in cancer therapy: Platelet-derived growth factor (PDGF) and vascular endothelial growth factor (VEGF) and their receptors: Biological functions and role in malignancy. Recent Results in Cancer Research. 2010;180:51-81. DOI: 10.1007/978-3-540-78281-0_5
  104. 104. Martínez CE, Smith PC, Palma Alvarado VA. The influence of platelet-derived products on angiogenesis and tissue repair: A concise update. Frontiers in Physiology. 2015;6:1-7. DOI: 10.3389/fphys.2015.00290
  105. 105. Bergers G, Benjamin LE. Tumorigenesis and the angiogenic switch. Nature Reviews Cancer. 2003;3:401-410. DOI: 10.1038/nrc1093
  106. 106. Wu MP, Tzeng CC, Wu LW, Huang KF, Chou CY. Thrombospondin-1 acts as a fence to inhibit angiogenesis that occurs during cervical carcinogenesis. Cancer Journal. 2004;10:27-32. DOI: 10.1097/00130404-200401000-00007
  107. 107. Baeriswyl V, Christofori G. The angiogenic switch in carcinogenesis. Seminars in Cancer Biology. 2009;19:329-337. DOI: 10.1016/j.semcancer.2009.05.003
  108. 108. Rofstad EK, Sundfør K, Lyng H, Tropé CG. Hypoxia-induced treatment failure in advanced squamous cell carcinoma of the uterine cervix is primarily due to hypoxia-induced radiation resistance rather than hypoxia-induced metastasis. British Journal of Cancer. 2000;83:354-359. DOI: 10.1054/bjoc.2000.1266
  109. 109. Brown LM, Cowen RL, Debray C, Eustace A, Erler JT, Sheppard FC, et al. Reversing hypoxic cell chemoresistance in vitro using genetic and small molecule approaches targeting hypoxia inducible factor-1. Molecular Pharmacology. 2006;69:411-418. DOI: 10.1124/mol.105.015743
  110. 110. Du R, Lu KV, Petritsch C, Liu P, Ganss R, Passegué E, et al. HIF1alpha induces the recruitment of bone marrow-derived vascular modulatory cells to regulate tumor angiogenesis and invasion. Cancer Cell. 2008;13:206-220. DOI: 10.1016/j.ccr.2008.01.034
  111. 111. Ferrara N. VEGF and the quest for tumour angiogenesis factors. Nature Reviews. Cancer. 2002;2:795-803. DOI: 10.1038/nrc909
  112. 112. Goodsell DS. The molecular perspective: VEGF and angiogenesis. Stem Cells. 2003;21:118-119. DOI: 10.1634/stemcells.21-1-118
  113. 113. Pepper MS, Ferrara N, Orci L, Montesano R. Potent synergism between vascular endothelial growth factor and basic fibroblast growth factor in the induction of angiogenesis in vitro. Biochemical and Biophysical Research Communications. 1992;189:824-831. DOI: 10.1016/0006-291x(92)92277-5
  114. 114. Giavazzi R, Sennino B, Coltrini D, Garofalo A, Dossi R, Ronca R, et al. Distinct role of fibroblast growth factor-2 and vascular endothelial growth factor on tumor growth and angiogenesis. The American Journal of Pathology. 2003;162:1913-1926. DOI: 10.1016/s0002-9440(10)64325-8
  115. 115. Fujii T, Kuwano H. Regulation of the expression balance of angiopoietin-1 and angiopoietin-2 by Shh and FGF-2. In Vitro Cellular & Developmental Biology. Animal. 2010;46:487-491. DOI: 10.1007/s11626-009-9270-x
  116. 116. Nasarre P, Thomas M, Kruse K, Helfrich I, Wolter V, Deppermann C, et al. Host-derived angiopoietin-2 affects early stages of tumor development and vessel maturation but is dispensable for later stages of tumor growth. Cancer Research. 2009;69:1324-1333. DOI: 10.1158/0008-5472.Can-08-3030
  117. 117. Monk BJ, Sill MW, Burger RA, Gray HJ, Buekers TE, Roman LD. Phase II trial of bevacizumab in the treatment of persistent or recurrent squamous cell carcinoma of the cervix: A gynecologic oncology group study. Journal of Clinical Oncology. 2009;27:1069-1074. DOI: 10.1200/jco.2008.18.9043
  118. 118. Ikeda Y, Takano M, Oda K, Kouta H, Goto T, Kudoh K, et al. Weekly administration of bevacizumab, gemcitabine, and oxaliplatin in patients with recurrent and refractory ovarian cancer: A preliminary result of 19 cases. International Journal of Gynecological Cancer. 2013;23:355-360. DOI: 10.1097/IGC.0b013e31827de69e
  119. 119. Schefter TE, Winter K, Kwon JS, Stuhr K, Balaraj K, Yaremko BP, et al. A phase II study of bevacizumab in combination with definitive radiotherapy and cisplatin chemotherapy in untreated patients with locally advanced cervical carcinoma: Preliminary results of RTOG 0417. International Journal of Radiation Oncology, Biology, Physics. 2012;83:1179-1184. DOI: 10.1016/j.ijrobp.2011.10.060
  120. 120. Schefter T, Winter K, Kwon JS, Stuhr K, Balaraj K, Yaremko BP, et al. RTOG 0417: Efficacy of bevacizumab in combination with definitive radiation therapy and cisplatin chemotherapy in untreated patients with locally advanced cervical carcinoma. International Journal of Radiation Oncology, Biology, Physics. 2014;88:101-105. DOI: 10.1016/j.ijrobp.2013.10.022
  121. 121. Liu P, Zhang C, Liao Y, Liu J, Huang J, Xia M, et al. High expression of PTPRM predicts poor prognosis and promotes tumor growth and lymph node metastasis in cervical cancer. Cell Death & Disease. 2020;11:687. DOI: 10.1038/s41419-020-02826-x
  122. 122. Cui J, Zhang C, Lee J-E, Bartholdy BA, Yang D, Liu Y, et al. MLL3 loss drives metastasis by promoting a hybrid epithelial–mesenchymal transition state. Nature Cell Biology. 2023;25:145-158. DOI: 10.1038/s41556-022-01045-0
  123. 123. Vasaikar SV, Deshmukh AP, den Hollander P, Addanki S, Kuburich NA, Kudaravalli S, et al. EMTome: A resource for pan-cancer analysis of epithelial-mesenchymal transition genes and signatures. British Journal of Cancer. 2021;124:259-269. DOI: 10.1038/s41416-020-01178-9
  124. 124. Yang J, Antin P, Berx G, Blanpain C, Brabletz T, Bronner M, et al. Guidelines and definitions for research on epithelial–mesenchymal transition. Nature Reviews Molecular Cell Biology. 2020;21:341-352. DOI: 10.1038/s41580-020-0237-9
  125. 125. Vergara D, Simeone P, Franck J, Trerotola M, Giudetti A, Capobianco L, et al. Translating epithelial mesenchymal transition markers into the clinic: Novel insights from proteomics. EuPA Open Proteomics. 2016;10:31-41. DOI: 10.1016/j.euprot.2016.01.003
  126. 126. Konieczny P, Adamus T, Sułkowski M, Skrzypek K, Majka M. Impact of AMPK on cervical carcinoma progression and metastasis. Cell Death & Disease. 2023;14:43. DOI: 10.1038/s41419-023-05583-9

Written By

Natalia Garcia-Becerra, Carlos A. Garcia-Becerra, Leonardo Fernandez-Avila, Jose Roberto Cruz-Lozano, Veronica Soltero-Molinar, Isabel Arias-Gallardo, Sofia Briseida Leyva-Delgado, Angel E. Chávez-Torres, Dalia I. Murillo-Geraldo and Jesús E. Juarez-Garcia

Submitted: 29 June 2023 Reviewed: 16 July 2023 Published: 21 December 2023