Open Access

Role of the tumor microenvironment in the lymphatic metastasis of cervical cancer (Review)

  • Authors:
    • Lufang Wang
    • Shuyan Yi
    • Yun Teng
    • Wenhan Li
    • Jing Cai
  • View Affiliations

  • Published online on: September 1, 2023     https://doi.org/10.3892/etm.2023.12185
  • Article Number: 486
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Lymphatic metastasis is the primary type of cervical cancer metastasis and is associated with an extremely poor prognosis in patients. The tumor microenvironment primarily includes cancer‑associated fibroblasts, tumor‑associated macrophages, myeloid‑derived suppressor cells, immune and inflammatory cells, and blood and lymphatic vascular networks, which can promote the establishment of lymphatic metastatic sites within immunosuppressive microenvironments or promote lymphatic metastasis by stimulating lymphangiogenesis and epithelial‑mesenchymal transformation. As the most important feature of the tumor microenvironment, hypoxia plays an essential role in lymph node metastasis. In this review, the known mechanisms of hypoxia, and the involvement of stromal components and immune inflammatory cells in the tumor microenvironment of lymphatic metastasis of cervical cancer are discussed. Additionally, a summary of the clinical trials targeting the tumor microenvironment for the treatment of cervical cancer is provided, emphasizing the potential and challenges of immunotherapy.

1. Introduction

Cervical cancer (CC), as one of the most common causes of female mortality, poses a serious threat to women's lives and health. Globally, in 2020, there were an estimated 604,127 CC cases and 341,831 related deaths, with a corresponding age-standardized incidence of 13.3 cases per 100,000 women-years and a mortality rate of 7.2 deaths per 100,000 women-years (1). Lymph node metastasis (LNM) is the most common type of CC metastasis and is closely related to prognosis. The more extensive the LNM is, the worse the prognosis of patients. Studies have confirmed that the overall 5-year survival rates of CC patients with 0, 1-2, 3-9 and 10 or more metastatic lymph nodes are 90, 69, 57 and 35%, respectively (2). According to the 2009 FIGO staging principle, LNM does not affect the International Federation of Obstetrics and Gynecology (FIGO) CC staging. However, the FIGO staging system released in 2018 clearly states that once CC patients are diagnosed with LNM, they can be directly diagnosed with stage IIIC or above CC, which fully demonstrates the important role of LNM in the progression of CC (3). Unfortunately, little is known regarding the LNM mechanism in CC, which remains one of the biggest challenges in treating CC (4).

The tumor microenvironment (TME) is primarily composed of fibroblasts, endothelial cells, different subsets of infiltrating immune cells (IICs), bone marrow-derived progenitor cells, platelets, and inflammatory cytokines (5). Previous studies have confirmed that tumor cells or host-derived cells (immune cells and fibroblasts, amongst others) in the TME can release various lymphatic angiogenic factors, such as vascular endothelial growth factor (VEGF)-A, C and D, lymphatic vascular factor angiogenin-2, and hepatocyte growth factor, which can stimulate angiogenesis and lymphangiogenesis (6). Tumor cells, tumor stromal cells, and infiltrating white blood cells release chemokines to recruit different immune cell types into the TME. Chemokines can be grouped into four main classes, depending on the location of the first two cysteine (C) residues of their primary protein structure, namely, the C, CC, CXC and CX3C chemokines. All chemokines signal by binding to cognate heterotrimeric G protein-coupled receptors (GPCRs) of the rhodopsin-like family found on migratory cells (7). According to the special needs of migration in each environment, chemokines can act as tumor angiogenesis media, directly interact with chemokine receptors on endothelial cells, and induce tumors to promote the release of growth factors. These growth factors can promote tumor growth in a paracrine signaling manner, thus improving migration, proliferation, and endothelial cell survival. In addition, chemokines can also cooperate with other angiogenesis promoters. For example, VEGF-, CXCL8- and CXCL12-induced upregulation of VEGF expression produces a positive feedback effect, and VEGF further stimulates the production of angiogenic chemokines (8). In addition, lymphatic endothelial cells (LECs) in tumor-draining lymph nodes have been proven to proliferate, leading to the expansion of the lymphatic sinus (9). Hypoxia can stimulate the formation of lymphatic vessels (lymphangiogenesis) and blood (angiogenesis), such that cancer cells can escape from the unfavorable tumor microenvironment and spread to an environment conducive to its survival, ultimately leading to metastatic diseases and mortality (10). Currently, research has confirmed that hypoxia promotes lymphatic metastasis primarily through HIF-1α promoting VEGF-A/-C/-D, TGF-β transcriptional activation of lymphatic vessel generation mediated by signal cascades such as Prox-1. Multiple factors, such as ET-1, C/EBP- δ, EGR-1, AP-1, MIF and NF-κB, can also promote lymphatic metastasis by promoting the proliferation and migration of LECs (11).

In the present review, the known mechanisms of hypoxia, and the involvement of stromal components and immune inflammatory cells in the tumor microenvironment in lymphatic metastasis of CC is discussed, and a summary of the clinical trials for strategies targeting the tumor microenvironment for the treatment of CC is provided.

2. Mechanism of lymphatic metastasis in cancer

Tumor cell entry into the lymphatic vasculature is the first step of metastasis. The lymphatic system primarily regulates fluid homeostasis and the immune response. Lymphatic metastasis plays an active role in the spread and metastasis of primary tumors (12). Similar to angiogenesis, lymphangiogenesis is a multi-step process. On the one hand, activated LECs proliferate and migrate under specific stimuli to form new blood vessels; on the other hand, cancer cells invading the afferent lymphatic vessels spread to the tumor-draining lymph nodes, which are an important hub for the stagnation and growth of metastatic cells, immune regulation, and secondary dissemination to distant sites (13). The process of tumor cells entering lymphatic vessels primarily includes the following steps: i) Cancer cell invasion through the basement membrane; ii) tumor-associated lymphoid hyperplasia; iii) tumor cell recruitment and clustering around lymphatic vessels; iv) secretion of cytokines by lymph endothelial cells to change the microenvironment and cause immune escape; and v) tumor cell entry into lymphatic vessels. The primary mechanisms of the last method include: i) Mechanical destruction of the lymphatic endothelial wall; ii) infiltration dependent on the CCL21 concentration gradient and via the lymphatic endothelial valve; iii) increased lymphatic permeability induced by mechanisms such as upregulation of α4β1 integrin and its ligand VCAM-1 in LECs; iv) release of chemicals to induce contraction of LECs to form an invasion site. The specific mechanisms are shown in Fig. 1.

Cancer cell invasion through the basement membrane

Tumor cells at the invasion front usually show infiltrative behaviors and penetrate into surrounding tissues in the form of cell stripes or clusters or individual cells, and this process is primarily mediated by epithelial to mesenchymal transformation (EMT) in cancer cells, which is an evolutionarily conserved cell process and is critical to embryogenesis and pathological reactions (such as wound healing or tissue repair) (14). EMT mainly occurs by activating Wnt or TGF-β signal transduction, hypoxia, and inflammation-related pathways, further inducing the expression of several key transcription factors of the twist, Snai1 and Zeb families in the TME (15). In turn, these transcription factors mediate several phenotypic changes in cancer cells; they mediate the downregulation of epithelial traits (including cell polarity and cell connectivity) and induction of mesenchymal characteristics, such as cytoskeleton remodeling and the expression of extracellular matrix (ECM)-degrading proteases, allowing cells to invade surrounding tissue effortlessly. It is worth noting that EMT is a gradual process and may occur throughout the entire process of tumor progression (16).

Tumor-associated lymphangiogenesis

Research has revealed that increased expression of the lymphatic angiogenic factors VEGF-C and VEGF-D can significantly promote LNM in esophageal squamous cell carcinoma (17). Compared with nonmetastatic tumors, metastatic melanoma is characterized by increased lymphangiogenesis, and the degree of tumor lymphangiogenesis is an important indicator for predicting the overall survival rate and LNM in patients (18). Tumor lymphangiogenesis and VEGF-C expression can serve as indicators of sentinel LNM during surgical resection of primary melanoma (19). In addition to increasing the quality of tumor-related lymphatic vessels, lymphangiogenic factors can also increase the expression of chemokines or adhesion molecules and receptors involved in tumor cell-LEC interactions by activating lymphatic endothelial cells, thus actively promoting cancer dissemination (20). In general, lymphatic hyperplasia occurs first in tumor LNM. The increased number and size of peripheral lymphatic vessels may provide more opportunities for cancer cells to infiltrate (lymph infiltration), but tumor drainage lymphatic vessels may also promote tumor proliferation by increasing lymphatic flow and drainage mediated by VEGF-C (21).

Tumor cells are attracted to and cluster around lymphatic vessels

Lymphatic hyperplasia serves as a prerequisite for LNM, which primarily involves cancer cell migration to lymphatic vessels and recruitment of cancer cells and supporting cells to the lymph nodes, providing a very conducive tumor microenvironment for cancer stem cells; this microenvironment regulates the antitumor immune response at the level of primary tumors and metastatic lymph nodes. In most normal tissues, lymphatic vessels can secrete a large amount of the chemokine CCL21, which can enter the lymphatic system by binding to the activated CCR7 receptor on DCs and can ultimately be excreted from lymph nodes to initiate an in vivo immune response (22). In vitro studies have confirmed that enhanced lymphatic flow can increase CCL21 production in the lymphatic endothelium (23). Importantly, when CCR7 was overexpressed in transplanted melanoma cells, LNM was greatly enhanced in an experimental tumor model, which indicates that LECs can serve as guiding clues for metastatic cancer cells in the physiological function of the immune system (24).

Chemokine CXCL12 (matrix-derived factor 1) was reported for the first time as a dependent factor involved in the in vivo lymphatic metastasis of Paget's disease (25). CXCL12 is upregulated in the lymphatic vascular endothelium of the subcapsular sinus of tumor-draining lymph nodes and tumor-associated lymphatic endothelial cells, while its receptor CXCR4 is expressed by invading tumor cells (26). In addition, the expression of CXCR3 in tumor cells is also associated with an increased LNM rate (27). CCL1 chemokines produced by the lymphatic sinuses mediate the entry of melanoma cells expressing CCR8 into the lymph nodes while blocking CCR8 suppresses LN metastasis. In particular, CCR8 inhibition leads to the stagnation of tumor cells in clusters of lymphatic vessels at the junction with the subcapsular LN sinus (28).

Tumor cell intravasation into lymphatic vessels

Previous studies have confirmed that tumor cells enter lymphatic vessels primarily via one of four ways: i) Mechanical injury or destruction of the lymphatic endothelial wall; ii) by detection of a CCL21 gradient and infiltrating through the junctions of the lymphatic endothelial valves; iii) by increasing lymphatic permeability, such as through cell growth factor (TGF-β1)-induced upregulation of α4β1 integrin and its ligand VCAM-1 in LECs; and iv) LEC contraction and formation of the invasion gate (CCID) induced by the release of chemical repellents [such as 12 (s)-hete] (29). Additionally, research has confirmed that CCR7+ cancer cells can escape the primary tumor and drift to the tumor-draining lymph nodes by increasing the expression of CCL21 in tumor-related lymphatic vessels via VEGF-C (30).

Lymph endothelial cells secrete cytokines to alter the microenvironment to facilitate immune escape

Lymph endothelium can not only provide a stable microenvironment for a cancer with stem cell-like characteristics but also provides a protective environment for long-term survival and subsequent metastasis of tumor cells. In addition, tumor cells may remain dormant in draining lymph nodes for a long period of time after primary tumor resection (31). The clinical observation of so-called ‘transit metastasis’ (that is, a metastatic tumor that has developed in the lymphatic vessel between the draining LN and the primary tumor) revealed that CXCR4 melanoma cells (CD133+ melanoma cells) were located near the lymphatic vessel-producing CXCL12 in the metastatic lymph nodes and lungs. The metastatic activity of CXCR4+/CD133+ cells was higher than that of CXCR4-/CD13- cells. The study also confirmed that the combined use of the alkylating agent dacarbazine and CXCR4 blocker, which are widely used to treat human melanoma, is significantly better than the single use of dacarbazine in inhibiting the growth, migration, and metastasis of melanoma (32). In addition to these direct effects of LECs on the survival of cancer cells, lymphatic vessels may also provide an immunoprotective microenvironment through the secretion of chemokines (33). Studies have shown that CCL21 may transform the host immune response from immunogenicity to tolerance, which may potentially promote tumor progression (34). It has been reported that LN lymphatic vessels activated by VEGF-C not only promote tumor metastasis of melanoma but also induce immune tolerance, which increases the difficulty of treatment (35). Recently, study revealed that LECs induce tolerance via programmed cell death 1 (PD1) ligand 1 (PD-L1) and lack of costimulation leading to high-level PD-1 expression on CD8 T cells (36). Therefore, the activities and interactions of various cells in the TME play a decisive role in lymphatic metastasis.

3. Role of the tumor microenvironment in the lymphatic metastasis of CC

Characteristics of the tumor microenvironment in CC

The unique tumor microenvironment of CC is primarily estrogen receptor α (ERα) matrix activation, sustained high-risk human papillomavirus infection, hypoxia, and matrix and immune inflammatory cells (mainly including CAFs, TAMs and MDSCs), T cells, and neutrophils, which ultimately promote angiogenesis and inflammation (37,38). First, 17-β-estradiol (E2) interacts with the matrix ERα on the surface of myofibroblasts and fibroblasts and further induces an increase in the secretion of anti-apoptotic factors, inflammatory chemokines, extracellular matrix enzymes, and proangiogenic factors. In addition, epithelial cells with persistent high-risk HPV infection can attract monocytes (monocyte chemoattractant protein-1 and macrophage inflammatory protein-3α), natural killer cells, and Th17 lymphocytes. Positive expression of cancer proteins (E6 and E7) in high-risk HPV strains can trigger a series of events to promote CC metastasis through their host cervical epithelial cells. In short, in contrast to the matrix progesterone cascade mediated by the progesterone receptor (PR), the synergistic effect between the activity of stromal ERα and high-risk HPV oncoproteins induces CC proliferation and promotes inflammation and angiogenesis, participating in mesenchymal-epithelial and EMT changes (39,40).

In CC, the lymphatic vessels in the diffusion area of tumor cells are in a dynamic process of change, and the lymphatic changes that promote tumor metastasis play a leading role in solid tumor metastasis (41). The biggest obstacle for tumor cells to invade lymphatic vessels is the intact lymphatic vessel structure, which makes it difficult for them to break through. The integrity of lymphatic vessels is mainly related to proteins in the adhesion links between LECs (42). In addition, the complete lymphatic endothelial barrier and sound repair function also depend on the interstitial cells and their secreted cytokines in the surrounding environment (43). The stromal cells and immunoinflammatory cells in the TME can secrete certain cytokines, such as VEGF-C/D, to promote the formation of lymphatic vessels, induce the inactivation of CD4+ and CD8+ T cells, help tumor cells escape immune surveillance, induce tumor cell EMT and enhance tumor cell invasive ability, finally leading to lymphatic metastasis of CC (44). Studies have confirmed that hypoxia can induce the enhancement of the invasive capacity of CC cells and foster a tumor-supporting environment through increased CCL8 secretion and TAM recruitment to promote lymphatic vessel entry and angiogenesis (38). In this review, the mechanisms of hypoxia, and the involvement of stromal components and immune inflammatory cells in the tumor microenvironment in the lymphatic metastasis of CC is summarized.

The role of hypoxia in the lymphatic metastasis of CC

Hypoxia is a key factor that has been identified to lead to a poor prognosis in patients with prostate cancer, pancreatic cancer, breast cancer, head and neck cancer, and other types of tumors. Targeting hypoxia is one of the directions of LNM imaging and staging. Therefore, addressing tumor regional hypoxia is an urgent problem to improve cancer prognosis. The main reasons for the formation of a hypoxic tumor microenvironment are imbalances in blood supply and abnormal tumor metabolism. Once the hypoxic microenvironment is formed, the tumor invasive and metastatic abilities are enhanced, and the tumor cells can present features of a dormant state to avoid immune surveillance, resulting in the failure of immunotherapy. In addition, the activation of HIF can induce EMT, which is the first step for tumor cells to break through the basement membrane and metastasize to a distant location (45). In addition, hypoxia has been proven to stimulate molecular changes in cancer cells to facilitate a state of mitotic arrest to make the cells appear dormant. Increasing evidence shows that dormancy is crucial for cancer cell survival; the cells need to delay their invasive behaviors until the distant ‘hostile’ microenvironment is transformed to enable them to escape from immune surveillance upon treatment (46). However, the underlying mechanisms of cancer cell EMT, progression and escape from apoptosis/necrosis in hypoxic environments remain unclear. Ju et al (47) found that CSN8, as a key factor in hypoxia-induced cell dormancy and EMT occurrence, can promote colorectal cancer cells to evade immune surveillance and attack, significantly improving their invasive and metastatic ability. Hsin et al (48) found that upregulation of carbonic anhydrase IX can promote EMT, and this phenotype combined with upregulation of PFKFB4 ultimately improves the migration and metastasis of CC cells. Hypoxia induces EMT in CC to facilitate further lymphatic metastasis.

The difficulty in early prediction and research of LNM lies in the lack of highly specific molecular markers, especially markers of lymphatic vessels and blood vessels around tumors, which has led to extremely slow progress in research on LNM for decades. Breakthroughs have been made following the discovery of VEGF-C/D and its specific receptor VEGFR-3; thus, research on lymphatic metastasis is gradually increasing. Sugiura et al (49) found that in oral tumors, hypoxia in the surgical area can lead to an increase in local lymphangiogenesis, accompanied by a significant increase in CD11b+ cell infiltration and LNM. In CC, Cairns and Hill (50) found that acute hypoxia can reduce the volume of the primary tumor lesion in nude mice but significantly increases the number of LNMs. Chaudary et al (51) found that hypoxic treatment of CC cells can significantly upregulate VEGFR3 and promote lymphatic metastasis. Downregulation of VEGFR3/VEGFC or the use of VEGFR3/VEGFC blockers can significantly reduce hypoxia-induced tumor lymphatic proliferation and metastasis. These studies further confirm that hypoxic conditions are more conducive to the increase in VEGFR3/VEGFC and promote the lymphatic metastasis of CC. Lee et al (52) found that the prognosis of CC patients with LNM is poor. Identifying novel treatment methods based on the expression of CA9 to prevent LNM is expected to significantly improve the prognosis of CC patients. CA9 is currently recognized as one of the most commonly used markers of hypoxia (53,54). Kim et al (55) found that extended-field irradiation (EFI) has a significant inhibitory effect on the recurrence of para-aortic lymph nodes in CA9-positive tumor patients, but it is not significant for improving long-term survival. The reason may be related to increased local and distant metastasis rates. Hypoxia promotes LNM by inducing the release of lymphangiogenic factors, such as VEGF-C, from malignant tumor cells to induce lymphatic dilation (lymphangiogenesis) of the primary tumor and draining sentinel LN.

Hypoxia can also lead to lymphatic metastasis through the release of lymphangiogenic active factors and other factors that recruit TAMs to accumulate in lymphatic vessels and form metastatic areas. Chen et al (56) found that high expression of ZEB1 under hypoxic conditions can promote tumor metastasis by increasing TAM recruitment and CCL8 secretion. Targeting ZEB1 can improve the prognosis of patients with metastatic tumors by destroying the hypoxic microenvironment. They also showed that hypoxic TAMs near lymphatic vessels are the primary cells producing IL-10, and a sharp increase in IL-10 concentration induces upregulation of Sp1 expression in LECs, promoting lymphatic angiogenesis in tumors (57). They further confirmed that macrophages recruited to the hypoxic microenvironment of CC tend to transform into the M2 phenotype and induce an increase in Nrp-1 in CC. This suggests that reversing the polarization of TAM towards an M2 phenotype and interfering with Nrp-1 represents a novel strategy for improving the hypoxic microenvironment of CC (58). As an important feature of solid tumors, hypoxia accompanies almost the entire process of tumor metastasis. The hypoxia of the tumor increases invasion and metastasis and helps disguise tumor cells as cells in a dormant state to avoid immune monitoring and attack, increasing the risk of chemotherapy and immunotherapy resistance. Targeted hypoxia therapy is an important direction for improving patient prognosis in the future.

The role of CAFs, TAMs, MDSCs, and immunoinflammatory cells in the lymphatic metastasis of CC

In addition to hypoxia, stromal components and inflammatory immune cells also play an essential role in the process of lymphatic metastasis of CC. These cells will change in morphology and function with tumor progression to promote tumor invasion, migration, lymphatic metastasis, and hematogenous migration. Here, a focus on the role of CAFs, TAMs, MDSCs, and immune cells in the lymphatic metastasis of CC is discussed.

CAFs

One of the primary obstacles in tumor cell infiltration through the lymphatic system is the integrity of the lymphatic endothelium, which is closely related to the protein complexes that make up junctions between endothelial cells. In addition, cell homeostasis, and cytokine dynamic balance in the microenvironment around lymphatic vessels are the basis for maintaining the structural integrity and barrier function of the lymphatic endothelium. CAFs have important physiological functions in maintaining the stability and integrity of most tissues. This function is realized during the progression of metastasis and can induce the formation of a metastasis-promoting microenvironment (43).

Activated CAFs can change the components of the ECM to reshape the tumor microenvironment, promote local angiogenesis, tumor cell proliferation, and metastasis and play a role in the formation of chemotherapy resistance by activating multiple signaling pathways and secreting activated cytokines. The signaling of CAFs and tumor cells plays an important role in tumor progression and treatment. The current view is that CAFs and the tumor immune microenvironment (TIME) mainly promote tumor progression. The antitumor components in the TIME and TME are in dynamic balance and are primarily composed of immune cell populations in tumors. CAFs interact with tumor-infiltrating immune cells to form a tumor-immune suppressive microenvironment by secreting growth factors, cytokines, exosomes, chemokines, and other effectors, assisting tumor cell escape from immune surveillance and attack and promoting tumor cell proliferation and distant metastasis (59). Deep investigation of the mechanisms and interactions between CAFs and tumor cells, as well as between CAFs and other immune cells, may provide novel ideas for immunotherapy.

Previous studies have confirmed that TGF-β1-activated CAFs promote breast cancer EMT, invasion, and metastasis by overexpression of FAP-α, and autophagy in breast cancer. Treatment with both the autophagy inhibitor 3-methyladenine and FAP-α knockdown can reverse EMT and eliminate lung metastasis and invasion caused by CAFs, indicating that autophagy and FAP-α in CAFs are prerequisites for the metastasis of breast cancer to the lungs (60). Wang et al (61) revealed that epiregulin reprograms CAFs via the JAK2/STAT3 pathway to facilitate oral squamous cell carcinoma invasion. Zhou et al (62) found that CAFs induce EMT functional changes in tongue squamous cell carcinoma. In CC, Murata et al (63) reconstituted a metastatic TME by co-transplanting CAFs and cancer cells into nude mice to reconstruct the microenvironment of tumor metastasis. It was surprising to find that 40% of nude mice co-transplanted with two kinds of cells had LNMs, while nude mice transplanted with a single cancer cell had no LNMs. They also showed that CC CAFs secreted large quantities of heparin-bound epidermal growth factor (HB-EGF), and the platelet-derived growth factor produced by ME180 cells enhances the expression of CAF HB-EGF, which in turn can significantly promote the proliferation of ME180 cells (64). Xiao et al (65) found that overexpression of TGF-β1 and SDF-1 in CAFs promoted colony formation, growth, and invasion of CC cells, while when cocultured with TGF-β1 and SDF-1 neutralizing antibodies, these phenomena were reversed. CAFs secrete a series of growth factors through interaction with tumor cells, causing tumor cell invasion, migration, and EMT, eventually leading to LNM of CC, as confirmed by in vivo experiments.

CAFs, as matrix-supporting cells around LECs, play a supportive role in maintaining the lymphatic endothelial barrier. Before lymphatic metastasis, CAFs can induce LNM by damaging the lymphatic endothelial barrier. Wei et al (66) found a new subgroup of CAFs, namely, periostin+ CAFs, which significantly increased infiltration in patients with CC LNM, and the more infiltration there was, the poorer the prognosis was. Further mechanistic research confirmed that periostin+ CAFs activated the integrin FAK/Src-VE cadherin signaling pathway in LECs, disrupted the lymphatic endothelial barrier, allowed cancer cells to enter the lymphatic vessels, and ultimately cause CC LNM. If the FAK/Src-VE cadherin signaling pathway was inhibited, the effect of periostin+ CAFs was weakened. This study highlights a novel approach to the treatment of CC LNMs, identifying potential targets for blocking CAF-dependent metastasis that destroys the lymphatic endothelial barrier and strengthening and consolidating the integrity and stability of the lymphatic endothelial barrier is essential for blocking CAF-dependent LNM. These studies comprehensively show that CAFs can play a key role in CC lymphatic metastasis by impairing lymphatic endothelial barrier function and secreting growth factors and activating related signaling pathways to promote tumor invasion. Targeting CAFs is thus a relatively novel method for preventing CC lymphatic metastasis.

TAMs

TAMs are involved in almost the entire process of tumor occurrence and development. In the early stages of tumor development, the immune system quickly responds, summoning T cells and macrophages to attack and clear tumor cells through killing and phagocytic functions. However, under certain conditions, macrophages can be easily educated by tumors and converted into TAMs, which can actually promote tumor progression and assist tumor cell metastasis. TAMs can also assist in local infiltration and distant dissemination of tumor cells by participating in lymphangiogenesis and angiogenesis. To a certain extent, TAMs can aggregate and even play a leading role in tumor cell metastasis. Previous studies have confirmed that depletion of TAMs is equivalent to turning off the switch on angiogenesis, and eliminating Tie2 TAMs can inhibit angiogenesis in mouse glioma (67,68).

Hypoxia is a common phenomenon in most solid tumors, as there are several novel blood vessels in the hypoxic area due to low oxygenation. Therefore, hypoxia is considered the primary driving force for angiogenesis. Research has revealed that hypoxic areas often accompany the aggregation of TAMs, which is primarily caused by hypoxia stimulating the production of a series of chemotactic and active factors, such as CCL-2, CXCL4 and VEGF, in the tumor and interstitial cells. In addition, TAMs can respond to hypoxia by upregulating the expression of HIF and downstream proangiogenic factors (69). Therefore, as the cells at the leading edge of the invading tumor, TAMs are also known as the cells that aggregate to form the premetastatic niche; thus, these cells can be used to identify the direction of tumor cell metastasis to blood and lymphatic vessels and assist in the prevention of metastasis.

The evidence that TAMs are directly involved in lymphangiogenesis is that TAM depletion can significantly weaken VEGFC and VEGFR3 signaling in LECs, weakening lymphatic vessel formation in early-stage tongue SCC (70,71). Hosono et al (72) found that in esophageal squamous cell carcinoma (ESCC), TAMs can release CXCL8 and bind to CXCR1/2 (known as CXCL8 receptors) of ESCC cell lines, promoting ESCC invasion by suppressing Akt and Erk1/2 phosphorylation. Neutralizing antibodies against CXCL8, CXCR1 and CXCR2 can inhibit these effects. Clinical analysis confirmed that CXCL8+ TAMs are associated with LNM in esophageal cancer. Chen et al (57) uncovered a new LNM model for CC in the hypoxic TME: ZEB1 increases CCL8 secretion and recruits TAMs to encapsulate the lymphatic vessel to form network centers, promoting CC LNM.

There are several studies on M2-like macrophages in the invasion and LNM of CC. Guo et al (73) found that the infiltration of CD68+ TAMs and CD163+ M2 TAMs is correlated with tumor progression. CD163+ M2 TAM infiltration is correlated with LNM and an advanced FIGO stage. Tan et al (74) found that activated T-cell nuclear factor 1+ TAMs, as the M2 TAM subtype, are significantly more abundant in CC tissues and can promote CC cell proliferation and metastasis by activating the c-Myc/PKM2 pathway. Jiang et al (75) found that during the progression from CIN I-III to stage I-IV CC, the levels of TAM aggregation and neovascularization in the TME increased synchronously, which fully confirmed that TAMs and tumor angiogenesis play a key role. Li et al (76) found that the number of M2-TAMs in CC was higher than that in the surrounding tissues, and the number of M2-TAMs in the diffusion infiltration pattern (DIP) was higher than that in a pushing border pattern (PBP), indicating a strong relationship between M2-TAMs and the invasive behavior of CC. The above research comprehensively showed that TAMs promote the LNM of CC. The identification of its regulatory mechanism not only provides a novel target for the development of therapies to counter metastasis but also provides a basis for selecting specific patient cohorts that may benefit from certain molecular-targeted drugs.

MDSCs

MDSCs are a group of heterogeneous immature myocytes that are blocked from maturing in cancer. They are one of the primary driving forces behind the immunosuppressive TME. According to phenotypic and morphological differences, these cells can be divided into two subgroups: Granulocytic MDSCs (G-MDSCs), which are similar to neutrophils, and monocytic MDSCs (Mo-MDSCs) (77). The increase in the number of MDSCs in CC patients was first confirmed in 2014 and the number of MDSCs, especially G-MDSCs, significantly increased in the tumor tissue of CC patients. MDSCs are important immune components in the TME and are considered to mediate the immunosuppression of tumor-bearing mice and cancer patients (78). In addition to enhancing immunosuppression, MDSCs have also been shown to enhance tumor progression by stimulating cancer cell invasion, metastasis, and tumor angiogenesis (79). In the TME of CC, tumor cells secrete various molecules to recruit MDSCs from immature myocytes, including granulocyte colony-stimulating factor (G-CSF) (80), IL-6(81), and highly expressed C-X-C chemokine receptor 2 (CXCR2) chemokines, such as CXCL1, CXCL2, CXCL3, CXCL5 and CXCL8(82). Mo-MDSCs enhance the stemness of pancreatic cancer cells by producing IL-6 and subsequently activating STAT3 activator in cancer cells (83). Peng et al (84) recently showed that MDSCs enhance the dryness of breast cancer cells by producing IL-6 and nitric oxide and subsequently activating the STAT3 and Notch signaling pathways, respectively. Kuroda et al (85) found that G-CSF-induced MDSCs enhance the dryness of CC cells by producing prostaglandin E2 (PGE2). It was also demonstrated that the inhibition of MDSCs or PGE2 effectively inhibits the induction of CSCs and enhances the efficacy of cisplatin in CC. Ni et al (86) revealed that patients with high levels of METTL3 and CD33 expression in MDSCs in tumor tissues have a poor prognosis, and these phenotypes are independent risk factors for the prognosis of CC patients. Heeren et al (87) found that elevated MDSC levels increase CC LNM and weaken sensitivity to radiotherapy and chemotherapy. Rodríguez and Ochoa (88) further confirmed that MDSC-mediated niche formation before LNM can induce FDG uptake during FDG positron emission tomography/CT scans and leads to false-positive detection of an LNM. Using the HPV-mediated CC mouse model, researchers have demonstrated that MDSCs mediate immunosuppressive activity through IL-6/JAK/STAT3 signaling. The activation of STAT6 mediated by the proinflammatory cytokine IL-3 may be the reason for the expansion of MDSCs, which then accelerates tumor growth (89). In addition, MDSCs interact with B lymphocytes in the TME of CC through B-cell activating factor (BAFF) expressed on the surface of MDSCs. MDSCs induce B cells to differentiate into Breg cells by acting on BAFF receptors expressed in B cells. In addition, IL-10 secreted by Breg cells can promote STAT3 phosphorylation and activate MDSCs, thereby establishing a positive feedback loop. The continuous differentiation of Breg cells and the activation of MDSCs induce immunosuppressive states and lead to tumor immune escape in CC patients (90). MDSCs also stimulate tumor angiogenesis by secreting Bv8 (a proangiogenic molecule), which increases the expression of tumor G-CSF and MDSCs. CC patients have poor survival rates, and G-CSF-producing CC is sensitive to cisplatin after splenectomy or administration of anti-Gr-1 antibodies, leading to the depletion of MDSCs (91). In summary, MDSCs utilize multiple mechanisms to enhance the proliferation and metastasis of CC. After being recruited to the TME, they mainly exert strong immunosuppressive effects by inhibiting T-cell function.

Immune and inflammatory cells

During the immune escape process of tumors, regulatory T cells (Tregs) secrete large quantities of IL-10, TGF-β, and IL-35, which inhibits antigen presentation by dendritic cells and CD4 helper T-cell function, regulates the expression of inhibitory receptors and CD8-tumor-infiltrating lymphocyte (TIL) depletion-related transcriptome characteristics, promote T-cell depletion in tumors, downregulate antitumor immunity, and produce tumor-specific CD8 cytotoxic T lymphocytes (92). As the role of adaptive immune cells in the tumor microenvironment has been elucidated, the first treatment scheme that interferes with the function of T cells has been successfully approved by the U.S. FDA for antibody-based treatment of patients with advanced cancers, such as Nivolumab, Balstilimab and Zalifllimab. Wu et al (93) showed that a significant number of CD4+ CD25+ FOXP3+ Treg cells accumulate around tumor cells and that the proportion of FOXP3+ T cells in CC is higher than that in cervical intraepithelial neoplasia. Moreover, the proportion of FOXP3+ T cells in CC with LNM is significantly higher than that in CC without LNM. Nakamura et al (94) found that the higher the Foxp3+/CD4+ Treg cell ratio was, the greater the rate of LNM was. Foxp3+ Treg cells contact the immunoregulatory enzyme indoleamine 2,3-dioxygenase (IDO)+ APC. Foxp3+ Treg cells form a premetastatic microecology and establish a network with IDO to induce immune escape. Compared with that in the satellite lymph nodes without tumors, the proportion of Foxp3 Treg cells is significantly increased in lymph node metastases (94). There is a high incidence of LNMs at Foxp3 Treg cell aggregation sites. Foxp3+ Treg cells can directly contact IDO APCs in the context of LNM. In the metastatic microenvironment of CC, Treg cells first infiltrate and accumulate to form an immunosuppressive microenvironment to attract cancer cells to the site of metastasis. These studies provide evidence that the recruitment of Foxp3+ Tregs can promote cancer cell migration.

Previous studies have noted that based on flow cytometry analysis, CC patients with a low CD8 T-cell/Treg ratio, high Treg level, and high levels of PD-L1 and major histocompatibility complex, class II, DR (HLA)-DR myeloid cells are more likely to have LNMs; therefore, this environment can also be considered an immunosuppressive microenvironment. Heeren et al (95) found that delineated fields of Treg-associated immune suppression in anatomically colocalized tumor-draining lymph nodes primarily form a micro-transfer microenvironment to further integrate and expand to further regions, which provides a basis for early surgical intervention. In another study by the same lab, it was found that a higher number of CD8+ T cells significantly reduced the frequency of CD4+ cells, increased the expression of the memory marker CD45RO and activation markers [PD-1, inducible T cell costimulator, HLA-DR and cytotoxic T-lymphocyte-associated protein (CTLA-4)], and significantly promoted LNM. Furthermore, they discovered that in metastatic lymphoid tissue, the proportions of the FoxP3+ Tregs and regulatory antigen-presenting cells (APCs) [PD-L1+ CD11c(hi), CD14+], APC/myeloid suppressor cell subpopulations increased significantly. After treatment with different Toll-like receptor ligands, the expression of IFN-γ in metastatic lymphoid tissue significantly decreased, but IL-10, IL-6, and TNF-α expression significantly increased (87). Wang et al (96) used 18F fluorodeoxyglucose microPET/CT and bioluminescence imaging analysis of mouse neck xenograft tumors to confirm that PD-L1 overexpression promoted tumor glucose uptake by activating the ITGB4/SNAI1/SIRT3 signaling pathway, ultimately leading to LNM. At present, it is suggested that targeting PD-L1 and CTLA4 is a potential approach for the treatment of LNM in CC.

Research on the relationship between tumors and inflammation is developing constantly. Tumor-induced inflammation can result in DNA damage and micrometastases. Systemic inflammatory responses can aggravate malnutrition in patients. Inflammatory indicators have certain reference values in the diagnosis and prognosis of various malignant tumors, and research has confirmed that inflammation may promote tumor progression (97). Zhang et al (98) found that the neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) were valuable for predicting LNM in gastric cancer. The NLR is superior to the PLR in predicting the gastric cancer survival rate. Ayhan et al (99) found that a high NLR and PLR was significantly correlated with tumor volume (>2 cm) and deep muscle interstitial infiltration. A high monocyte-to-lymphocyte ratio was significantly correlated with abdominal aortic LNM, pelvic LNM, and locally advanced CC (IB3-IIIC2). Lee et al (100) confirmed that an NLR ≥3.1 was indicative of a shift in the immune response from tumor suppression to cancer promotion, often accompanied by radiation resistance, rapid tumor progression, and LNM, which is associated with a poor prognosis in CC patients. The use of inflammatory indices in predicting the clinical outcomes of patients with lymphatic metastasis of CC is deserved of further attention in light of the convenient and low-cost means of access to the data.

4. Therapeutic strategies for targeting the TME of CC

The standard method of treatment for advanced CC includes radiotherapy and chemotherapy; however, the patient survival rate is very low (101). In recent years, the gradual promotion and application of immunotherapy has provided renewed hope. The role of the TME in tumor metastasis has been confirmed; cancer cells can recruit Tregs, downregulate cancer cell surface antigens, induce T-cell apoptosis, secrete immunosuppressive factors, and evade immune surveillance and attack, eventually forming the immunosuppressive TME on which they rely for survival (102). The primary purpose of immunotherapy is to reactivate the antitumor immune response or reshape the immunosuppressive microenvironment. The US Food and Drug Administration (FDA) has approved three immunotherapy-based drugs for the treatment of CC: Pembrolizumab, Tisotumab Vedotin and Nivolumab. Other immunotherapeutic strategies for the TME are still in the exploratory stage, and the progress of TME treatment strategies for CC is briefly summarized below.

Targeting hypoxia

Hypoxia increases the risk of local invasion, metastasis, and treatment failure in CC. Hypoxia represents an attractive therapeutic target, and a number of strategies have been researched. The known studies on hypoxia-targeted strategies are as follows: i) Increasing intratumoral oxygen: A phase II clinical trial of 139 patients with locally advanced CC concluded that the addition of carbogen and nicotinamide hypoxia modification to standard therapy was feasible and safe (103). ii) Decreasing tumor oxygen consumption: Metformin, an antidiabetic agent, reduces cancer incidence in patients with diabetes (104). Subsequent experiments highlight a complex interplay with hypoxia-associated molecular pathways, possibly through the inhibition of the mammalian target of the rapamycin-HIF-1α axis (105). Two phase II randomized clinical trials, NCT02394652 and NCT04275713, are currently investigating the use of metformin as a hypoxia-modifying therapy for locally advanced CCs. These trials will assess metformin-induced changes in tumor hypoxia using imaging and gene expression biomarkers (3). Hypoxia-specific radiosensitizers and cytotoxins: There are several classes of hypoxia-specific cytotoxins. Quinone-based agents selectively activate hypoxia through a reductive mechanism and induce DNA alkylation-mediated cytotoxicity. Sharma et al (106) performed a study with 160 patients with locally advanced squamous cell carcinoma of the uterine cervix that participated in a multicenter phase III trial that randomized participants to receive radiotherapy alone or radiotherapy with concomitant mitomycin C. Despite improved 4-year disease-free survival rates in the intervention group, the study failed to show a significant benefit in overall survival or local recurrence rates. Discovered ~35 years ago by Zeman et al (107) and Brown (108), tirapazamine was the first purely hypoxic cytotoxin and is one of the most advanced bioreductive drugs in clinical trials. The best-known aromatic N-oxide is used as an anticancer drug that undergoes enzymatic one-electron reduction and converts to an electron-donating mono-N-oxide metabolite (tirapazamine radical). Murine model experiments showed considerably more tumor cell death was observed when tirapazamine was combined with radiotherapy or cisplatin chemotherapy compared with monotherapy (108), but unfortunately, the follow-up clinical trial of DiSilvestro et al (109) failed to show a clinically meaningful result. iv) Hyperthermia: A strategy that encompasses a variety of hypoxia-targeting mechanisms is hyperthermia. It is assumed to improve oxygenation by causing vasodilatation, direct cellular damage, immune-mediated killing of tumor cells, and inhibition of DNA repair. In CC, hyperthermia has been used to sensitize tumors to radiotherapy, and the evidence suggests that combining radiotherapy with hyperthermia results in improved locoregional control when compared with using radiotherapy alone (77 vs. 52%) (38). Effectively decreasing hypoxia would clearly improve the response to therapy and reduce the likelihood of metastatic spread in CC.

Targeting immune checkpoint molecules

The tumor microenvironment plays a key role in tumor metastasis. Cancer cells can recruit Tregs, downregulate tumor antigen expression, induce T-cell tolerance and/or apoptosis, and generate immunosuppressive cytokines to stimulate immunosuppressive immune checkpoints, which leads to a unique and highly immunosuppressive tumor microenvironment (TME) (102). To overcome these immunosuppressive conditions, immune checkpoints may be modulated by either agonist or antagonist monoclonal antibodies used to enhance T-cell activation and eliminate inhibition of T-cell activation, respectively, to reactivate T cells to attack tumors (110). At present, immune checkpoint inhibitors in CC primarily include the following: i) Programmed death ligand 1 (PD-L1): PD-L1 is an immunomodulator that is expressed on antigen-presenting cells (APCs) and 20-50% of human cancer cells. Tumor-induced PD-L1 inhibits T-cell function and induces immune tolerance but also induces T-cell apoptosis. By contrast, PD-L1 induces expansion registration of T cells. Therefore, blocking this ligand on tumor cells and APCs can improve tumor defense, and T cells with anticancer properties can restore their effector functions (111). ii) Anti-CTLA4 antibody: Under physiological conditions, T cells are stimulated by CD28, and CD28 interacts with B7-1 and B7-2 on dendritic cells. In addition to the ‘key’ CD28, T cells also express CTLA4, which can be regarded as ‘key off’. CTLA4 acts as a symbiotic factor on activated T cells to regulate their immune response (112). The application of immune checkpoint inhibitor targeting in CC is summarized in Table I.

Table I

Summary of clinical trials targeting immune checkpoint molecules in cervical cancer.

Table I

Summary of clinical trials targeting immune checkpoint molecules in cervical cancer.

First author, yearNCT Number/phase of clinical trialImmunotherapeutic regimenAdditional therapyPatient population (n)Trial status/clinical efficacy(Refs.)
Naumann, 2019NCT02488759/Phase 2Nivolumab (anti-PD-1 antibody)  Recurrent/metastatic cervical, vaginal or vulvar cancer (24 participants)Completed/ORR, 26.3%; MOS, 21.9 months(113)
O'Malley, 2022NCT03495882/Phase 2Balstilimab (anti-PD-1 antibody) and Zalifrelimab (anti-CTLA-4 antibody) Recurrent and/or metastatic cervical cancer (155 participants)Completed/ORR, 25.6%; PDL-1+, 32.8%; PDL-1(-): 9.1%.(114)
Santin, 2020NCT02257528/Phase 2Nivolumab (anti-PD-1 antibody) Persistent, recurrent, metastatic cervical cancer (26 participants)Completed/PFS and OS at 6 months were 16 and 78.4%, respectively.(115)
Frenel, 2017NCT02054806/Phase 1Pembrolizumab (anti-PD-1 antibody) Advanced cervical cancer (42 participants)Completed/ORR, 17%(116)
Chung, 2019NCT02628067/Phase 2Pembrolizumab (anti-PD-1 antibody) Advanced cervical cancer (46 participants)Completed/ORR, 12.2%; PD-L1+ ORR, 14.6%(117)
De Jaeghere, 2023NCT03192059/Phase 2Pembrolizumab (anti-PD-1 antibody)Radiotherapy, vitamin D, Aspirin, Lansoprazole, Cyclophosphamide, and CurcuminRefractory or persistent endometrial, cervical, or uterine cancer patients (43 participants)Completed/ORR, 11.1%; MOS, 39.6 weeks(118)

[i] ORR, objective response rate; MOS, median overall survival; NCT, National Clinical Trial; HPV, human papillomavirus; PD-1, programmed cell death 1; PD-L1, PD1 ligand 1; CTLA, cytotoxic T-lymphocyte-associated protein; PFS, progression-free survival.

Early data suggested that immune cells, in particular CD8+ T cells, play a key role in tumor cell death within a radiation field (119). Radiation therapy causes migration of dendritic cells and cross-penetration of tumor antigens, which can result in T-cell activation and proliferation. Furthermore, radiation therapy increases the density of TILs within a tumor, likely via extravasation of TILs within the vasculature of tumors and chemokine activation (120). It is known that radiation therapy alters the T-cell receptor repertoire of peripheral T-cell clones (121). Thus, there is a strong rationale for the combination of radiation with immune checkpoint blockade. There are limited data surrounding the optimal dose and fractionation needed to provoke an ideal immune response when combining immunotherapy with radiation in CC. Sequencing of CTLA-4 blockade with immunotherapy in preclinical models demonstrates that when anti-CTLA-4 is delivered prior to radiotherapy, there is increased efficacy compared to delivery after radiotherapy (122). Studies have also demonstrated that radiotherapy increases PD-L1 expression, which may act as a negative feedback mechanism preventing T-cell-mediated tumor rejection (123). Radiotherapy and chemotherapy combined with PD-1 and CTLA-4 immune checkpoint blockades provide a more effective scheme than monotherapy for the treatment of advanced and recurrent cervical cancer.

Targeting suppressive immune cells

Suppressive immune cells, such as Tregs, MDSCs, and type 2 macrophages, form an immunosuppressive microenvironment to assist tumor cell escape from immune surveillance. Research has shown that practical CXCR2 antagonist therapy can weaken the proliferation and migration of CC cells (124). In addition, the method of targeting the CSF-1/CSF-1R axis of TAMs is being assessed in a mouse model. CSF-1R inhibition weakened the turnover rate of TAMs and increased the number of CD8 T cells infiltrating tumor tissue (125). The antitumor effect of anti-PD-1 therapy is enhanced by inhibiting CXCR2, the primary chemokine receptor for MDSC recruitment in human pancreatic cancer (126). However, these studies are still limited to in vitro and in vivo experiments, although they provide novel ideas for future clinical trials. In addition, metabolites targeting suppressive immune cells, such as Arg-1 and IDO, are also novel avenues for targeting suppressive immune cells. The arginase inhibitor INCB001158 is being used to treat metastatic solid tumors (NCT02903914). Treatment of IL-6 knockout mice with IDO inhibitors has been proven to inhibit the expression of IDO. In addition, combination therapy with therapeutic vaccines leads to a decrease in polymorphonuclear MDSCs and Treg cells in tumors, supporting IL-6 and IDO as immunometabolic adjuvants for immunotherapy against CC (127). Combination therapy targeting inhibitory immune cells and metabolites within the TME represents a new strategy for antitumor therapy.

Anti-lymphangiogenesis and anti-inflammatory therapy

Since lymphatic vessels and lymphatic remodeling play a key role in lymphatic metastasis, targeting lymphatic vessels is the key to the treatment of metastatic CC. The VEGF-C/VEGFR-3 signaling axis induces tumor lymphatic vessel formation. In an experimental model, blocking VEGF-C/VEGFR-3 has been proven to reduce tumor lymphatic vessel formation and metastasis (128). However, this study has not yet entered a clinical trial stage. Inflammation plays a certain role in tumor metastasis. Nonsteroidal anti-inflammatory drugs combined with chemotherapy and radiotherapy can increase the sensitivity of patients with locally advanced cervical cancer. Studies have confirmed that blocking the inflammatory signaling pathway (COX/PGE2) and regulating the immune response against HPV and targeting the virus are the best choices for antitumor treatment of cervical cancer (129). The interaction of various cells in the TME is very complex, and the effect of any single therapy is limited. Combination therapy may provide a breakthrough for improving the prognosis of patients with recurrent and metastatic CC in the future.

5. Conclusions and future perspectives

Lymphatic metastasis is a key factor affecting the prognosis of patients with cervical cancer. CAFs, TAMs, and immune and inflammatory cells (primarily T cells and neutrophils) in the tumor microenvironment promote lymphatic metastasis by releasing a series of cytokines (such as VEGF-A/C/D and TGF-β) to induce tumor cell EMT, lymphatic vessel proliferation, and immune evasion, which ultimately leads to lymphatic metastasis. The above effects are enhanced under hypoxic conditions through hypoxia-related signaling pathways and transcription factors (such as HIFs). Although progress has been made in clinical trials on hypoxia-targeting strategies, and PD-1 and CTLA-4 immune checkpoint blockades in advanced CC, there are few clinical trials and drugs that specifically target markers for predicting and treating lymphatic metastasis in CC (130). A few clinical trials have shown that simultaneous radiotherapy combined with immunotherapy is more effective than monotherapy, but the specific mechanism remains unclear, and it is meaningful to expand the population to further study the mechanism of action to guide clinical treatment. The tumor heterogeneity of individual CC patients increases the complexity of treatment and leads to differences in the involvement of factors related to lymphatic metastasis among patients. Hypoxia can allow tumor cells to appear dormant, increase the difficulty of treatment, and recruit more TAMs as a lymphatic angiogenesis switch. TAMs directly participate in lymphatic angiogenesis. CAFs damage the lymphatic endothelial barrier and destroy the integrity of the lymphatic endothelium, and immune-inflammatory cells to create an immunosuppressive microenvironment. These complex and orderly steps involved in tumor microenvironment formation eventually leading to LNM. However, the mechanism is still unclear. There remain several aspects that need to be studied and explored in the future to understand and reduce the incidence of lymphatic metastasis of CC and improve the survival rate. Development of individualized treatments based on the tumor microenvironment is an important direction that is expected to be an important strategy for treating lymphatic metastasis of CC in the future.

Acknowledgements

Not applicable.

Funding

Funding: This work was supported by the National Natural Science Foundation of China (grant no. 81902140).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

LFW and JC conceived the concept of the review and revised the manuscript. LFW, SYY, YT, and WHL drafted the manuscript. LFW and WHL prepared the figures. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethical approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Singh D, Vignat J, Lorenzoni V, Eslahi M, Ginsburg O, Lauby-Secretan B, Arbyn M, Basu P, Bray F and Vaccarella S: Global estimates of incidence and mortality of cervical cancer in 2020: A baseline analysis of the WHO global cervical cancer elimination initiative. Lancet Glob Health. 11:e197–e206. 2023.PubMed/NCBI View Article : Google Scholar

2 

Tax C, Rovers MM, de Graaf C, Zusterzeel PL and Bekkers RL: The sentinel node procedure in early stage cervical cancer, taking the next step; a diagnostic review. Gynecol Oncol. 139:559–567. 2015.PubMed/NCBI View Article : Google Scholar

3 

Bhatla N, Aoki D, Sharma DN and Sankaranarayanan R: Cancer of the cervix uteri. Int J Gynaecol Obstet. 143 (Suppl 2):S22–S36. 2018.PubMed/NCBI View Article : Google Scholar

4 

Jürgenliemk-Schulz IM, Beriwal S, de Leeuw AAC, Lindegaard JC, Nomden CN, Pötter R, Tanderup K, Viswanathan AN and Erickson B: Management of nodal disease in advanced cervical cancer. Semin Radiat Oncol. 29:158–165. 2019.PubMed/NCBI View Article : Google Scholar

5 

Dadafarin S, Carnazza M, Islam HK, Moscatello A, Tiwari RK and Geliebter J: Noncoding RNAs in papillary thyroid cancer: Interaction with cancer-associated fibroblasts (CAFs) in the tumor microenvironment (TME) and regulators of differentiation and lymph node metastasis. Adv Exp Med Biol. 1350:145–155. 2021.PubMed/NCBI View Article : Google Scholar

6 

Solis-Castillo LA, Garcia-Romo GS, Diaz-Rodriguez A, Reyes-Hernandez D, Tellez-Rivera E, Rosales-Garcia VH, Mendez-Cruz AR, Jimenez-Flores JR, Villafana-Vazquez VH and Pedroza-Gonzalez A: Tumor-infiltrating regulatory T cells, CD8/Treg ratio, and cancer stem cells are correlated with lymph node metastasis in patients with early breast cancer. Breast Cancer. 27:837–849. 2020.PubMed/NCBI View Article : Google Scholar

7 

Griffith JW, Sokol CL and Luster AD: Chemokines and chemokine receptors: Positioning cells for host defense and immunity. Annu Rev Immunol. 32:659–702. 2014.PubMed/NCBI View Article : Google Scholar

8 

Singh S, Sadanandam A and Singh RK: Chemokines in tumor angiogenesis and metastasis. Cancer Metastasis Rev. 26:453–467. 2007.PubMed/NCBI View Article : Google Scholar

9 

He M, He Q, Cai X, Chen Z, Lao S, Deng H, Liu X, Zheng Y, Liu X, Liu J, et al: Role of lymphatic endothelial cells in the tumor microenvironment-a narrative review of recent advances. Transl Lung Cancer Res. 10:2252–2277. 2021.PubMed/NCBI View Article : Google Scholar

10 

Schito L: Hypoxia-dependent angiogenesis and lymphangiogenesis in cancer. Adv Exp Med Biol. 1136:71–85. 2019.PubMed/NCBI View Article : Google Scholar

11 

Ji RC: Hypoxia and lymphangiogenesis in tumor microenvironment and metastasis. Cancer Lett. 346:6–16. 2014.PubMed/NCBI View Article : Google Scholar

12 

Dieterich LC, Tacconi C, Ducoli L and Detmar M: Lymphatic vessels in cancer. Physiol Rev. 102:1837–1879. 2022.PubMed/NCBI View Article : Google Scholar

13 

Chen JM, Luo B, Ma R, Luo XX, Chen YS and Li Y: Lymphatic endothelial markers and tumor lymphangiogenesis assessment in human breast cancer. Diagnostics (Basel). 12(4)2021.PubMed/NCBI View Article : Google Scholar

14 

Lambert AW and Weinberg RA: Linking EMT programmes to normal and neoplastic epithelial stem cells. Nat Rev Cancer. 21:325–338. 2021.PubMed/NCBI View Article : Google Scholar

15 

Bakir B, Chiarella AM, Pitarresi JR and Rustgi AK: EMT, MET, plasticity, and tumor metastasis. Trends Cell Biol. 30:764–776. 2020.PubMed/NCBI View Article : Google Scholar

16 

Sinha D, Saha P, Samanta A and Bishayee A: Emerging concepts of hybrid epithelial-to-mesenchymal transition in cancer progression. Biomolecules. 10(1561)2020.PubMed/NCBI View Article : Google Scholar

17 

Kumagai Y, Tachikawa T, Higashi M, Sobajima J, Takahashi A, Amano K, Fukuchi M, Ishibashi K, Mochiki E, Yakabi K, et al: Vascular endothelial growth factors C and D and lymphangiogenesis at the early stage of esophageal squamous cell carcinoma progression. Dis Esophagus. 31:2018.PubMed/NCBI View Article : Google Scholar

18 

García-Silva S, Benito-Martín A, Nogués L, Hernández-Barranco A, Mazariegos MS, Santos V, Hergueta-Redondo M, Ximénez-Embún P, Kataru RP, Lopez AA, et al: Melanoma-derived small extracellular vesicles induce lymphangiogenesis and metastasis through an NGFR-dependent mechanism. Nat Cancer. 2:1387–1405. 2021.PubMed/NCBI View Article : Google Scholar

19 

Dadras SS, Lange-Asschenfeldt B, Velasco P, Nguyen L, Vora A, Muzikansky A, Jahnke K, Hauschild A, Hirakawa S, Mihm MC and Detmar M: Tumor lymphangiogenesis predicts melanoma metastasis to sentinel lymph nodes. Mod Pathol. 18:1232–1242. 2005.PubMed/NCBI View Article : Google Scholar

20 

Roy S, Kumaravel S, Banerjee P, White TK, O'Brien A, Seelig C, Chauhan R, Ekser B, Bayless KJ, Alpini G, et al: Tumor lymphatic interactions induce CXCR2-CXCL5 axis and alter cellular metabolism and lymphangiogenic pathways to promote cholangiocarcinoma. Cells. 10(3093)2021.PubMed/NCBI View Article : Google Scholar

21 

Gogineni A, Maresa C, Ailey C, Lee CR, Fuh G, van Bruggen N, Ye W and Weimer RM: Inhibition of VEGF-C modulates distal lymphatic remodeling and secondary metastasis. PLoS One. 8(e68755)2013.PubMed/NCBI View Article : Google Scholar

22 

Aebischer D, Iolyeva M and Halin C: The inflammatory response of lymphatic endothelium. Angiogenesis. 17:383–393. 2014.PubMed/NCBI View Article : Google Scholar

23 

Miteva DO, Rutkowski JM, Dixon JB, Kilarski W, Shields JD and Swartz MA: Transmural flow modulates cell and fluid transport functions of lymphatic endothelium. Circ Res. 106:920–931. 2010.PubMed/NCBI View Article : Google Scholar

24 

Wiley HE, Gonzalez EB, Maki W, Wu MT and Wang ST: Expression of CC chemokine receptor-7 and regional lymph node metastasis of B16 murine melanoma. J Nat Cancer Inst. 93:1638–1643. 2001.PubMed/NCBI View Article : Google Scholar

25 

Mezzapelle R, Leo M, Caprioglio F, Colley LS, Lamarca A, Sabatino L, Colantuoni V, Crippa MP and Bianchi ME: CXCR4/CXCL12 activities in the tumor microenvironment and implications for tumor immunotherapy. Cancers (Basel). 14(2314)2022.PubMed/NCBI View Article : Google Scholar

26 

Hirakawa S, Detmar M, Kerjaschki D, Nagamatsu S, Matsuo K, Tanemura A, Kamata N, Higashikawa K, Okazaki H, Kameda K, et al: Nodal lymphangiogenesis and metastasis: Role of tumor-induced lymphatic vessel activation in extramammary Paget's disease. Am J Pathol. 175:2235–2248. 2009.PubMed/NCBI View Article : Google Scholar

27 

Kawada K, Hosogi H, Sonoshita M, Sakashita H, Manabe T, Shimahara Y, Sakai Y, Takabayashi A, Oshima M and Taketo MM: Chemokine receptor CXCR3 promotes colon cancer metastasis to lymph nodes. Oncogene. 26:4679–4688. 2007.PubMed/NCBI View Article : Google Scholar

28 

Das S, Sarrou E, Podgrabinska S, Cassella M, Mungamuri SK, Feirt N, Gordon R, Nagi CS, Wang Y, Entenberg D, et al: Tumor cell entry into the lymph node is controlled by CCL1 chemokine expressed by lymph node lymphatic sinuses. J Exp Med. 210:1509–1528. 2013.PubMed/NCBI View Article : Google Scholar

29 

Fujimoto N and Dieterich LC: Mechanisms and clinical significance of tumor lymphatic invasion. Cells. 10(2585)2021.PubMed/NCBI View Article : Google Scholar

30 

Issa A, Le TX, Shoushtari AN, Shields JD and Swartz MA: Vascular endothelial growth factor-C and C-C chemokine receptor 7 in tumor cell-lymphatic cross-talk promote invasive phenotype. Cancer Res. 69:349–357. 2009.PubMed/NCBI View Article : Google Scholar

31 

Meier F, Will S, Ellwanger U, Schlagenhauff B, Schittek B, Rassner G and Garbe C: Metastatic pathways and time courses in the orderly progression of cutaneous melanoma. Br J Dermatol. 147:62–70. 2002.PubMed/NCBI View Article : Google Scholar

32 

Kim M, Koh YJ, Kim KE, Koh BI, Nam DH, Alitalo K, Kim I and Koh GY: CXCR4 signaling regulates metastasis of chemoresistant melanoma cells by a lymphatic metastatic niche. Cancer Res. 70:10411–10421. 2010.PubMed/NCBI View Article : Google Scholar

33 

Farnsworth RH, Karnezis T, Maciburko SJ, Mueller SN and Stacker SA: The interplay between lymphatic vessels and chemokines. Front Immunol. 10(518)2019.PubMed/NCBI View Article : Google Scholar

34 

Shields JD, Kourtis IC, Tomei AA, Roberts JM and Swartz MA: Induction of lymphoidlike stroma and immune escape by tumors that express the chemokine CCL21. Science. 328:749–752. 2010.PubMed/NCBI View Article : Google Scholar

35 

Lund AW, Duraes FV, Hirosue S, Raghavan VR, Nembrini C, Thomas S, Issa A, Hugues S and Swartz MA: VEGF-C promotes immune tolerance in B16 melanomas and cross-presentation of tumor antigen by lymph node lymphatics. Cell Rep. 1:191–199. 2012.PubMed/NCBI View Article : Google Scholar

36 

Tewalt EF, Cohen JN, Rouhani SJ, Guidi CJ, Qiao H, Fahl SP, Conaway MR, Bender TP, Tung KS, Vella AT, et al: Lymphatic endothelial cells induce tolerance via PD-L1 and lack of costimulation leading to high-level PD-1 expression on CD8 T cells. Blood. 120:4772–4782. 2012.PubMed/NCBI View Article : Google Scholar

37 

De Nola R, Loizzi V, Cicinelli E and Cormio G: Dynamic crosstalk within the tumor microenvironment of uterine cervical carcinoma: Baseline network, iatrogenic alterations, and translational implications. Crit Rev Oncol Hematol. 162(103343)2021.PubMed/NCBI View Article : Google Scholar

38 

Datta A, West C, O'Connor JPB, Choudhury A and Hoskin P: Impact of hypoxia on cervical cancer outcomes. Int J Gynecol Cancer. 31:1459–1470. 2021.PubMed/NCBI View Article : Google Scholar

39 

Rojo-León V, García C, Valencia C, Méndez MA, Wood C and Covarrubias L: The E6/E7 oncogenes of human papilloma virus and estradiol regulate hedgehog signaling activity in a murine model of cervical cancer. Exp Cell Res. 381:311–322. 2019.PubMed/NCBI View Article : Google Scholar

40 

De Nola R, Menga A, Castegna A, Loizzi V, Ranieri G, Cicinelli E and Cormio G: The crowded crosstalk between cancer cells and stromal microenvironment in gynecological malignancies: Biological pathways and therapeutic implication. Int J Mol Sci. 20(2401)2019.PubMed/NCBI View Article : Google Scholar

41 

Lea JS and Lin KY: Cervical cancer. Obstet Gynecol Clin North Am. 39:233–253. 2012.PubMed/NCBI View Article : Google Scholar

42 

Baluk P, Fuxe J, Hashizume H, Romano T, Lashnits E, Butz S, Vestweber D, Corada M, Molendini C, Dejana E and McDonald DM: Functionally specialized junctions between endothelial cells of lymphatic vessels. J Exp Med. 204:2349–2362. 2007.PubMed/NCBI View Article : Google Scholar

43 

Tacconi C, Correale C, Gandelli A, Spinelli A, Dejana E, D'Alessio S and Danese S: Vascular endothelial growth factor C disrupts the endothelial lymphatic barrier to promote colorectal cancer invasion. Gastroenterology. 148:1438–1451.e8. 2015.PubMed/NCBI View Article : Google Scholar

44 

Chen C, Shen N, Chen Y, Jiang P, Sun W, Wang Q, Wang Z, Wang Y, Cheng W, Fu S and Wang S: LncCCLM inhibits lymphatic metastasis of cervical cancer by promoting STAU1-mediated IGF-1 mRNA degradation. Cancer Lett. 518:169–179. 2021.PubMed/NCBI View Article : Google Scholar

45 

Alavi A, Carlin SD, Werner TJ and Zaghal AA: Suboptimal sensitivity and specificity of PET and other gross imaging techniques in assessing lymph node metastasis. Mol Imaging Biol. 21:808–811. 2019.PubMed/NCBI View Article : Google Scholar

46 

Phan TG and Croucher PI: The dormant cancer cell life cycle. Nat Rev Cancer. 20:398–411. 2020.PubMed/NCBI View Article : Google Scholar

47 

Ju S, Wang F, Wang Y and Ju S: CSN8 is a key regulator in hypoxia-induced epithelial-mesenchymal transition and dormancy of colorectal cancer cells. Mol Cancer. 19(168)2020.PubMed/NCBI View Article : Google Scholar

48 

Hsin MC, Hsieh YH, Hsiao YH, Chen PN, Wang PH and Yang SF: Carbonic anhydrase IX promotes human cervical cancer cell motility by regulating PFKFB4 expression. Cancers (Basel). 13(1174)2021.PubMed/NCBI View Article : Google Scholar

49 

Sugiura K, Nakajima S, Kato I, Okubo-Sato M, Nakazawa Y, Mitsudo K and Kioi M: Hypoxia and CD11b+ cell influx are strongly associated with lymph node metastasis of oral cancer. Anticancer Res. 40:6845–6852. 2020.PubMed/NCBI View Article : Google Scholar

50 

Cairns RA and Hill RP: Acute hypoxia enhances spontaneous lymph node metastasis in an orthotopic murine model of human cervical carcinoma. Cancer Res. 64:2054–2061. 2004.PubMed/NCBI View Article : Google Scholar

51 

Chaudary N, Milosevic M and Hill RP: Suppression of vascular endothelial growth factor receptor 3 (VEGFR3) and vascular endothelial growth factor C (VEGFC) inhibits hypoxia-induced lymph node metastases in cervix cancer. Gynecol Oncol. 123:393–400. 2011.PubMed/NCBI View Article : Google Scholar

52 

Lee S, Shin HJ, Han IO, Hong EK, Park SY, Roh JW, Shin KH, Kim TH and Kim JY: Tumor carbonic anhydrase 9 expression is associated with the presence of lymph node metastases in uterine cervical cancer. Cancer Sci. 98:329–333. 2007.PubMed/NCBI View Article : Google Scholar

53 

Li Z, Jiang L, Chew SH, Hirayama T, Sekido Y and Toyokuni S: Carbonic anhydrase 9 confers resistance to ferroptosis/apoptosis in malignant mesothelioma under hypoxia. Redox Biol. 26(101297)2019.PubMed/NCBI View Article : Google Scholar

54 

Hu HM, Mao MH, Hu YH, Zhou XC, Li S, Chen CF, Li CN, Yuan QL and Li W: Artemisinin protects DPSC from hypoxia and TNF-α mediated osteogenesis impairments through CA9 and Wnt signaling pathway. Life Sci. 277(119471)2021.PubMed/NCBI View Article : Google Scholar

55 

Kim JH, Kim JY, Yoon MS, Kim YS, Lee JH, Kim HJ, Kim H, Kim YJ, Yoo CW, Nam BH, et al: Prophylactic irradiation of para-aortic lymph nodes for patients with locally advanced cervical cancers with and without high CA9 expression (KROG 07-01): A randomized, open-label, multicenter, phase 2 trial. Radiother Oncol. 120:383–389. 2016.PubMed/NCBI View Article : Google Scholar

56 

Chen XJ, Deng YR, Wang ZC, Wei WF, Zhou CF, Zhang YM, Yan RM, Liang LJ, Zhong M, Liang L, et al: Hypoxia-induced ZEB1 promotes cervical cancer progression via CCL8-dependent tumour-associated macrophage recruitment. Cell Death Dis. 10(508)2019.PubMed/NCBI View Article : Google Scholar

57 

Chen XJ, Wei WF, Wang ZC, Wang N, Guo CH, Zhou CF, Liang LJ, Wu S, Liang L and Wang W: A novel lymphatic pattern promotes metastasis of cervical cancer in a hypoxic tumour-associated macrophage-dependent manner. Angiogenesis. 24:549–565. 2021.PubMed/NCBI View Article : Google Scholar

58 

Chen XJ, Wu S, Yan RM, Fan LS, Yu L, Zhang YM, Wei WF, Zhou CF, Wu XG, 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. 58:388–397. 2019.PubMed/NCBI View Article : Google Scholar

59 

Mao X, Xu J, Wang W, Liang C, Hua J, Liu J, Zhang B, Meng Q, Yu X and Shi S: Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: New findings and future perspectives. Mol Cance. 20(131)2021.PubMed/NCBI View Article : Google Scholar

60 

Huang M, Fu M, Wang J, Xia C, Zhang H, Xiong Y, He J, Liu J, Liu B, Pan S and Liu F: TGF-β1-activated cancer-associated fibroblasts promote breast cancer invasion, metastasis and epithelial-mesenchymal transition by autophagy or overexpression of FAP-α. Biochem Pharmacol. 188(114527)2021.PubMed/NCBI View Article : Google Scholar

61 

Wang Y, Jing Y, Ding L, Zhang X, Song Y, Chen S, Zhao X, Huang X, Pu Y, Wang Z, et al: Epiregulin reprograms cancer-associated fibroblasts and facilitates oral squamous cell carcinoma invasion via JAK2-STAT3 pathway. J Exp Clin Cancer Res. 38(274)2019.PubMed/NCBI View Article : Google Scholar

62 

Zhou B, Chen WL, Wang YY, Lin ZY, Zhang DM, Fan S and Li JS: A role for cancer-associated fibroblasts in inducing the epithelial-to-mesenchymal transition in human tongue squamous cell carcinoma. J Oral Pathol Med. 43:585–592. 2014.PubMed/NCBI View Article : Google Scholar

63 

Murata T, Mekada E and Hoffman RM: Reconstitution of a metastatic-resistant tumor microenvironment with cancer-associated fibroblasts enables metastasis. Cell Cycle. 16:533–535. 2017.PubMed/NCBI View Article : Google Scholar

64 

Murata T, Mizushima H, Chinen I, Moribe H, Yagi S, Hoffman RM, Kimura T, Yoshino K, Ueda Y, Enomoto T and Mekada E: HB-EGF and PDGF mediate reciprocal interactions of carcinoma cells with cancer-associated fibroblasts to support progression of uterine cervical cancers. Cancer Res. 71:6633–6642. 2011.PubMed/NCBI View Article : Google Scholar

65 

Xiao L, Zhu H, Shu J, Gong D, Zheng D and Gao J: Overexpression of TGF-β1 and SDF-1 in cervical cancer-associated fibroblasts promotes cell growth, invasion and migration. Arch Gynecol Obstet. 305:179–192. 2022.PubMed/NCBI View Article : Google Scholar

66 

Wei WF, Chen XJ, Liang LJ, Yu L, Wu XG, Zhou CF, Wang ZC, Fan LS, Hu Z, Liang L and Wang W: Periostin+ cancer-associated fibroblasts promote lymph node metastasis by impairing the lymphatic endothelial barriers in cervical squamous cell carcinoma. Mol Oncol. 15:210–227. 2021.PubMed/NCBI View Article : Google Scholar

67 

Nielsen SR and Schmid MC: Macrophages as key drivers of cancer progression and metastasis. Mediators Inflamm. 2017(9624760)2017.PubMed/NCBI View Article : Google Scholar

68 

Mazzieri R, Pucci F, Moi D, Zonari E, Ranghetti A, Berti A, Politi LS, Gentner B, Brown JL, Naldini L and De Palma M: Targeting the ANG2/TIE2 axis inhibits tumor growth and metastasis by impairing angiogenesis and disabling rebounds of proangiogenic myeloid cells. Cancer Cell. 19:512–526. 2011.PubMed/NCBI View Article : Google Scholar

69 

Yeo EJ, Cassetta L, Qian BZ, Lewkowich I, Li JF, Stefater JA III, Smith AN, Wiechmann LS, Wang Y, Pollard JW and Lang RA: Myeloid WNT7b mediates the angiogenic switch and metastasis in breast cancer. Cancer Res. 74:2962–2973. 2014.PubMed/NCBI View Article : Google Scholar

70 

Ji H, Cao R, Yang Y, Zhang Y, Iwamoto H, Lim S, Nakamura M, Andersson P, Wang J, Sun Y, et al: TNFR1 mediates TNF-α-induced tumour lymphangiogenesis and metastasis by modulating VEGF-C-VEGFR3 signalling. Nat Commun. 5(4944)2014.PubMed/NCBI View Article : Google Scholar

71 

Kimura S, Noguchi H, Nanbu U and Nakayama T: Macrophage CCL22 expression promotes lymphangiogenesis in patients with tongue squamous cell carcinoma via IL-4/STAT6 in the tumor microenvironment. Oncol Lett. 21(383)2021.PubMed/NCBI View Article : Google Scholar

72 

Hosono M, Koma YI, Takase N, Urakawa N, Higashino N, Suemune K, Kodaira H, Nishio M, Shigeoka M, Kakeji Y and Yokozaki H: CXCL8 derived from tumor-associated macrophages and esophageal squamous cell carcinomas contributes to tumor progression by promoting migration and invasion of cancer cells. Oncotarget. 8:106071–106088. 2017.PubMed/NCBI View Article : Google Scholar

73 

Guo F, Kong W, Zhao G, Cheng Z, Ai L, Lv J, Feng Y and Ma X: The correlation between tumor-associated macrophage infiltration and progression in cervical carcinoma. Biosci Rep. 41(BSR20203145)2021.PubMed/NCBI View Article : Google Scholar

74 

Tan J, Yang L, Zhao H, Ai Y, Ren L, Zhang F, Dong W, Shi R, Sun D and Feng Y: The role of NFATc1/c-myc/PKM2/IL-10 axis in activating cervical cancer tumor-associated M2 macrophage polarization to promote cervical cancer progression. Exp Cell Res. 413(113052)2022.PubMed/NCBI View Article : Google Scholar

75 

Jiang S, Yang Y, Fang M, Li X and Yuan XJ: Co-evolution of tumor-associated macrophages and tumor neo-vessels during cervical cancer invasion. Oncol Lett. 12:2625–2631. 2016.PubMed/NCBI View Article : Google Scholar

76 

Li Y, Huang G and Zhang S: Associations between intratumoral and peritumoral M2 macrophage counts and cervical squamous cell carcinoma invasion patterns. Int J Gynaecol Obstet. 139:346–351. 2017.PubMed/NCBI View Article : Google Scholar

77 

Dou A and Fang J: Heterogeneous myeloid cells in tumors. Cancers (Basel). 13(3772)2021.PubMed/NCBI View Article : Google Scholar

78 

Mabuchi S, Matsumoto Y, Kawano M, Minami K, Seo Y, Sasano T, Takahashi R, Kuroda H, Hisamatsu T, Kakigano A, et al: Uterine cervical cancer displaying tumor-related leukocytosis: A distinct clinical entity with radioresistant feature. J Natl Cancer Inst. 106(dju147)2014.PubMed/NCBI View Article : Google Scholar

79 

Marvel D and Gabrilovich DI: Myeloid-derived suppressor cells in the tumor microenvironment: Expect the unexpected. J Clin Invest. 125:3356–3364. 2015.PubMed/NCBI View Article : Google Scholar

80 

Mabuchi S, Komura N, Sasano T, Shimura K, Yokoi E, Kozasa K, Kuroda H, Takahashi R, Kawano M, Matsumoto Y, et al: Pretreatment tumor-related leukocytosis misleads positron emission tomography-computed tomography during lymph node staging in gynecological malignancies. Nat Commun. 11(1364)2020.PubMed/NCBI View Article : Google Scholar

81 

Lee BR, Kwon BE, Hong EH, Shim A, Song JH, Kim HM, Chang SY, Kim YJ, Kweon MN, Youn JI and Ko HJ: Interleukin-10 attenuates tumour growth by inhibiting interleukin-6/signal transducer and activator of transcription 3 signalling in myeloid-derived suppressor cells. Cancer Lett. 381:156–164. 2016.PubMed/NCBI View Article : Google Scholar

82 

Kim KH, Sim NS, Chang JS and Kim YB: Tumor immune microenvironment in cancer patients with leukocytosis. Cancer Immunol Immunother. 69:1265–1277. 2020.PubMed/NCBI View Article : Google Scholar

83 

Panni RZ, Sanford DE, Belt BA, Mitchem JB, Worley LA, Goetz BD, Mukherjee P, Wang-Gillam A, Link DC, Denardo DG, et al: Tumor-induced STAT3 activation in monocytic myeloid-derived suppressor cells enhances stemness and mesenchymal properties in human pancreatic cancer. Cancer Immunol Immunother. 63:513–528. 2014.PubMed/NCBI View Article : Google Scholar

84 

Peng D, Tanikawa T, Li W, Zhao L, Vatan L, Szeliga W, Wan S, Wei S, Wang Y, Liu Y, et al: Myeloid-derived suppressor cells endow stem-like qualities to breast cancer cells through IL6/STAT3 and NO/NOTCH cross-talk signaling. Cancer Res. 76:3156–3165. 2016.PubMed/NCBI View Article : Google Scholar

85 

Kuroda H, Mabuchi S, Yokoi E, Komura N, Kozasa K, Matsumoto Y, Kawano M, Takahashi R, Sasano T, Shimura K, et al: Prostaglandin E2 produced by myeloid-derived suppressive cells induces cancer stem cells in uterine cervical cancer. Oncotarget. 9:36317–36330. 2018.PubMed/NCBI View Article : Google Scholar

86 

Ni HH, Zhang L, Huang H, Dai SQ and Li J: Connecting METTL3 and intratumoural CD33+ MDSCs in predicting clinical outcome in cervical cancer. J Transl Med. 18(393)2020.PubMed/NCBI View Article : Google Scholar

87 

Heeren AM, Koster BD, Samuels S, Ferns DM, Chondronasiou D, Kenter GG, Jordanova ES and de Gruijl TD: High and interrelated rates of PD-L1+CD14+ antigen-presenting cells and regulatory T cells mark the microenvironment of metastatic lymph nodes from patients with cervical cancer. Cancer Immunol Res. 3:48–58. 2015.PubMed/NCBI View Article : Google Scholar

88 

Rodríguez PC and Ochoa AC: Arginine regulation by myeloid derived suppressor cells and tolerance in cancer: Mechanisms and therapeutic perspectives. Immunol Rev. 222:180–191. 2008.PubMed/NCBI View Article : Google Scholar

89 

Galliverti G, Wullschleger S, Tichet M, Murugan D, Zangger N, Horton W, Korman AJ, Coussens LM, Swartz MA and Hanahan D: Myeloid cells orchestrate systemic immunosuppression, impairing the efficacy of immunotherapy against HPV+ cancers. Cancer Immunol Res. 8:131–145. 2020.PubMed/NCBI View Article : Google Scholar

90 

Jianyi D, Haili G, Bo Y, Meiqin Y, Baoyou H, Haoran H, Fang L, Qingliang Z and Lingfei H: Myeloid-derived suppressor cells cross-talk with B10 cells by BAFF/BAFF-R pathway to promote immunosuppression in cervical cancer. Cancer Immunol Immunother. 72:87–89. 2023.PubMed/NCBI View Article : Google Scholar

91 

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. 5(18217)2015.PubMed/NCBI View Article : Google Scholar

92 

Sawant DV, Yano H, Chikina M, Zhang Q, Liao M, Liu C, Callahan DJ, Sun Z, Sun T, Tabib T, et al: Adaptive plasticity of IL-10+ and IL-35+ Treg cells cooperatively promotes tumor T cell exhaustion. Nat Immunol. 20:724–735. 2019.PubMed/NCBI View Article : Google Scholar

93 

Wu MY, Kuo TY and Ho HN: Tumor-infiltrating lymphocytes contain a higher proportion of FOXP3(+) T lymphocytes in cervical cancer. J Formos Med Assoc. 110:580–586. 2011.PubMed/NCBI View Article : Google Scholar

94 

Nakamura T, Shima T, Saeki A, Hidaka T, Nakashima A, Takikawa O and Saito S: Expression of indoleamine 2, 3-dioxygenase and the recruitment of Foxp3-expressing regulatory T cells in the development and progression of uterine cervical cancer. Cancer Sci. 98:874–881. 2007.PubMed/NCBI View Article : Google Scholar

95 

Heeren AM, de Boer E, Bleeker MC, Musters RJ, Buist MR, Kenter GG, de Gruijl TD and Jordanova ES: Nodal metastasis in cervical cancer occurs in clearly delineated fields of immune suppression in the pelvic lymph catchment area. Oncotarget. 6:32484–32493. 2015.PubMed/NCBI View Article : Google Scholar

96 

Wang S, Li J, Xie J, Liu F, Duan Y, Wu Y, Huang S, He X, Wang Z and Wu X: Programmed death ligand 1 promotes lymph node metastasis and glucose metabolism in cervical cancer by activating integrin β4/SNAI1/SIRT3 signaling pathway. Oncogene. 37:4164–4180. 2018.PubMed/NCBI View Article : Google Scholar

97 

Stein M and Eckert KA: Impact of G-quadruplexes and chronic inflammation on genome instability: Additive effects during carcinogenesis. Genes (Basel). 12(1779)2021.PubMed/NCBI View Article : Google Scholar

98 

Zhang LX, Wei ZJ, Xu M and Zang JH: Can the neutrophil-lymphocyte ratio and platelet-lymphocyte ratio be beneficial in predicting lymph node metastasis and promising prognostic markers of gastric cancer patients? Tumor maker retrospective study. Int J Surg. 56:320–327. 2018.PubMed/NCBI View Article : Google Scholar

99 

Ayhan S, Akar S, Kar İ, Turan AT, Türkmen O, Kiliç F, Aytekin O, Ersak B, Ceylan Ö, Moraloğlu Tekin Ö and Kimyon Comert G: Prognostic value of systemic inflammatory response markers in cervical cancer. J Obstet Gynaecol. 42(2411)2022.PubMed/NCBI View Article : Google Scholar

100 

Lee WH, Kim GE and Kim YB: Prognostic factors of dose-response relationship for nodal control in metastatic lymph nodes of cervical cancer patients undergoing definitive radiotherapy with concurrent chemotherapy. J Gynecol Oncol. 33(e59)2022.PubMed/NCBI View Article : Google Scholar

101 

Polgár C, Major T and Varga S: Radiotherapy and radio-chemotherapy of cervical cancer. Magy Onkol. 66:307–314. 2022.PubMed/NCBI(In Hungarian).

102 

Vinay DS, Ryan EP, Pawelec G, Talib WH, Stagg J, Elkord E, Lichtor T, Decker WK, Whelan RL, Kumara HMCS, et al: Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin Cancer Biol. 35 (Suppl):S185–S198. 2015.PubMed/NCBI View Article : Google Scholar

103 

van Weelden WJ, Sekarutami SM, Bekkers RL, Kaanders JH, Bussink J, Gondhowiardjo S, Leer JW and Span PN: The effect of carbogen breathing and nicotinamide added to standard (chemo) radiation treatment of advanced cervical cancer in Indonesia. Int J Gynecol Cancer. 24:1628–1635. 2014.PubMed/NCBI View Article : Google Scholar

104 

Samsuri NAB, Leech M and Marignol L: Metformin and improved treatment outcomes in radiation therapy-A review. Cancer Treat Rev. 55:150–162. 2017.PubMed/NCBI View Article : Google Scholar

105 

Lin A and Maity A: Molecular pathways: A novel approach to targeting hypoxia and improving radiotherapy efficacy via reduction in oxygen demand. Clin Cancer Res. 21:1995–2000. 2015.PubMed/NCBI View Article : Google Scholar

106 

Sharma A, Arambula JF, Koo S, Kumar R, Singh H, Sessler JL and Kim JS: Hypoxia-targeted drug delivery. Chem Soc Rev. 48:771–813. 2019.PubMed/NCBI View Article : Google Scholar

107 

Zeman EM, Brown JM, Lemmon MJ, Hirst VK and Lee WW: SR-4233: A new bioreductive agent with high selective toxicity for hypoxic mammalian cells. Int J Radiat Oncol Biol Phys. 12:1239–1242. 1986.PubMed/NCBI View Article : Google Scholar

108 

Brown JM: SR 4233 (tirapazamine): A new anticancer drug exploiting hypoxia in solid tumours. Br J Cancer. 67:1163–1170. 1993.PubMed/NCBI View Article : Google Scholar

109 

DiSilvestro PA, Ali S, Craighead PS, Lucci JA, Lee YC, Cohn DE, Spirtos NM, Tewari KS, Muller C, Gajewski WH, et al: Phase III randomized trial of weekly cisplatin and irradiation versus cisplatin and tirapazamine and irradiation in stages IB2, IIA, IIB, IIIB, and IVA cervical carcinoma limited to the pelvis: A gynecologic oncology group study. J Clin Oncol. 32:458–464. 2014.PubMed/NCBI View Article : Google Scholar

110 

Seidel JA, Otsuka A and Kabashima K: Anti-PD-1 and anti-CTLA-4 therapies in cancer: Mechanisms of action, efficacy, and limitations. Front Oncol. 8(86)2018.PubMed/NCBI View Article : Google Scholar

111 

Chen DS and Mellman I: Oncology meets immunology: The cancer-immunity cycle. Immunity. 39:1–10. 2013.PubMed/NCBI View Article : Google Scholar

112 

Yao S, Zhu Y and Chen L: Advances in targeting cell surface signalling molecules for immune modulation. Nat Rev Drug Discov. 12:130–146. 2013.PubMed/NCBI View Article : Google Scholar

113 

Naumann RW, Hollebecque A, Meyer T, Devlin MJ, Oaknin A, Kerger J, López-Picazo JM, Machiels JP, Delord JP, Evans TRJ, et al: Safety and efficacy of Nivolumab Monotherapy in recurrent or metastatic cervical, vaginal, or vulvar carcinoma: Results from the phase I/II CheckMate 358 trial. J Clin Oncol. 37:2825–2834. 2019.PubMed/NCBI View Article : Google Scholar

114 

O'Malley DM, Neffa M, Monk BJ, Melkadze T, Huang M, Kryzhanivska A, Bulat I, Meniawy TM, Bagameri A, Wang EW, et al: Dual PD-1 and CTLA-4 checkpoint blockade using Balstilimab and Zalifrelimab combination as second-line treatment for advanced cervical cancer: An open-label phase II study. J Clin Oncol. 40:762–771. 2022.PubMed/NCBI View Article : Google Scholar

115 

Santin AD, Deng W, Frumovitz M, Buza N, Bellone S, Huh W, Khleif S, Lankes HA, Ratner ES, O'Cearbhaill RE, et al: Phase II evaluation of nivolumab in the treatment of persistent or recurrent cervical cancer (NCT02257528/NRG-GY002). Gynecol Oncol. 157:161–166. 2020.PubMed/NCBI View Article : Google Scholar

116 

Frenel JS, Le Tourneau C, O'Neil B, Ott PA, Piha-Paul SA, Gomez-Roca C, van Brummelen EMJ, Rugo HS, Thomas S, Saraf S, et al: Safety and efficacy of Pembrolizumab in advanced, programmed death ligand 1-positive cervical cancer: Results from the phase Ib KEYNOTE-028 trial. J Clin Oncol. 35:4035–4041. 2017.PubMed/NCBI View Article : Google Scholar

117 

Chung HC, Ros W, Delord JP, Perets R, Italiano A, Shapira-Frommer R, Manzuk L, Piha-Paul SA, Xu L, Zeigenfuss S, et al: Efficacy and safety of pembrolizumab in previously treated advanced cervical cancer: results from the phase II KEYNOTE-158 study. J Clin Oncol. 37:1470–1478. 2019.PubMed/NCBI View Article : Google Scholar

118 

De Jaeghere EA, Tuyaerts S, Van Nuffel AMT, Belmans A, Bogaerts K, Baiden-Amissah R, Lippens L, Vuylsteke P, Henry S, Trinh XB, et al: Pembrolizumab, radiotherapy, and an immunomodulatory five-drug cocktail in pretreated patients with persistent, recurrent, or metastatic cervical or endometrial carcinoma: Results of the phase II PRIMMO study. Cancer Immunol Immunother. 72:475–491. 2023.PubMed/NCBI View Article : Google Scholar

119 

Lee Y, Auh SL, Wang Y, Burnette B, Wang Y, Meng Y, Beckett M, Sharma R, Chin R, Tu T, et al: Therapeutic effects of ablative radiation on local tumor require CD8+ T cells: Changing strategies for cancer treatment. Blood. 114:589–595. 2009.PubMed/NCBI View Article : Google Scholar

120 

Sharabi AB, Lim M, DeWeese TL and Drake CG: Radiation and checkpoint blockade immunotherapy: Radiosensitisation and potential mechanisms of synergy. Lancet Oncol. 16:e498–e509. 2015.PubMed/NCBI View Article : Google Scholar

121 

Twyman-Saint Victor C, Rech AJ, Maity A, Rengan R, Pauken KE, Stelekati E, Benci JL, Xu B, Dada H, Odorizzi PM, et al: Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature. 520:373–377. 2015.PubMed/NCBI View Article : Google Scholar

122 

Young KH, Baird JR, Savage T, Cottam B, Friedman D, Bambina S, Messenheimer DJ, Fox B, Newell P, Bahjat KS, et al: Optimizing timing of immunotherapy improves control of tumors by hypofractionated radiation therapy. PLoS One. 11(e0157164)2016.PubMed/NCBI View Article : Google Scholar

123 

Dovedi SJ, Adlard AL, Lipowska-Bhalla G, McKenna C, Jones S, Cheadle EJ, Stratford IJ, Poon E, Morrow M, Stewart R, et al: Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade. Cancer Res. 74:5458–5468. 2014.PubMed/NCBI View Article : Google Scholar

124 

Sun J and Yuan J: Chemokine (C-X-C motif) ligand 1/chemokine (C-X-C motif) receptor 2 autocrine loop contributes to cellular proliferation, migration and apoptosis in cervical cancer. Bioengineered. 13:7579–7591. 2022.PubMed/NCBI View Article : Google Scholar

125 

Strachan DC, Ruffell B, Oei Y, Bissell MJ, Coussens LM, Pryer N and Daniel D: CSF1R inhibition delays cervical and mammary tumor growth in murine models by attenuating the turnover of tumor-associated macrophages and enhancing infiltration by CD8+ T cells. Oncoimmunology. 2(e26968)2013.PubMed/NCBI View Article : Google Scholar

126 

Steele CW, Karim SA, Leach JDG, Bailey P, Upstill-Goddard R, Rishi L, Foth M, Bryson S, McDaid K, Wilson Z, et al: CXCR2 inhibition profoundly suppresses metastases and augments immunotherapy in pancreatic ductal adenocarcinoma. Cancer Cell. 29:832–845. 2016.PubMed/NCBI View Article : Google Scholar

127 

Pagni RL, Souza PDC, Pegoraro R, Porchia BFMM, da Silva JR, Aps LRMM, Silva MO, Rodrigues KB, Sales NS, Ferreira LCS and Moreno ACR: Interleukin-6 and indoleamine-2,3-dioxygenase as potential adjuvant targets for papillomavirus-related tumors immunotherapy. Front Immunol. 13(1005937)2022.PubMed/NCBI View Article : Google Scholar

128 

He Y, Kozaki K, Karpanen T, Koshikawa K, Yla-Herttuala S, Takahashi T and Alitalo K: Suppression of tumor lymphangiogenesis and lymph node metastasis by blocking vascular endothelial growth factor receptor 3 signaling. J Natl Cancer Inst. 94:819–825. 2002.PubMed/NCBI View Article : Google Scholar

129 

García-Quiroz J, Vázquez-Almazán B, García-Becerra R, Díaz L and Avila E: The interaction of human papillomavirus infection and prostaglandin E2 signaling in carcinogenesis: A focus on cervical cancer therapeutics. Cells. 11(2528)2022.PubMed/NCBI View Article : Google Scholar

130 

Peng H, He X and Wang Q: Immune checkpoint blockades in gynecological cancers: A review of clinical trials. Acta Obstet Gynecol Scand. 101:941–951. 2022.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

October-2023
Volume 26 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang L, Yi S, Teng Y, Li W and Cai J: Role of the tumor microenvironment in the lymphatic metastasis of cervical cancer (Review). Exp Ther Med 26: 486, 2023
APA
Wang, L., Yi, S., Teng, Y., Li, W., & Cai, J. (2023). Role of the tumor microenvironment in the lymphatic metastasis of cervical cancer (Review). Experimental and Therapeutic Medicine, 26, 486. https://doi.org/10.3892/etm.2023.12185
MLA
Wang, L., Yi, S., Teng, Y., Li, W., Cai, J."Role of the tumor microenvironment in the lymphatic metastasis of cervical cancer (Review)". Experimental and Therapeutic Medicine 26.4 (2023): 486.
Chicago
Wang, L., Yi, S., Teng, Y., Li, W., Cai, J."Role of the tumor microenvironment in the lymphatic metastasis of cervical cancer (Review)". Experimental and Therapeutic Medicine 26, no. 4 (2023): 486. https://doi.org/10.3892/etm.2023.12185