Next Article in Journal
Comparative Characterization of Iron and Silver Nanoparticles: Extract-Stabilized and Classical Synthesis Methods
Previous Article in Journal
Pyroptosis: A Promising Mechanism Linking SARS-CoV-2 Infection to Adverse Pregnancy Outcomes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Crosstalk of Immune Cells and Platelets in an Ovarian Cancer Microenvironment and Their Prognostic Significance

by
Katarzyna Aneta Pankowska
*,
Grażyna Ewa Będkowska
,
Joanna Chociej-Stypułkowska
,
Małgorzata Rusak
,
Milena Dąbrowska
and
Joanna Osada
Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(11), 9279; https://doi.org/10.3390/ijms24119279
Submission received: 28 April 2023 / Revised: 17 May 2023 / Accepted: 23 May 2023 / Published: 25 May 2023
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)

Abstract

:
Ovarian cancer (OC) is one of the deadliest gynecological cancers, largely due to the fast development of metastasis and drug resistance. The immune system is a critical component of the OC tumor microenvironment (TME) and immune cells such as T cells, NK cells, and dendritic cells (DC) play a key role in anti-tumor immunity. However, OC tumor cells are well known for evading immune surveillance by modulating the immune response through various mechanisms. Recruiting immune-suppressive cells such as regulatory T cells (Treg cells), macrophages, or myeloid-derived suppressor cells (MDSC) inhibit the anti-tumor immune response and promote the development and progression of OC. Platelets are also involved in immune evasion by interaction with tumor cells or through the secretion of a variety of growth factors and cytokines to promote tumor growth and angiogenesis. In this review, we discuss the role and contribution of immune cells and platelets in TME. Furthermore, we discuss their potential prognostic significance to help in the early detection of OC and to predict disease outcome.

1. Introduction

Ovarian cancer (OC) is one of the biggest problems in gynecological oncology, causing over 207,000 deaths worldwide in 2020. Ovarian malignant neoplasm ranks eighth in the world among cancers in women [1,2]. It is distinguished by a poor prognosis, remarkably high death rate, and a five-year survival rate that is continuously under 45%. If a cancer is detected at an early stage (International Federation of Obstetrics and Gynecology (FIGO I)) before it spreads outside of the ovaries, the majority of women can be effectively treated using standard therapeutic approaches, with a survival rate of up to 90%. When OC spreads to the pelvic organs (FIGO II), the abdominal organs (FIGO III), and beyond the peritoneal cavity (FIGO IV), the situation dramatically changes. The poor survival rate for OC is associated with detection at an advanced stage due to the absence of distinct signs and symptoms in the initial stages of the disease and a lack of proper screening. In the majority of patients, nonspecific gastrointestinal symptoms such as flatulence, a feeling of fullness, and reproductive organ problems such as excessive vaginal bleeding or feeling pressure in the pelvis may occur more than a year before diagnosis [3,4]. This leads to approximately 70% of women being diagnosed in the advanced stages of the disease (FIGO III and IV) [5].
More than 90% of ovarian malignant neoplasms in developed nations are of an epithelial origin, with the remaining 10% coming from stromal cells (5–6%) or germ cells (2–3%) [6]. High- and low-grade serous ovarian cancers account for 70% of all epithelial-derived ovarian tumors; the remaining 30% are endometrial, mucinous, and clear cell carcinomas. Here, two lines of ovarian carcinogenesis development with the degree of histological and molecular differentiation were identified and taken into consideration. Serous (low-grade serous ovarian carcinoma (LGSOC)), clear cell, and endometrial cancers are examples of well-differentiated tumors that fall under type I and are characterized by slow development and minimal spreading potential. Serous (high-grade serous ovarian carcinomas (HGSOC)), endometrial, and sarcomas are examples of type II low-differentiated carcinomas that exhibit rapid growth and a strong metastatic potential. Over 95% of cases of HGSOC are correlated with genetic mutations in the tumor suppressor protein p53, which account for the late stage (FIGO III/IV) diagnosis of the disease when the cancer has advanced to the peritoneum [7]. Additionally, almost 85% of hereditary OC cases and more than one-fifth of all occurrences of OC are linked to mutation in genes such as BRCA1 and BRCA2 [8,9].
Combination therapy, which consists of surgery and platinum-based chemotherapy, is the recommended approach for treating OC. However, the effectiveness of this treatment varies depending on the histological nature of the tumor. Recent clinical trials suggest that drugs targeting signaling pathways, such as the PI3K- and RAS-signaling pathways, may be a new and promising option for treating OC [10]. Despite this, recurrence is seen in the majority of women within 18 months [10,11].
To gain a better understanding of OC development and progression, it is vital to recognize the functioning of the ovarian tumor microenvironment, particularly the activity and characteristics of immune cells. Various soluble factors (cytokines, chemokines, and proteins) and direct cell-to-cell interactions comprise the active network required for the development of local immunosuppression, allowing cancer cells to survive, grow, and develop metastatic qualities.
Platelets, which are derived from megakaryocytes, are anucleate cell fragments that play a significant role in primary hemostasis via their multilayer interaction between platelet and cancer cells in the tumor microenvironment, bloodstream, and peritoneal fluid. Increasing evidence in recent years suggests their crucial involvement in cancer metastatic spread, notably in the promotion of the invasiveness, angiogenesis, and anti-apoptotic activity in OC. Moreover, increasing evidence suggests that platelets may play important roles in cancer [12]. In OC, thrombocytosis, both at the time of initial diagnosis and at recurrence, has been associated with worse disease outcomes [13].
The purpose of this review is to summarize and better understand the involvement of immune cells and platelets in the development and progression of OC, as well show as their prognostic significance.

2. Tumor Development

The presence of leukocytes within tumors, discovered by Rudolf Virchow in the 19th century, offered the first indication of a possible relationship between inflammation and cancer. A role for inflammation in carcinogenesis is now well accepted, and an inflammatory microenvironment is an essential component of many cancers [14].
Several studies have linked ovarian surface inflammation-related factors to the development of OC [15,16]. There are two strong carcinogenesis models: ovulation theory and chronic inflammation, both of which propose that incessant ovulation or chronic exposure to external or endogenous triggers a cascade of immune cells to injury, causing damage to the ovarian endothelium, DNA damage through reactive oxygen species (ROS) release, and cytokine production, all of which increase the risk of malignant transformation. Chronic inflammation can promote and support a variety of cellular events that lead to the production or selection of aggressive cancer cells [17].
As previously stated, genomic instability caused by DNA damage could be one of the mechanisms of cancer initiation. A pro-inflammatory microenvironment is constantly replenished by a wide range of cytokines, growth factors, and ROS, all of which can damage DNA, switch to an antiapoptotic pathway, and trigger cancer transformation. During transformation, cells activate pro-survival signaling pathways rather than pro-apoptotic pathways that oncogenes normally activate to initiate the transformation of normal cells. The presence of an abundance of inflammatory mediators promotes a pro-inflammatory microenvironment, which can trigger the reprogramming of the surrounding cells to establish a tumor microenvironment (TME) (Figure 1) [18]. Besides malignant-transformed cells, TME is composed of normal cells including epithelial cells, fibroblasts, muscle cells, and inflammatory immune cells. Several innate and adaptive immune cells, such as macrophages, neutrophils, myeloid-derived suppressor cells (MDSCs), and T regulatory cells (Treg cells), shape the peritoneal TME directly or indirectly (via soluble interactions), creating a permissive environment for tumor development. The formation of a complex immune suppression network that effectively neutralizes anticancer activity is one of the primary causes of disease progression and therapy failure. In this part of the review, we closely focus on selected players of pro- and anti-cancer immunity in TME.

2.1. Tumor-Associated Macrophages

Macrophages are an essential population of immune cells that participate in inflammation and tumorigenesis. Among them, macrophages that either reside or are recruited to the tumor tissue are termed tumor-associated macrophages (TAMs) [19].
TAMs can be derived from resident macrophages or infiltrating macrophages from bone marrow monocytes circulating in the blood. TAMs can exhibit either the anti-tumor M1 macrophage or the pro-tumor M2 macrophage phenotype depending on the stimuli in the TME [20,21]. TAMs are the most prevalent type of immune cells in OC TME by volume and number alone, and numerous studies have connected tumor growth and survival to the characteristic properties of those cells [22].
Monocytes are differentiated into M1 macrophages in response to stimulation with interferon-gamma (IFN-γ), lipopolysaccharide (LPS), and granulocyte-macrophage-colony-stimulating factor (GM-CSF), which can secrete IL-1, IL-12, tumor necrosis factor (TNFα), and stromal cell-derived factor 1 (SDF-1) [23,24]. M1 macrophages possess cytotoxicity, tumor suppression, and immune-stimulation functions.
Monocytes developed into M2 macrophages when activated by cytokines such as IL-4 and IL-13. In OC, TAMs are predominantly M2 macrophages that produce and secrete anti-inflammatory/immunosuppressive cytokines such as IL-10 and transforming growth factor beta 1(TGFB1) and chemokines (CCL17, CCL18, and CCL22) associated with tumor invasion, angiogenesis, metastasis, and early recurrence [25]. Moreover, monocytes develop into M2 macrophages more quickly in an immunosuppressive environment [26,27].
TAM heterogeneity cannot be explained by a simple dichotomy of M1/M2 macrophages. Transcriptome analysis revealed a TAM spectrum model. M1 and M2 macrophages can be thought of as two endpoints of a spectrum with a wide range of functional states; TAM subpopulations in between the two ends can share characteristics of both M1 and M2 types [28,29]. Singhal et al. discovered that TAMs can co-express M1/M2 markers, together with T cell co-inhibitory and co-stimulatory receptors [30]. The dynamic nature of the TME cellular environment provides a foundation for TAM plasticity. Macrophages undergo reversible changes in their functional phenotypes and distribution in response to numerous microenvironmental cues, including tissue and tumor-specific cytokines and locally generated molecules. As a result, TAMs with varying degrees of infiltration and functional states can be found in distinct tumor histotypes and microregions of the same tumor. According to the genome-wide expression analysis of TAMs in HGSOC patients, not only are traditional M2 markers such as IL-10 and CD163 upregulated, but so are several M1 markers such as CD86 and TNF-α [31].
TAM polarization is strongly influenced by soluble and insoluble components found in the OC TME, which includes both the peritoneal and primary sites. TME is thought to stimulate TAMs to exhibit an immunosuppressive and pro-tumoral phenotype, encouraging cancer growth and progression. However, the key players in TAMs education, are the cancer cells themselves [32,33].
The TAMs pro-tumoral phenotype is also initiated by colony-stimulated factor 1 (CSF-1). CSF-1 is a key cytokine that regulates macrophage differentiation, development, and function, in addition to transforming macrophages into an M2-polarized phenotype [34]. Furthermore, cancer cells are the primary generator of CSF-1 in TME [31,32,35,36]. Colony stimulating factor 1 receptor (CSF-1R) inhibition significantly increases the proportion of M1 macrophages expressing CCR2, IL-12, and IFN-γ, decreasing the immunosuppressive state of TME [37]. Additionally, allowing monocytes to consume CSF-1 in the presence of high levels of leukemia inhibitory factor (LIF) and IL-6 in OC ascites can boost the generation of TAMs [38]. It was discovered that OC cell mucins trigger a TAMs immunosuppressive profile, as seen by the strong IL-10 production [39]. When co-cultured with OC cells, macrophages unregulated the expression of M2-associated macrophage mannose receptor (MR; CD206) [33]. MR was reported to be expressed in TAMs isolated from OC patients. TAMs are directed toward an immune-suppressive phenotype when activated by tumor mucins such as cancer antigen 125 (CA125) and tumor-associated glycoprotein 72 (TAG-72) [39]. Mucin2 (MUC2) is overexpressed in OC cells and is an independent poor prognostic factor in OC patients. MUC2 expression on tumor cells is inversely associated with TAM M1/M2 ratio and promotes cancer growth [40].
M2-like TAMs participate in OC development through their significant immunosuppressive impact on immune cells in TME. Firstly, M2 macrophages secrete transforming growth factor (TGF-α), IL-6, IL-10, CCL18, and CCL22, which attract Treg cells and promote T cell differentiation towards the Th2 phenotype [41]. Secondly, IL-10 and TGF-β inhibit the cytotoxic activity of NK cells and cytotoxic T lymphocytes (CTL). In addition, IL-10 blocks the maturation of DC. CCL18 promotes T cell anergy and unresponsiveness [41].
Another mechanism through which M2-like TAMs regulate T cell activity is their influence on the checkpoint programmed cell death protein 1 (PD-1). The interaction of PD-1 with its ligand PD-L1 on macrophages inhibits T cell activation by depleting l-arginine, which is essential for proper T cell function. Arginase I (Arg1) is a l-arginine processing enzyme that is found in M2 macrophages. Arg1 decomposes l-arginine in the TME to form l-ornithine and urea. L-arginine deficiency inhibits the re-expression of the CD3ζ chain, which is internalized by antigen stimulation and receptor (TCR) signaling. T cells become unresponsive, manifested in the inhibition of their proliferation, cytotoxicity, the production of cytokines, finally resulting in the blocking of tumor-specific T cell responses. The PI3K/AKT and Ras-MEK-ERK pathways are the main signaling pathways inhibited via triggered PD-1 [42].
Additionally, more than 70% of TAMs of OC patients are characterized by the expression of B7-H4, a coinhibitory molecule induced by IL-6 and IL-10. B7-H4 is a transmembrane protein belonging to the B7 family of costimulatory proteins. B7-H4 binds the putative receptor on activated CD4+ and CD8+ T cells, causing a decrease in their proliferation and reduction in IL-2 production [43]. Selectively blocking B7-H4 expressed on the surface of ovarian TAMs significantly increased T-cell proliferation [43]. Moreover, the intensity of B7-H4 on TAMs was associated with the number of Treg cells [40].
Exosomes produced by M2-like TAMs in the ovarian TME inhibit immunological effector activities. TAM-derived exosomes, in particular, contain a high concentration of proteins as well as DNA, mRNA, and miRNA molecules, which together suppress T cell activity and promote an imbalance between Treg cells and T helper 17 cells (Th17) cells by directly targeting signal transducer and activator of transcription 3 (STAT3) in CD4+ T cells [44].

2.2. Dendric Cell

Dendritic cells (DCs) operate as a bridge between the innate and adaptive immune systems by capturing endogenous or foreign antigens, processing them, and presenting the antigenic peptides to other immune cells [9,45,46,47]. Based on functional and phenotypic features, DC can be divided into two main subsets: the conventional DC (cDC) that is specialized in antigen presentation, and the plasmacytoid DC (pDC) that produces IFNγ upon antigen stimulation, aside from activating lymphocytes and other myeloid cells plasma cells [48,49].
When exposed to an antigen, DC goes through a maturation process that is marked by an increase in costimulatory molecules, downregulation of pre-existing chemokine receptors, and the acquisition of CCR7. The latter drives DC to lymph nodes through the secreted chemokines CCL19 (MIP-3β) and CCL21. Mature DC can bind with the CD40 ligand on other cells to secrete IL-12, in addition to activating naïve CD8+ T cells [45,50,51,52,53].
In addition to T cell activation, DCs are essential to the expansion of the population of CTL within TME. Intratumoral cDCs are the only type of phagocytosing tumor myeloid cell that can promote CD8+ T cell proliferation and are responsible for intratumoral CTL proliferation both in vivo and in vitro [54,55].
Unfortunately, cDC are rarely present in ovarian TME and show signs of immaturity, especially in the early stages of the disease, suggesting that they might contribute to the development of tumors [56]. Immune-suppressive milieu of OC are abundant with cytokines and inhibitory molecules, which promote the differentiation of immature DCs cells, enhance tolerance, and accelerate tumor growth [35,57,58].
Immune checkpoint signaling could play a role in DC dysfunction. OC cells may enhance PD-L1 expression in DCs by secreting (TGF-β) and prostaglandin E2 (PGE2) into TME [59]. To impair the antitumor effect, specific DCs interact with immunosuppressive cells. Inducible co-stimulator (ICOS) is expressed on immunosuppressive Treg cells and pDCs in the OC TME activate Treg cells by expressing the ICOS ligand, resulting in tumor progression [60].
In OC, insulin-like growth factor (IGF) also affects DCs. Via the RAS-ERK and PI3K-AKT pathways, IGF contributes to protein synthesis, cell growth, and cell proliferation [61]. IGF-treated DCs are unable to mature and secrete higher levels of TNF-α and IL-10, both of which are immune suppressors in the microenvironment of OC [62]. The differentiation of DCs into cDCs is negatively correlated with the high expression of the insulin-like growth factor type I receptor (IGF1R) in OC [63].
Scarlett et al. discovered that DC depletion at advanced stages significantly delays aggressive malignant progression. [59]. It is worth noting that mature myeloid DCs, due to their antitumor immune responses, have been used in clinical trials of DC vaccine therapy in OC [36,64].
Because DC functions can be influenced by their interactions with the proximal milieu, different DC locations may result in different functions [48]. Labidi-Galy et al. discovered that tumor pDCs released fewer pro-inflammatory cytokines than pDCs from ascites or peripheral blood in OC patients [48].
Furthermore, the DC performance can change depending on the stage of tumor development. In an OC mouse model, infiltrating DCs prevented tumor growth at an early stage. However, at an advanced stage, DCs become immunosuppressive in the TME, inhibiting anti-tumor T cell activity and DC depletion, significantly delaying the disease progression [59]. Similarly, Krempski et al. discovered that infiltrating DCs acquired a more immunosuppressive phenotype as the tumor progressed over time in a mouse model of OC, as shown by the increased PD-1 expression [65].

2.3. Myeloid-Derived Suppressor Cells

A diverse group of myeloid cells known as myeloid-derived suppressor cells (MDSCs) co-express the myeloid surface markers GR-1 and CD11b [66,67]. Three phenotypes of MDSCs are present: polymorphonuclear PMN-MDSC (also known as granulocytic MDSC), which are morphologically similar to neutrophils; M-MDSC, which morphologically and phenotypically resemble monocytes; and a limited subset of cells that are capable of producing myeloid colonies, such as myeloid progenitors and precursors [68].
The mechanism that induces MDSCs into the TME of OC is poorly understood. Many studies have proposed various pathways of MDSC production among tumor cell lines, emphasizing the relevance of each tumor type’s unique combination of inflammatory cytokines.
MDSCs are recruited to ascites of OC in a CXCR4-dependent way that needs cyclooxygenase-2 (COX2), the major enzyme in PGE2 synthesis. PGE2 is essential for both the production of CXCL12 and the expression of CXCR4 in MDSCs [69]. Furthermore, PGE2 and COX2 redirect a potential pathway for the recruitment and development of CD1a+ DCs to CD14 + CD33 + CD34+ monocytic MDSCs and induce the expression of MDSC-associated immunosuppressive factors [69,70].
Another potential mechanism for the induction of MDSCs in the TME of OC involves IL-6. In OC, IL-6 has a significant role in the development of tumor growth, angiogenesis, and tumor myeloid cell infiltration. IL-6 may act as a mediator in the differentiation of MDSCs. According to an experimental investigation by Wouters et al., IL-6 and its receptor IL-6R are opposing markers for the survival and infiltration of tumor-infiltrating myeloid cells. Longer disease-specific survival and lower mature myeloid cell infiltration were observed in tumors with a high IL-6R expression. On the other hand, tumors with a high epithelial IL-6 expression showed a significant tumor-infiltrating myeloid infiltration and were associated with a reduced survival time [71].
One of the primary ways MDSCs can restrict T cell differentiation is by producing Arg-1 [42]. Additionally, MDSCs release ROS and nitric oxide (NO), which nitrate signaling molecules downstream of the Fc gamma receptor IIIa (FcRIIIa) and decrease the activity of T cells and NK cells, respectively [68,72]. The direct inhibition of T cells occurs when superoxide and NO combine to form peroxynitrite (PNT), which nitrates T-cell receptors and limits the response of antigen−MHC complexes. PNT also inhibits chemokines that are unique to T cells, which reduces the combination of antigenic peptides to MHC and hinders T cell migration [73].
Furthermore, MDSCs induce the activation and proliferation of regulatory Treg cells [74]. It has also been proposed that MDSCs support both the conversion of naïve CD4+ T cells to translate into induce Treg cells (iTreg) and the expansion of natural Treg cells (nTreg) [75,76].
The expression of ADAM Metallopeptidase Domain 17 (ADAM17) on MDSC reduced the expression of CD62 ligand (CD62L) on CD4+ CD8+ T cells, limiting recirculation into the lymph nodes [77].
Last, but not least, multiple checkpoint molecules have been implicated in MDSC-mediated immunosuppression, with PD-L1 and cytotoxic T cell antigen 4 (CTLA-4) being key negative regulators of T cell activities [78,79,80]. CTLA-4 is expressed by Treg cells and primarily interacts with CD80/CD86 expressed by antigen-presenting cell (APC)-like DC. This interaction reduces APC-dependent T cell activation [81].

2.4. Regulatory T Cells (Treg)

Regulatory T cells are a diverse group of CD4+ T lymphocytes that express CD25+ and the transcription factor forkhead box P3 (FOXP3) [82]. Treg cells are critical for maintaining tolerance and preventing autoimmunity. However, developing cancers including OC, recruit Treg cells to establish local immunosuppression through a variety of mechanisms [82]. Tregs cells identified as CD4 + CD25+ FOXP3+ are abundant in OC patients’ peripheral blood, TME and ascites [83,84].
Numerous pathways contribute to the induction and differentiation of Tregs in the OC TME. The most important axis in the selective trafficking of Tregs cells to tumors is CCR4/CCL22 and CCL17 signaling. TAMs are the main source of CCL22, which has been shown to be substantially expressed in OC and ascites cells compared with normal ovaries at the mRNA level [83]. According to a study performed on 104 patients with OC, the accumulation of Tregs cells in tumors induced by CCL22 is associated with decreased survival rates and a higher risk of patient death. The migration of T cells was significantly reduced by CCL22 inhibition [83]. Treg cell infiltration is also reduced by blocking CCR5/CCL5, which were also involved in the trafficking of Tregs cells [85].
A significant Th17 response in the early stages of tumor development has been demonstrated to be replaced by a preponderance of Treg cells in the late stages, demonstrating that tumor progression can shape Treg participation in the local immune milieu [86]. Idoleamine 2,3-dioxygenase (IDO) accumulation in tumors causes the tryptophan catabolite kynurenine to be activated. Kynurenine then binds the aryl hydrocarbon receptor (AhR) on T cells, shifting the Th17/Tregs cell balance in favor of Treg cell generation. Moreover, kynurenine can bind AhR on TAMs, triggering the production of IDO in a feedback loop. Furthermore, tumor-associated DCs infiltrating in the OC TME influence Treg cell transformation. Dysfunctional DCs infiltrating the TME directly contribute to the induction of IL-10-producing Tregs [87]. Notably, hypoxia-induced CCL28 overexpression enhances Treg cell migration to the OC TME via a mechanism involving CCR10, which also leads to IL-10 secretion [88].
Cell-to-cell contact is achieved through the production of molecules such as CTLA-4 and the secretion of soluble mediators such as IL-10 and TGF-β, which limit immune activation [89]. The co-expression of CD39, CD73, and glycoprotein-A repetition predominate protein (GARP) on Treg cells may also result in enhanced suppression [90,91,92]. Interestingly, in both animal tumor models and human head and neck cancer, CD39 and CD73 have been linked to reduced local anti-tumor immunity [93,94,95]. When CD39 and CD73 are co-expressed, they can convert ATP to adenosine, inhibiting the metabolic activity of effector T cells, whereas GARP is implicated in relaying TGF-β inhibitory signals and increasing FOXP3 production [92,96].
TNF receptor 2 (TNFR2) expression on Treg cells has been characterized as a maximally suppressive Treg cell population [97,98,99]. TNF-induced induction of TNFR2 expression on Tregs occurs in an inflammatory environment [16]. TNF levels in OC patient serum and ascites are elevated, and existing research suggests the establishment of a “TNF network” that supports tumor progression [100]. Research has shown that TNFR2 + Tregs cells from ascites were the most potent suppressor T cell population [101]. They were abundantly present within the ascites and more suppressive than in the peripheral blood of OC patients [101].

2.5. Tumor Infiltrating Lymphocytes (TIL)

Tumor infiltrating lymphocytes (TILs) are lymphocytes that left the vasculature and have localized in the tumor stroma or intraepithelial. TILs are segregated into those that penetrate the tumor islet (intraepithelial) and those that reside in the peritumoral space (stromal). TILs are dominated by CD3+ cells, while the proportions of CD4+ and CD8+ cells vary between cancers. TILs have been proven to be a powerful factor that imposes tremendous pressure on many types of cancer, including OC [102].
TIL local infiltration denotes local immunity of immune cells surrounding or within the tumor [103]. The abundance of TILs in OC shows that this tumor has immunogenic potential. Moreover, numerous tumor-associated antigens (TAAs) can be identified by T cells, highlighting the potential benefits of immunotherapy in OC treatment approaches [89,104]. DCs are immune system sentinels that detect TAAs expressed on the surface of tumor cells and transport them to lymph nodes, where they activate T cell-mediated immunity, allowing T cells to bypass the immune system working station and attack the tumor [105]. These physiological processes occur in all OC patients with improved survival because normally generated lymphocytes are insufficient to attack and eliminate the tumor in advance [106,107].

2.6. NK Cells

NK cells are a subset of the innate lymphoid cells that play a significant role in safeguarding the organism from viral infection, early malignant transformation, and metastatic tumor dissemination. Based on CD16 and CD56 expression, they can be divided into two main populations: CD56bright/CD16− predominantly function to produce cytokines in the bloodstream and CD56dim/CD16+, which causes cytotoxicity in the tissues. NK cells develop from the common lymphoid progenitor in the bone marrow, are most closely related to T cells, and have similar capacities to release perforin and granzymes for direct target cell killing. Furthermore, they signal target cell death via the Fas and TNF-related apoptosis-inducing ligand (TRAIL) pathways and release pro-inflammatory cytokines such as IFN-γ and TNF [108].
The CD16 receptor, natural killer group 2D (NKG2D) receptor, and natural cytotoxicity receptors, such as NKp30, are the most significant cytotoxicity receptors that mediate NK cell-dependent immunosurveillance. Malignant diseases have been associated with defects in NK cell function, including impaired cytotoxicity and cytokine secretion, aberrant receptor and ligand expression, decreased NK cell number, and NK cell anergy [109]. Two primary mechanisms that block the function of NKG2D and NKp30 have been described in OC [110,111,112].
Ascites fluid from OC patients contains cancer cells that emit macrophage migration inhibitory factor (MIF), a chemokine that promotes the growth, migration, and metastasis of tumor cells. NKG2D is transcriptionally downregulated by MIF in NK cells, which reduces their capacity to eradicate tumor cells [110]. Second, elevated levels of soluble B7-H6, one of the ligands for the NKp30 receptor, suppress NK cell function in ascites. Increased soluble B7-H6 expression was linked to decreased NKp30 expression in tumor-associated NK cells and decreased NK cell activity [112]. Immune escape may be facilitated by reduced NK cell activities in the OC TME, and additional research into the underlying mechanism is necessary.
NK cells in the presence of IL-18 release the chemokines CCL3 and CCL4 that recruit immature DCs. Cross-talk between NKs and DCs increases the expression of CXCL9, CXCL10, and CCL5 on DCs, which can recruit effector CD8+ T cells to the TME [113]. NK cells are powerful inducers of the cDC chemoattractants XCL1 and CCL5. Tumor PGE2 synthesis could interfere with this mechanism and the ability of DC to secrete chemokines [114].

3. Tumor Progression

The transcoelomic route is the most common route of OC cell metastasis, and it is predicated on cancer cells detaching from the primary tumor and moving through the peritoneal fluid to the omentum, parietal, and visceral peritoneum, as well as direct extension of the tumor lesions to the adjacent organs. The additional route is via lymphatic vessels to pelvic and paraaortic lymph nodes. According to studies, about 70% of women with OC had peritoneal cavity metastases at the time of diagnosis [115,116]. OC tumor cells primarily colonize the omentum and form metastatic lesions, although micro-metastases frequently appear on the peritoneal surfaces. Microvasculature perfusion is inadequate in metastatic omentum and peritoneal tissues, according to immunohistochemical studies. Furthermore, the normal collagen network is disrupted in the metastatic omentum and peritoneum [117].
Presently, two hypotheses for the peritoneal metastasis model in OC have been proposed. The first hypothesis, which is related to the “seed and soil” concept, proposes that peritoneal metastasis of OC is induced by circulating tumor cells within the peritoneal cavity, which preferentially metastasize to the peritoneum through transcoelomic, hematogenous, or lymphatic routes [118]. The second hypothesis, known as the metaplasia hypothesis, argues that the OC metastatic omental sites are the consequence of a synchronized malignant transformation of the peritoneum or omentum due to the ovarian epithelium and omentum’s similar origin [115].
Although the etiology of ascites is unknown, preclinical and clinical studies have shown that the vascular endothelial growth factor (VEGF) is responsible for ascites accumulation, and that the obstruction of lymphatic vessels by cancer cells may also cause the accumulation of ascites. Ascitic fluid in most cases of OC is composed of malignant cells, a large number of immune cells, and a high level of lactate dehydrogenase (LDH) [119,120].
It should be emphasized that the conventional patterns of metastasis via the hematogenous pathway occur in OC, yet they are not the dominant way, and they are primarily responsible for distant metastases [13]. In clinical trials, inferior vena cava filters enhance the risk of hematogenous dissemination in OC by activating platelets and causing a proinflammatory response [121]. Moreover, circulating tumor cells detached from original lesions could be identified in OC patients upon early diagnosis [122].
The underlying mechanisms creating the strong OC tropism and complex interplay between tumor cells, immune cells, and platelets within the TME are unknown.

3.1. Tumor-Associated Macrophages

TAMs play a critical role in the transcoelomic dissemination of OC cells, their survival in ascitic fluid, and the formation of spheroids in malignant ascites. Furthermore, many spheroids in OC patients are heterogeneous, containing TAM-OC cells. TAMs have been found to promote spheroid formation and tumor growth in an ID8-bearing mouse model by secreting endothelial growth factor (EGF) [123].
To promote the interaction between TAMs and tumor tissues, EGF secreted by TAMs in the core of the spheroid upregulated the αMβ2 integrin on TAMs and intercellular cell adhesion molecule-1 (ICAM-1) on tumor cells. EGF influences ICAM-1 expression by inducing VEGF-C production, which promotes VEGF receptor 2 (VEGFR3) signaling in tumor cells and induces integrin/ICAM-1, which ultimately leads to tumor growth and migration [120,124]. In an animal model, inhibiting VEGF/VEGFR signaling or neutralizing ICAM-1 slowed spheroid and tumor growth [38].
Hypoxia is another essential component in the accumulation of TAMs. TAMs have been found in high-density hypoxic areas of tumors in many studies. According to Wen et al., increased 5-lipoxygenase (5-LOX) metabolites from OC cells under hypoxic conditions stimulate TAM migration and invasion by upregulating matrix metalloproteinase-7 (MMP-7) via the p38 pathway. The tumor-bearing mouse model demonstrates that inhibiting 5-LOX selectively reduces MMP-7 expression and the number of TAMs in tumor tissues [125].
Monocytes and macrophages obtained from OC patients had an increased number, a less differentiated phenotype, deficient cytotoxicity and phagocytic abilities, and impaired antitumor activities when compared with cells isolated from healthy donors [126]. An animal investigation showed that chemically depleting macrophages significantly reduced tumor dissemination and ascites build-up in OC-bearing mice [127].
In OC cells, the expression of MIF and extracellular matrix metalloproteinase inducer (EMMPRIN) upregulated MMP secretion by macrophages and overexpressed MMP promotes OC invasion and angiogenesis [128]. MIF inhibition in ovarian tumor-bearing mice decreases macrophage infiltration, as well as IL-6, IL-10, and TNF-α [129]. Furthermore, downregulated MIF in OC cells decreased the production of CCL2 and CCL22 in vitro [129]. CCL22, which is produced and increased by ovarian TAMs, can stimulate Treg cell trafficking to the tumor [83]. CCL18, an immunosuppressive chemokine expressed by TAMs, has been found in high levels in patients with OC. CCL18 levels in ascites and the serum of OC patients are elevated, which increases tumor migration and metastasis [130,131]. IFN-γ therapy can decrease CCL18 secretion and shift TAMs from being immunosuppressive to immunostimulatory [38].
Scavenger receptor-A (SR-A, CD204) expressed on TAMs and dependent on the presence of TNF-α was found to be involved in tumor cell invasion [33]. In vitro, SR-A-deficient macrophages diminish the invasiveness of OC cells. Tumor progression and metastasis are decreased in SR-A (-/-) mice when compared with the control group [132].
The crucial mechanism through which TAMs promote OC cell invasiveness is through the activation of nuclear factor κB (NFκB) activity. Numerous data from in vitro investigations on different cell lines clearly show that TNF-α is a crucial player in NFκB overexpression and activation in cancer cells. Blocking the NFκB signaling pathways in ovarian TAMs reduces M2 cytokines while increasing M1 IL-12 and NOS, thus inhibiting tumor cell invasion [133]. The effect of TNF-α on a tumor is determined by the dose and time. Prolonged low-dose exposure increases tumor progression, whereas a single high-dose exposure causes tumor regression. After interacting with its receptors, TNF-α increases the activity of OC cells, changes their shape, and promotes carcinogenesis and angiogenesis [133]. TNF-α expression is closely associated with the accumulation of IL-6 and CXCL12, which are implicated in an autocrine cytokine network, which was previously confirmed in OC [100]. In both a mouse model and human OC specimens, the “TNF network” played a paracrine function in angiogenesis, myeloid cell infiltration, and NOTCH signaling [100].
An elevated level of IL-6 and TGF-β in ascites and the serum of OC patients was associated with the generation of TAMs. STAT3 is a crucial point of junction for several carcinogenic pathways and is activated by TAMs IL-6, which is necessary for OC cell migration, motility, survival, and proliferation [134].
CXCR4 is found to be constitutively expressed in OC, while CXCL12 has been found to be highly concentrated in OC ascites. The CXCR4−CXCL12 pathway promotes invasion, the recruitment of immunosuppressive cells, and angiogenesis. Blocking the CXCR4−CXCL12 pathway improves tumor-bearing mice survival by decreasing the number of Treg cells and increasing the CD8/Treg ratio [135]. A study by Kajiyama et al. found that high levels of CXCL12 upregulated by TGF-β1 were accumulated in OC patient’s human peritoneal mesothelial cells (HPMCs), which served as scaffolding for the first step of peritoneal metastasis, enhancing crosstalk between tumor cells and HPMCs and promoting peritoneal metastasis [136].
TAMs may also be implicated in angiogenesis. The functional interaction of macrophages and endothelial cells is a hot topic in cancer and vascular biology. Co-cultivating OC cell lines with TAMs greatly boosts endothelial cell migration and tube formation, as well as the concentration of the pro-angiogenic cytokine IL-8 [137]. TAMs have also been linked to lymphangiogenesis in ovarian cancer. TAMs were reported to enhance lymphangiogenesis by stimulating lymphatic endothelial cell proliferation, migration, and capillary-like tube formation in a study of 108 ovarian tissue specimens. Its property may be enhanced by combining it with high-mobility group box protein 1 (HMGB1) [138].
To summarize, TAMs play a significant role in the course of OC disease. TAMs are a potent immunosuppressive driver within the TME. TAMs also promote metastasis and angiogenesis via several signaling pathways, which is especially important in OC, which is frequently diagnosed after the tumor has already metastasized.

3.2. Myeloid Derived Suppressor Cells

MDSCs not only suppress immune responses within the TME, but also promote cancer progression by stimulating tumor angiogenesis and enhancing tumor cell invasion and metastasis. MDSC under hypoxia-derived mediators, such as VEGF, basic fibroblast growth factor (FGF), and MMP-9, all play a crucial role in tumor angiogenesis and cancer cell invasion, regulating these processes [139]. Furthermore, new research suggests that MDSCs are involved in the establishment of epithelial-to-mesenchymal transition (EMT) or by forming “premetastatic niches” [140]. Crucially, data suggest that MDSCs promote “stemness”, which may be linked to resistance to conventional anticancer treatments such as chemotherapy or radiotherapy [141].
Additionally, by promoting miRNA101 expression and subsequently suppressing the co-repressor gene C-terminal binding protein-2 (CtBP2), MDSCs improve the stemness of OC cells. By specifically focusing on the stem-cell core genes, CtBP2 improves tumorigenic and metastatic potential while increasing the stemness of cancer cells [142].

3.3. Tumor Infiltrating Lymphocytes (TIL)

The absence of TILs in patients is linked to increased angiogenesis in OC, which creates a significant barrier to the infiltration of TILs. Overexpressed VEGF promotes endothelial cell proliferation, migration, and invasion in OC, which is associated with increased microvascular density [143]. Through the deregulation of ICAM-1 and vascular cell adhesion protein 1 (VCAM-1), VEGF-A can also lessen the adhesion between lymphocytes and tumor vascular endothelial cells, which in turn reduces TIL penetration [144]. Moreover, VEGF-A can promote the expression of FasL in endothelial cells in conjunction with IL-10 and PGE2, and increased FasL expression is linked to enabling the selective trafficking of more Tregs than CD8+ T cells [145]. Additionally, it has been demonstrated that VEGF inhibits CD8+ T cells by recruiting in MDSCs, and that the MDSCs produced by VEGF have stronger immunosuppressive properties. Anti-VEGF therapies successfully reduced the tumor development and ascites generation in mice models of OC [146].

3.4. Neutrophils

Neutrophils are a remarkably diverse population with both pro- and anti-tumor abilities, which is mostly due to phenotypic plasticity. Contrary to the notion that mature neutrophils exit the bone marrow as terminally differentiated cells, there are distinct subsets of neutrophils that differ in terms of their level of maturity and activation, as well as their pro- and anti-cancer properties. Neutrophil subsets with opposing roles have so far been identified in cancer patients’ circulation and primary tumors. The significance of neutrophils in cancer is debatable as they have seemingly contradicting qualities that might promote or inhibit tumor growth [147]. TAN (tumor-associated-neutrophils) can have anti-tumorigenic (N1) or pro-tumorigenic (N2) functions [148]. According to studies, TAN displays different activity patterns depending on the microenvironment. TGF-β induces neutrophils to acquire an N2 pro-tumoral phenotype, whereas IFN-β induces neutrophils to acquire an N1 phenotype, which is more anti-tumoral [148,149].
TGF-β inhibition enhanced the proinflammatory potential of TANs (N1), resulting in an increased expression of proinflammatory cytokines such as TNF-α and CCL3, cytotoxic CD8+ T lymphocyte activation, and direct killing of tumor cells dependent on ROS and the costimulatory molecule ICAM-1. [148]. N2 neutrophils have higher levels of CXCR4, VEGF, MMP-9, and arginase, which promote angiogenesis, immunological suppression, and tumorigenesis [150]. Moreover, according to in vivo research, subpopulations of TANs in early lung cancer could be polarized to an anti-tumor phenotype in response to low doses of GM-CSF and IFN-γ [151].
Neutrophils derived from bone marrow are generally found in low abundance in the omentum and peritoneal fluid, and act as the first line of defense in response to infections or tissue damage [152,153]. It has been reported that neutrophils mobilize into the abdominal cavity in response to peritoneal infection or injury via specialized vessels known as high endothelial venules (HEVs) in omental milky spots [154]. Lee et al. hypothesized that the establishment of the premetastatic omental niche in OC involves neutrophil influx at this site. It was discovered that recruiting neutrophils to the omentum is an important step preceding OC cell invasion of this site [155]. Moreover, Mishalian et al. proposed that neutrophil behavior varies depending on the stage of tumor development. They discovered that neutrophils were almost exclusively found at the tumor’s periphery during the initial stages of tumor development, and TANs were more cytotoxic toward tumor cells and generated higher levels of TNF-α, NO, and hydrogen peroxide (H2O2). At advanced stages of disease, neutrophils were found scattered among the tumor cells, and TAN properties, such as suppressing adaptive antitumor immunity, were down-regulated and acquired a more N2-pro-tumorigenic phenotype [156].
Neutrophils recruited into the TME are a source of cytokine and chemokine secretion that impacts innate and adaptive immunity. TANs release cytokines and chemokines, which have the ability to regulate both the activation and recruitment of other immune cells, as well as their own recruitment [157].
Neutrophils regulate immune responses using a variety of mechanisms, including the release of chemokines/cytokines, production of variety of antimicrobial molecules, neutrophil extracellular traps (NETs), recruitment of Treg cells, and PD-L1/PD-1 interactions [158,159,160]. Although some research implies that neutrophils have an antitumorogenic role due to T cell stimulatory activities, the majority of studies thus far indicate that neutrophils mostly have immunosuppressive functions.
In the case of OC, neutrophils work in tandem with TNF-/IL-17 signaling to promote tumor development and enhance premetastatic niche creation via NETs [161]. Furthermore, neutrophils developed inhibitory behavior in response to ascites supernatants from advanced OC patients. Additionally, Emmons et al. demonstrated the ability of TANs (N2) to adhere to T cells and cause trogocytosis of T-cell membranes, resulting in T-cell immunoparalysis characterized by an impaired nuclear factor of activated T cell (NFAT) translocation [162].

3.5. Platelets

Activated platelets play a variety of roles in tumor metastasis progression, including facilitating tumor-cell EMT. They become less adherent, less polar, more mesenchymal, and have increased mobility. EMT is characterized by a decreased expression of E-cadherin following the increased expression of p38 and related pathways. It has been shown that E-cadherin is down-regulated by epidermal growth factor receptor (EGFR) activated by OC cells, and that cells with a low E-cadherin expression are particularly invasive [163].
The role of platelets in EMT has been connected to many factors derived from either platelets or tumors to induce platelet activation. Podoplanin (PDPN) is a glycoprotein that binds to C-type lectin-like receptor 2 (CLEC-2) on platelets, resulting in platelet activation. PDPN-positive cancer cell lines stimulate platelet activation, particularly platelet TGFβ-expression, which induces cancer cells to undergo EMT. TGF-β inhibition, on the other hand, strongly reduces PDPN-induced EMT and metastasis in mice injected with tumor cells, implying that podoplanin causes tumor metastasis by boosting platelet-derived TGF-β [164].
Among the platelet associated metastasis mediators, TGF-β plays an essential role. It has been reported that in OC, there is a significant correlation between elevated platelets counts with a higher incidence of intraperitoneal dissemination and higher TGF-β [165]. Furthermore, platelets promote EMT and the invasiveness of OC cells by activating the TGF-β/Smad pathway. All of these impacts were reversed through the use of antiplatelet agent A83-01 (a selective inhibitor of TβRI), which inhibits the activation of the Smad pathway and, as a result, blocked the pro-metastatic effects and reversed the EMT in tumor-bearing OC cells in mice [165].
Additionally, the contribution of platelets to metastasis is influenced by autotaxin (ATX), which regulates lysophosphatidic acid levels (LPA) [166]. LPA acts as a bridge between cancer cells and platelets in the process of tumor invasion and metastasis. LPA is prevalent in the malignant ascites of OC patients [167,168]. In contrast with normal ovarian epithelial cells, which do not produce LPA at levels sufficient to stimulate aberrant receptors, the constitutive production of LPA by both OC cells and peritoneal mesothelial cells accelerates the spread of cancer cell and the synthesis of IL-6 and IL-8 [169,170]. CD97, a member of the epidermal growth factor family that is activated in various types of cancer, activates the signaling pathway via the LPA receptor [171].
There is mounting evidence to support the notion that platelets shape the metastatic milieu in the setting of the early metastatic niche. This was demonstrated in a lung cancer mouse model, where tumor-aggregated platelets promoted the formation of metastatic sites by producing cytokines that attract granulocytes CXCL-5 and CXCL-7 [172]. Platelet-derived chemoattractants, such as C-X-C motif ligands, particularly trigger the deployment of granulocytes to early metastatic niches rather than monocytes, lymphocytes, or NK cells [172]. Orellana et al. revealed that platelets have a chemotactic effect on OC cells, with the subsequent phenotypic change favoring a mesenchymal phenotype, with an enhanced expression of the tissue factor (TF) [173]. Platelets thus not only give survival signals for tumor cells, but also recruit host cells to disseminate tumor foci [173].
In order for cancer cells to escape detachment-induced apoptosis (often known as anoikis), platelet−cancer-cell interaction is required. Experimental findings implicate a crucial role for platelets in inducing anoikis resistance and in the metastatic spread of cancer cells intraperitoneally and hematogenously by inducing a Yes-Associated-Protein 1(YAP1)-dependent transcriptional program in detached cancer cells, which promotes cell survival and metastasis [174]. Furthermore, the results suggest that reducing blood platelet counts or interfering with YAP1 signaling might be an important approach to limit OC metastasis [174]. Additionally, recent research by Rodriguez-Martinez et al. has strengthened the notion that platelet interaction may cause tumor cells to develop aggressive phenotypes such as EMT, stem-like phenotypes, and high rates of proliferation [175]. The findings also showed that platelets educate tumor cells through the very effective transfer of lipids, proteins, and RNA via various mechanisms [175].
Cancer cells are exposed to significant challenges such as shear forces during circulation before reaching the distant site [176]. Platelets support cancer spread by shielding cancer cells from shear pressures induced by blood flow. The co-incubation of A2780 OC cells with human platelets significantly reduces the cancer cells’ release of LDH, a marker for shear-induced membrane damage [176]. Platelets can protect OC cells against shear-induced damage. The disruption of platelet−cancer-cell cross talk could increase the shear- stress-induced destruction of cancer cells in vivo [176].
Immune attack, in which NK cells play a crucial role in the immune system, present another significant threat to metastasizing cells in circulation [177]. Through the formation of aggregates on the tumor cell surface, platelets protect tumor cells against NK cell lysis [177]. Both sterically and by preventing NK cells from performing their cytolytic function, platelets prevent NK cells from adhering to tumor cells. Platelet activation leads to the expression of the TNF receptor superfamily member glucocorticoid-induced TNF-related ligand (GITRL) in parallel with the α-granular activation marker P-selectin on their surface membrane [178]. GITRL binds to its receptor on the NK cell membrane and suppresses the latter’s cytotoxic properties by impairing its lytic activity and IFN-γ secretion [178]. Activated platelets also contain a plethora of soluble substances that suppress NK cells. TGF-β released during tumor-cell-induced platelet aggregation (TCIPA) has been shown to downregulate theNKG2D immunoreceptor, limiting lytic activity and IFN-γ secretion [179]. It has also been proposed that the transfer of platelet-derived MHC-I onto the surface of tumor cells during aggregation may restrict the immune system’s NK-mediated attack against the developing metastatic niche [179]. Apart from NK cells, platelets interact with other immune cells such as macrophages and T-cells, and their impact on them may contribute to the formation of an immunosuppressive milieu. The micrometastatic niche, for example, is rich in platelet and tumor-cell-derived TGF-β, which suppresses both CD4+ and CD8+ T-cell activity.
Furthermore, platelets have been linked to neutrophil activation and the formation of neutrophil NETs, a process known as NETosis. The relationship between platelets and neutrophils is bidirectional as platelet TLR4 promotes NETosis and extracellular DNA from NETs induces platelet activation [180]. It also contributes to cancer-associated thrombosis, which OC patients are at a higher risk of developing [181,182,183].
According to the research, either by the direct paracrine effect on megakaryocytes or through their ability to generate and activate platelets, cancer cells can affect platelet counts and physiological activation states. OC cells can directly activate platelets through contact or indirectly through the release of agonist-like substances that contribute to the stimulation of megakaryopoiesis and, consequently, thrombopoiesis in cancer patients [184,185,186]. Platelets are activated via key pathways such as thromboxane (TX)-A2, glycoprotein (GP)-Ib-IX, adenosine diphosphate (ADP), and GPIIb/IIIa [187].
Additionally, through participation in the coagulation pathway, cancer cells might indirectly activate platelets. TF, which may be generated and secreted by macrophages and endothelial cells as well as OC themselves, is released in great quantities by cancer cells and is a key factor in the procoagulant ability of tumors [188,189,190]. Furthermore, cancer cells have the capacity to release TF-rich procoagulant microparticles (MPs) that cause the production of thrombin [191]. Thrombin secretion is one of TCIPA’s most crucial processes [192,193]. In addition to activating the receptor PAR on the surface of platelets and the coagulation factors V, VIII, XI, and XII, thrombin transforms fibrinogen to fibrin [194].
Platelets have long been known to play an active role in angiogenesis, from the initial stages of vasculogenesis through the advanced stages.
While it has been demonstrated that platelets triggered by VEGF assist in the extravasation and metastasis of tumor cells, platelet-derived VEGF also enhances angiogenesis by encouraging the recruitment of further endothelial cells in the TME in OC [195]. In vitro investigations have shown that exposing OC cell lines to activated platelets significantly increases VEGF production in a cellular medium [196]. The endothelial markers VEGF and CD31 were discovered to co-localize with platelets in murine models of OC, while co-culture of human OC cell lines with platelets increased the secretion of many pro-angiogenic factors [197,198]. Surprisingly, this preclinical evidence reveals that metformin may inhibit platelet pro-angiogenic activity in OC [198]. Platelets may also play a role in tumor growth once anti-angiogenic therapy is discontinued; discontinuation of anti-VEGF medications is associated with accelerated tumor growth and simultaneous tumor platelet infiltration, while platelet depletion mitigates these effects in vivo [199].

4. Prognostic Significance of Immune Cell and Platelets in Ovarian Cancer

Many aspects of the immunological response to OC have predictive value. The presence of various immune cells and platelets infiltrating or interacting with the tumor has been linked to better and worse disease prognoses, indicating pro- or anti-tumor activities. Investigations on diverse tumor tissues have revealed significant infiltration of immune cells and platelets into the intratumoral and peritumoral areas. The presence of these cells is linked to tumor responses to a plethora of cytokines and chemokines produced and released by tumor cells. Its detection in OC allows for better clinical outcome prediction than other histological markers, indicating that the immunological profile can be useful in alternate forms of treatment, such as immunotherapy.

4.1. Tumor-Associated Macrophages

Research on the presence of TAMs in OC shows that the number of TAMs in malignant ovarian tumors is significantly higher than in benign and borderline tumors [200,201].
High numbers of M2-like TAMs in primary and metastatic OC of all histologies are associated with a lower sensitivity to treatment and a poor prognosis.
M1-like TAMs are a positive prognostic factor in women with EOC, owing to their ability to promote robust inflammatory responses that limit disease progression, despite the fact that their presence is significantly reduced in the TME of patients with advanced OC (Table 1).
Retrospective research of patients with advanced OC examined the expression of TAMs in tumor tissues using CD68 and CD163 as M1 and M2 macrophage markers, respectively. When comparing high vs. low CD163 (M2 TAMs), there was a substantial difference in progression-free (PFS) and overall survival (OS), with the low CD163+ groups significantly exceeding the high CD163+ groups [202]. Yafei et al. investigated the predictive value of CD68+ and CD163+ positive macrophages in 42 OC patients at various stages of disease. Immunohistochemical research revealed that a high proportion of CD163+ (M2 phenotype) in the total CD68+ macrophages predicted a poor prognosis [203].
A meta-analysis of 794 OC patients was performed to determine the relationship between the TAMs phenotype and clinical outcomes [204]. The infiltration of tumor tissue with CD163+ TAMs was associated with a poor prognosis, whereas a high M1-to-M2 macrophage ratio predicted a better prognosis for both OS and PFS. Another study on 112 patients with advanced OC found that a high M1/M2 ratio of TAMs in tumor specimens was associated better disease outcome [205].
Another M2-related marker was linked to a worse outcome in OC. While absolute CD206+ cell counts were not predictive, a high CD206/CD68 ratio was substantially associated with a worse PFS and worse OS [206]. Research on M1 (HLA-DR and iNOS) and M2-polarization (CD163 and VEGF) markers in OC patients found that a greater M1/M2 ratio was associated with improved patient survival [40,205]. Zhang et al. measured the M1/M2 ratio in the tumor and the stroma, and discovered that only the M1/M2 ratio of overall tumor macrophages or macrophages present intratumorally were prognostic, while the M1/M2 ratio in the tumor stroma was not, indicating that macrophages infiltrating tumor cells may play a more important role in tumor progression [205]. Furthermore, B7-H4 expression on the surface of TAMs, but not in ovarian tumor cells, was associated with decreased survival, and the number of B7-H4+ macrophages was significantly increased in advanced stages of disease [43].
Table 1. Characteristics of studies on tumor-associated macrophages and ovarian cancer prognostic significance.
Table 1. Characteristics of studies on tumor-associated macrophages and ovarian cancer prognostic significance.
Author/YearHistologyStageMethodImpactFindings
Tan et al., 2021 [207]n.an.aMeta-analysisDetrimentalHigh density of M2 TAMs negatively correlated with OS.
Sue-A-Quan et al., 2021 [208]CCOCI-IIIIHCDetrimentalHigh density of CD68+ TAMs was associated with shorter DFS, RFS and OS.
Macciò et al., 2020 [209]EOCIIIC-IVFCBeneficialHigh density of M1 TAMs, and high M1/M2 ratio correlated with longer OS, PFS and PFI.
Badmann et al., 2020 [210]EOCI-IVIHC, IFDetrimentalInfiltration of MDR1+ CD163 + CD68+
M2 TAMs was associated with poor prognosis.
Yuan et al., 2017 [204]n.aI-IVMeta-analysisDetrimentalHigher M1/M2 TAMs was associated with a favorable OS and PFS.
High density of CD163+ M2 TAMs correlated with poor disease outcome.
Yin et al., 2016 [123]EOCIII-IVIHCDetrimentalHigh density of EGF-secreting
M2-like TAMs correlated with
poor disease outcome.
Zhang et al., 2014 [205]EOCI-IVIHC, IF, FCBeneficialHigh M1/M2 TAMs ratio correlated with improved prognosis.
Reinartz et al., 2014 [31]HGSOCI-IVFCDetrimentalHigh density of CD163+ TAMs correlated with poor RFS and OS.
He et al., 2013 [40]EOCII-IVIHC, IFDetrimentalHigh density of M2 TAMs negatively correlated with OS.
EOC, various subtypes; CCOC, clear cell carcinoma; HGSOC, high-grade serous ovarian cancer; OS, overall survival; PFS, progression-free survival; PFI, platinum-free interval; RFS, relapse-free survival; DFS, disease-free survival, IHC, immunohistochemistry; IF, immunofluorescence; FC, flow cytometry; n.a, not available.

4.2. Dendritic Cells

Recently, studies have shown that DCs may infiltrate OC tumors and have a beneficial or adverse prognostic impact on patients, depending on the subpopulation (Table 2). According to research carried out on HGSOC cancer patients, the presence of mature, DC-LAMP+ DCs in TMA was correlated with improved OS. Furthermore, it should be noted that most mature DC-LAMP+ DCs are found in the tumor stroma and are linked to tertiary lymphoid structures (TLSs) rather than being in close proximity to malignant cells [211,212]. However, a higher density of mature DCs in the ovarian TME is associated with cytotoxic activity and Th1 polarization biomarkers, both of which are promising indicators in patients with OC [211,212].
Mastelic-Gavillet et al.’s investigation showed that a high expression of CLEC9A (cDC1s marker) in tissues of OC patients was associated with better OS. However, cDC1s are quantitatively and qualitatively impaired in patients with OC [213].
A high density of pDC in TME is often connected to immunosuppression and a worse outcome in OC. In the cohort studies of 44 patients, the accumulation of CD4+ BDCA2+ CD123+ pDC in TME, but not in ascites, was an independent prognostic factor associated with early relapse [48]. Notably, while pDC were the most abundant DC subgroup in TME and ascites, they were greatly diminished in the peripheral blood [48]. Similar results were obtained in immunohistochemical research performed on a cohort of 99 patients, where the presence of tissue-associated pCDs was correlated with PFS (14.6 months compared with 26.2 months in the absence of tissue-associated pDCs) [214]. According to these findings, pDC are mostly recruited into tumors where they exhibit a partially mature phenotype that would indicate in situ activation [48,214].
These findings indicate that while DC immunotherapies have considerable promise for the treatment of OC, they should be performed in conjunction with strategies for avoiding the quantitative and functional defects of DCs [215]. It has been demonstrated that treatment with CD40 and TLR3 agonists may transform tumor-associated DC into DC capable of stimulating anti-tumor activation of T-cells [216,217]. Moreover, it should target the subset that plays the most significant role in eliciting an anti-tumor response.
Table 2. Characteristics of studies on dendritic cells and ovarian cancer prognostic significance.
Table 2. Characteristics of studies on dendritic cells and ovarian cancer prognostic significance.
Author/YearHistologyStageMethodImpactFindings
Hensler et al., 2020 [211]HGSOCIII-IVIHCBeneficialHigh density of mature DC-LAMP+
DCs in peritoneal metastasis
correlated with improved disease outcome.
Mastelic-Gavillet et al., 2020 [213]n.aI-IVFCBeneficialHigh expression of CLEC9A (cDC1s marker) in tissues was associated with better OS.
Truxova et al., 2018 [212]HGSOCI-IVIHCBeneficialHigh density of tumor-infiltrating DC-LAMP+ mDCs correlated with cytotoxic activity and favorable OS.
Zhang et al., 2015 [218]EOCI-IVIHCBeneficialHigh density of CD1a+ mDCs correlated with improve OS.
Labidi-Galy et al., 2012 [214]n.an.aIHCDetrimentalCD4 + BDCA2 + CD123+ pDC in tumors correlated with poor disease outcome.
Conrad et al., 2012 [60]EOCII-IVIHC, FCDetrimentalHigh density of HLA-DR + CD123+ pDCs and ICOS+ FOXP3+ Treg cells correlated with poor disease outcome.
EOC various subtypes; HGSOC, high-grade serous ovarian cancer; OS, overall survival; IHC, immunohistochemistry; FC, flow cytometry; n.a, not available.

4.3. Myeloid-Derived Suppressor Cells

Correlative studies in humans linking the number and phenotype of MDSC with a poor prognosis, as well as animal studies, have revealed the clinical importance of MDSCs. A higher MDSC frequency in the tumor tissue, peripheral blood, or ascites is linked with a lower OS or relapse-free survival (Table 3). Mouse model studies have shown that MDSC accumulation during tumor formation and the ablation of MDSCs resulted in improved survival [219]. Lee et al. discovered that women with germline BRCA1- and BRCA2-mutation-associated OC have fewer circulating immunosuppressive immune cells compared with those with BRCA wild-type hereditary BRCA1 OC, which is thought to respond better to platinum-based chemotherapy than BRCA wild-type OC and has fewer circulating MDSCs and greater CD8+ T cells in peripheral blood mononuclear cells (PBMC) than BRCA wild-type OC [220]. Additionally, Li et al. found that metformin treatment in diabetic OC patients was related to lower circulating MDSCs, an increase in circulating CD8+ T cells, and a longer survival [221].
MDSCs decreased the antitumor immunity by using a variety of inflammatory mediators. Okła et al. performed a comprehensive analysis of the MDSC subpopulation and immunosuppressive factors, finding that high levels of M-MDSCs were associated with an advanced stage and high grade of OC and were significantly increased in blood-circulating immunosuppressive factors (Arg, IDO, and IL-10) compared with the healthy group. Furthermore, a decreased level of M-MDSCs was strongly correlated with prolonged survival in OC patients [222]. Wu et al. revealed that IL-6 and IL-10 from ascites increased the recruitment/production of M-MDSC in the peripheral blood and ascitic of OC patients and high levels of M-MDSC were correlated with a poor prognosis [223]. Additionally, the M-MDSC/DC cell ratio is an independent predictive factor for EOC survival and a high ratio is correlated with poor OS [224]. All of these findings suggest that the M-MDSCs subpopulation may have the highest clinical significance in MDSC populations.
Although all of these studies have demonstrated that MDSC has a predictive value in OC, they have significant drawbacks, such as a small study group, the use of variety of MDSC surface markers, and most studies being conducted in a single institution.
Table 3. Characteristics of studies on MDSC and ovarian cancer prognostic significance.
Table 3. Characteristics of studies on MDSC and ovarian cancer prognostic significance.
Author/YearHistologyStageMethodImpactFindings
Okła et al., 2020 [225]EOCI-IVIHCNoneHigh density of PD-L1+
MDSCs was not associated.
with disease outcome.
Komura et al. 2020 [141]n.an.aIHC, FCDetrimentalMDSC increases the stem cell-like properties and tumor PD-L1 expression by PGE2 production.
Okła et al., 2019 [222]EOCI-IVFCDetrimentalHigh density of M-MDSCs in peritoneal fluid correlated with poor disease outcome.
Lee et al., 2019 [220]HGSOC,
OCCC
III-IVFCBeneficialPatients with gBRCAm mutation correlated with low level of MDSC in peripheral blood and better disease outcome.
Li et al., 2018 [221]EOCI-IVFCDetrimentalMetformin inhibition of CD39+/CD73+ MDSCs improve antitumor T-cell immunity.
Taki et al., 2018 [226]HGSOCIII-IVIHC, FCDetrimentalHigh MDSC infiltration correlated with high CXCL1/2 level and short OS.
Santegoets et al., 2018 [224]EOCn.aFCDetrimentalHigh density of M-MDSC and low M-MDSC/DC ratio are associated with poor disease outcome.
Horikawa et al., 2017 [146]HGSOCIII-IVIHC, FCDetrimentalHigh VEGF expression induced MDSCs and correlated with poor prognosis.
Cui et al., 2013 [142]HGSOCI-IVIHC, FCDetrimentalHigh density of MDSC, significantly correlated with poor disease
outcome.
Obermajer et al., 2011 [135]n.aIII-IVFCDetrimentalHigh CXCL12 levels correlated.
with accumulation of MDSCs
in malignant ascites and poor
disease outcome.
EOC various subtypes; CCOC, clear cell carcinoma; HGSOC, high-grade serous ovarian cancer; OS, overall survival; IHC, immunohistochemistry; FC, flow cytometry; n.a, not available; gBRCAm, germline mutation.

4.4. Lymphocytes

There has been extensive research in the last 20 year regarding TILs and their potential as predictive biomarkers in patients with OC (Table 4). Several publications indicate that a higher level of intraepithelial CD3+, CD8+, and CD103+ cells is associated with a more favorable clinical outcome [227,228,229].
Importantly, a study of 186 OC patients with an advanced stage of disease (FIGO III/IV) found that patients with CD3+ TILs had a 5-year OS rate of 38.0%, while patients without detectable CD3+ TILs had a 5-year overall survival rate of only 4.5%. Moreover, complete responses (CR) occurred in 73.9% of patients with CD3+ TILs compared with 11.9% of patients without TILs who underwent surgical debulking and platinum-based chemotherapy [227].
According to Hamanishi et al., study patients with CD8+ TILs had longer PFS and OS [230]. Similar results were found by Sato et al.; patients with higher percentages of CD8+ TILs demonstrated improved survival compared with patients with lower percentages (55 months vs. 26 months) [227]. Nonetheless, the groups with high vs. low CD8+/CD4+ TILs ratios had a median survival of 74 and 25 months, respectively. These findings indicate that CD4+ TILs influenced the beneficial outcomes of CD8+ TILs [227]. In retrospective study of 500 OC patients, intraepithelial CD8+ TILs were correlated with improved disease-specific survival (DSS) from serous ovarian carcinomas, but not in endometrioid or clear cell carcinomas [231].
A meta-analysis of 19 studies comprising 6004 patients with HGSOC provided evidence of CD3+, CD4+, CD8+, and CD103+ TILs positively correlated with OS and PFS [232]. Furthermore, patients with primary or metastatic lesions with a high density of CD8+ TILs and a high CD8+/FOXP3+ cell ratio had a better DSS [233].
There are inconsistent data on Treg FoxP3+ cells, with the majority of studies supporting the association between Treg cells and a poor prognosis, while some studies revealed no association or even a beneficial effect on OS. Curiel et al. divided patients based on their numbers of CD4 + CD25 + FOXP3+ Treg cells (high, medium, and low), and found that Treg cells in tumor sites were associated with poor survival and a significant risk of mortality. Patients in the high Treg cell group experienced a 25.1-fold higher death hazard compared with those in the low Treg cell group [83]. Similarly, Herman et al. found that FoxP3+ T cells located within lymphoid aggregates surrounding the tumor were strongly associated with a reduced survival time [234]. Additionally, a high Treg/Th17 ratio was an independent poor prognostic factor for OS in OC patients [235]. In apparent contrast with other reports, Milne et al. revealed that the presence of intraepithelial FoxP3+ T cells was associated with increased DSS [236]. However, in a big meta-analysis across diverse types of cancer, including OC, it was not concluded that FoxP3+ Tregs cells in OC patients are a significant prognostic indicator of survival [237].
Furthermore, increased CD20+ TIL levels are associated with improved survival in OC patients [236,238]. Intratumor infiltration of CD27- atypical memory B cells, together with CD8+ T cells in HGSOC, is associated with a better prognosis [239]. Kroeger et al. discovered that a high infiltration of CD8+ T cells, CD20+ B cells, and plasma cells in OC tumors was associated with the presence of TLS in the TME and improved patient survival. Importantly, tumors containing CD8+ TILs only had an improved prognosis when identified in conjunction with CD20+ B cells, CD4+ T cells, and plasma cells, indicating cooperation between those subsets and their involvement in supporting antitumor immunity [240]. However, another study showed that a high expression of CD138+ plasma cells was correlated with a significantly reduced OS and OC-specific survival [241].
Furthermore, PD-1 and PD-L1 can be regarded as independent prognostic factors in OC [238]. Although the earliest findings on PD-L1 in OC suggested that PD-L1 expression on tumor cells was associated with a poor prognosis, some recent studies have revealed no association or even an improved outcome [230,242,243,244].
Table 4. Characteristics of studies on lymphocytes and ovarian cancer prognostic significance.
Table 4. Characteristics of studies on lymphocytes and ovarian cancer prognostic significance.
Author/YearHistologyStageMethodImpactFindings
Hao et al., 2020 [232]HGSOCI-IVMeta-analysesBeneficialCD3+, CD4+, CD8+, and CD103+ TILs positively correlated with PFS, OS.
Henriksen et al., 2020 [245]HGSOCI-IVIHCBeneficialHigh density of CD8+ TILs and PD-L1 expression correlated with favorable prognostic outcome.
Zhu et al., 2020 [246]EOCII-IVRetrospective analysisBeneficialHigh density of CD38+ was associated with longer DFS and increased OS.
Fucikova et al., 2019 [247]HGSOCI-IVIHC, FCDetrimentalHigh density of PD-1+ TIM-3+ CD8+ T cells correlated with poor disease outcome.
Truxova et al., 2018 [212]HGSOCI-IVIHCBeneficialHigh density of CD8+ T cells and CD20+ B cells correlated with improve survival.
Zhou et al., 2018 [235]EOCIII-IVFC, miRNA microarrayDetrimentalHigh Treg cells/Th17 cells ratio, derived by TAMs correlated with disease progression and metastasis potential.
Li et al., 2017 [248]EOCIII-IVMeta-analysisBeneficialIntraepithelial CD3+ and CD8+ CD103 + TILs correlated with improved survival.
Wang et al., 2017 [249]HGSOCI-IVIHCBeneficialCD3+ or CD8+ TILs were positively correlated with longer OS.
Montfort et al., 2017 [250]HGSOCIII-IVIHC, FCBeneficialHigh density of memory 20+ B cells in omental metastasis correlated with cytolytic favorable disease outcome.
Darb-Esfahan et al., 2016 [251]HGSOCI-IVIHCBeneficialHigh density of CD3+ T cells and high expression of PD-L1 correlated with PFS and OS.
Santoiemma et al., 2016 [238]EOCI-IVIHCBeneficialHigh density of CD8+ T cells and CD20+ B cells TILs positively correlated with OS
Lundgren et al., 2016 [241]EOCI-IVIHCDetrimentalHigh density of CD138+ plasma cells correlated with poor OS.
Kroeger et al., 2016 [240]HGSOCI-IVIHC, FCBeneficialTLS with high density of CD8+, CD4+ T cell, CD20+ B cells and plasma cells (CD20− CD38, CD138, and CD79a) correlated improve disease outcome.
Zhang et al., 2015 [218]EOCI-IVIHCBeneficialHigh density of CD45RO+ T cells correlated with higher survival rates
Shang et al., 2015 [237]n.aI-IVMeta-analysisNoneFoxP3 + Tregs cells did not correlated with OS.
Knutson et al., 2015 [252]EOCI-IVIFBeneficialHigh CD8+ T cells and CD4+/Treg cells ratio was associated with improved OS.
Webb et al., 2014 [253]EOCI-IIIIHC, FCBeneficialIntraepithelial CD103+, CD8+ TILs correlated with improved survival.
Hermans et al., 2014 [234]EOCI-IVIHCBeneficial/
Detrimental
CD8+ TILs positively correlated with OS. Treg cells lymphoid aggregates correlated with reduce OS.
Iglesia et al., 2014 [254]n.an.aRetrospective analysisBeneficialBCR gene segments correlated with improved prognosis.
Bachmayr-Heyda et al., 2013 [255]EOCII-IVIHCBeneficialHigh densities of CD8+ TILs correlated with better OS.
Hwang et al., 2012 [256]n.an.aMeta-analysesBeneficialIntraepithelial CD3+ CD8+ TILs correlated with a improve survival.
Nielsen et al., 2012 [239]HGSOCII-IVIHC, FCBeneficialThe presence of both CD20+ and CD8+ TIL correlated with increased patient survival. Additionally, CD27-CD20+ memory B cells correlated with cytolytic immune response and favorable prognosis.
Callahan et al., 2010 [257]EOCIIIB-IVIHCBeneficialTumor cell expression of HLA-CD8 + TILs correlated with improved survival.
Barnett et al., 2010 [258]EOCI-IVIHC, IFBeneficialCD8+ TILs positively correlated with improved disease outcome.
Leffers et al., 2009 [233]EOCI–IVIHCBeneficialHigh densities of CD8+, CD45R0+, Treg and a high ratio CD8+/FoxP3+ Treg correlated with DSS PFS, OS.
Milne et al., 2009 [236]HGSOCI-IVIHCBeneficialHigh density of CD3+, CD8+, Treg and CD20+ B cells correlated with improved disease outcome
Stumpf et al., 2009 [259]SOCIIIIHCBeneficialIntraepithelial CD3+ CD8+ correlated with improved DFS and OS
Hamanishi et al., 2007 [230]EOCI-IVIHCDetrimentalIntraepithelial CD8+ TILs count was poor prognostic factor for PFS and OS.
Kryczek et al., 2007 [43]EOCI-IVIFDetrimentalTreg cells and TAMs B7-H4, correlated with poor disease outcome.
Sato et al., 2005 [227]EOCI-IVIHCBeneficialHigh density of intraepithelial CD8+ T cells and high CD8+/Treg cells ratio correlated with favorable outcome.
Curiel et al., 2004 [83]n.aI-IVIF, FCDetrimentalTreg cells in tumor and malignant ascites were associated with poor survival.
EOC various subtypes; HGSOC, high-grade serous ovarian cancer; SOC, serous ovarian cancer; OS, overall survival; DFS, disease-free survival; DSS, disease specific survival; PFS, progression-free survival; IHC, immunohistochemistry; IF, immunofluorescence; FC, flow cytometry; CD45R0+, memory T-lymphocytes; TLS, tertiary lymphoid structures; n.a, not available.

4.5. NK Cells

Despite the fact that only a few studies have been conducted to assess the prognostic significance of NK cells in OC, most have presented promising outcomes (Table 5). Immunohistochemical research revealed that a high level of CD56+ NK cells was correlated with a favorable prognostic impact; the OS was 45 months in patients with a high level of CD57+ NK cells compared with a 29-month OS in patients with a low level of CD56+ NK cells [245]. According to similar findings, in ascites of OC patients, a higher percentage of CD56+ NK cells was associated with a better PFS, and their immunity was boosted by IL-15, which could be a promising direction for new immunotherapy [260]. Another study using anti-CD103+ antibodies discovered that CD103 was highly expressed by intraepithelial CD8+ T cells and in also some patients by NK cells TILs in human ovarian tumors. Significantly, CD103 + TILs comprising intraepithelial, CD8+ T cells, and NK cells were strongly associated with survival in HGSOC patients, indicating that CD103 represents a clinically beneficial TIL subset that merits further investigation [253].
The prognostic value of the blood NK cell count and intratumoral NK cell density in OC could be a promising therapeutic therapy for OC patients.

4.6. Neutrophils

In recent years, the role of neutrophils in OC has attracted more attention, and theneutrophilia and neutrophil-to-lymphocytes ratio (NLR) has been associated with detrimental outcome in OC patients (Table 6). A limited number of studies have investigated the prognostic role of neutrophilia in OC. Komura et al. suggested that neutrophilia is a poor prognostic factor, but is inferior to NLR [262]. However, according to two earlier findings, neutrophilia is not a prognostic factor in relation to the disease outcome [263,264].
Multiple studies have concentrated on the value of the inflammatory composite marker NLR in which most reported a strong predictive value for predicting the survival of patients with OC. In a retrospective analysis of 315 newly diagnosed EOC patients undergoing platinum-taxane chemotherapy, it was found that a high pre-treatment NLR count was an independent negative prognostic factor for PFS, but not OS [265]. A similar observation was reported by Wang et al., who recognized a preoperative high NLR count as a prognostic factor for both OS and PFS [266]. It also was suggested that a high preoperative NLR ratio was associated with a greater risk of 30-day postoperative morbidity and poor OS [267]. Notably, a similar observation was reported by Nakamura et al., correlating a high NLR count with a higher mortality within 100 days of unsuccessful first-line chemotherapy [268].
Furthermore, preoperative NLR combined with CA125 was shown to be useful for the early detection of OC and to be able to predict the pathological diagnosis of adnexal masses [269,270]. Additionally, a high baseline NLR ratio was more correlated with distant metastases in patients with advanced gynecological cancers, including OC [271].
The majority of cut-off values for NLR are between 2 and 4. However, Forget et al. showed comparable values (0.78 to 3.53) in healthy people [272]. As a result, it is necessary to establish the NLR cut-off values for clinical applications in predicting OC.
In addition to NLR, other neutrophil-related markers have been investigated as potential prognostic biomarkers. Kim et al. found that elevated levels of NET markers (histone–DNA complex, cell-free DNA, and neutrophil elastase) were correlated with worse OS and PFS. Specifically, neutrophil elastase was identified as an independent prognostic factor in patients with HGSOC [273]. Contradictory observations were made by Muqaku et al. indicating that the release of S100A8/A9 protein, associated with the formation of NETs and a high S100A8/CRP ratio, was correlated with a favorable survival of HGSOC patients [274].
Table 6. Characteristics of studies on neutrophils and ovarian cancer prognostic significance.
Table 6. Characteristics of studies on neutrophils and ovarian cancer prognostic significance.
Author/YearHistologyStageMethodImpactFindings
Kim et al., 2022 [273]HGSOCI-IVELISADetrimentalHigh circulating levels of NET markers (histone−DNA complex, cell free DNA, and neutrophil elastase) and prekallikrein, correlated with poor OS.
Marchetti et al., 2021 [275]HGSOCIII-IVRetrospective studyDetrimentalHigh NLR correlated with negative PFS and OS.
Henriksen et al., 2020 [245]EOCI-IVFCDetrimentalHigh NLR was associated with lack of response to chemotherapy and a poor prognosis.
He et al., 2020 [276]SOCI-IVRetrospective studyDetrimentalEarly onset neutropenia correlated with chemosensitivity and favorable PFS and OS.
Singel et al., 2019 [277]EOCIII-IVIFDetrimentalHigh ascites mtDNA and neutrophil elastase was associated with reduced PFS.
Posabella et al., 2019 [278]HGSOCII-IVIHCBeneficialHigh density of CD66b+ neutrophils was associated with chemosensitivity and longer RFS.
Chen et al., 2018 [279]n.an.aMeta-analysisDetrimentalHigh NLR ratio correlated negatively with PFS and OS.
Komura et al., 2018 [262]EOCI–IVRetrospective studyDetrimentalPre-treatment neutrophilia and high NLR correlated with poor disease outcome.
Huang et al., 2017 [280]EOCI-IVMeta-analysisDetrimentalElevated pre-treatment NLR correlated with poor disease outcome.
EOC various subtypes; HGSOC, high-grade serous ovarian cancer; SOC, serous ovarian cancer; OS, overall survival; PFS, progression-free survival; RFS, recurrence-free survival; IHC, immunohistochemistry; FC, flow cytometry; IF, immunofluorescence; ELISA, enzyme-linked immunosorbent assay; n.a, not available.

4.7. Platelets

Platelets have been studied as a potential prognostic marker in OC (Table 7). A high platelet count or thrombocytosis has been wildly associated with a poor disease outcome in patients with OC. Preoperative thrombocytosis is correlated with worse disease-free survival (DFS). Furthermore, in an early disease stage (FIGO I/II), there was a five-fold increase in the risk of death and a nearly eight-fold risk of disease recurrence associated with thrombocytosis [281]. Similarly, in a meta-analysis including a total of 4953 patients with OC pre-treatment, thrombocytosis was significantly associated with OS and PFS [282].
Among 875 patients with HGSOC who underwent primary staging or debulking surgery, thrombocytosis was not found to be a predictor for PFS. However, hyperfibrinogenemia was correlated with OS but not PFS [283]. In a retrospective analysis of 179 women with advanced EOC who underwent cytoreductive surgery and chemotherapy preoperative thrombocytosis, a high platelet ratio and persistent thrombocytosis after chemotherapy combined with high CA-125 levels after chemotherapy were independent poor prognostic factors of OS [284]. In a multicenter case-control study of 1308 patients with advanced and early-stage ovarian clear cell carcinoma (OCCC) and serous ovarian carcinoma (SOC), patients with advanced OCCC were associated with decreased survival outcomes and an increased incidence of venous thromboembolism (VTE) [284].
One of the widely used markers of platelet activity is the platelet-to-lymphocytes ratio (PLR). A high PLR ratio has been associated with a poor prognosis in OC [285]. In patients who underwent debulking surgery, a high PLR had a negative correlation with DFS and OS [286]. Winarno et al. discovered that a high post-operative PLR ratio of patients with an EOC was associated with an increase in the response to platinum-based chemotherapy [287]. Several researchers have also shown that PLR could play a predictive role in the early diagnosis and in distinguishing between benign and malignant tumors [269,270,288,289]. In a cohort of 43 patients, researchers found that preoperative PLR was a predictor for the recurrence of OC [290]. As with NLR, standardized universal cut-off values for PLR are not defined.
Table 7. Characteristics of studies on platelets and ovarian cancer prognostic significance.
Table 7. Characteristics of studies on platelets and ovarian cancer prognostic significance.
Author/YearHistologyStageMethodImpactFindings
Kim et al., 2022 [291]EOCIII-IVRetrospective studyDetrimentalReactive thrombocytosis after cytoreductive surgery and chemotherapy was associated with poor OS and PFS.
Canzler et al., 2020 [292]EOCI-IVRetrospective studyDetrimentalPatients with pretreatment thrombocytosis responded less to treatment and corelated with worse OS and PFS.
Ye et al., 2019 [282]EOCI-IVMeta-analysisDetrimentalPre-treatment thrombocytosis corelated with worse OS and PFS.
Yildirim et al., 2015 [269]n.aI-IVRetrospective studyDetrimentalPreoperative high PLR ratio may help identify OC in patients with adnexal masses.
Ma et al., 2014 [293]EOCI-IVRetrospective studyDetrimentalThrombocytosis and MAR correlated negatively with OS.
Allensworth et al., 2013 [281]n.aI-IVRetrospective studyDetrimentalPreoperative thrombocytosis correlated with worse DFS and OS
Lee et al., 2011 [284]EOCIII-IVRetrospective studyDetrimentalPreoperative and post-chemotherapy thrombocytosis correlated with poor prognosis.
Asher et al., 2011 [285]EOCI-IVRetrospective studyDetrimentalPre-surgery high PLR ratio was associated with poor OS.
EOC various subtypes; OS, overall survival; PFS, progression-free survival; DFS, disease-free survival; MAR, maximal aggregation rate; n.a, not available.

5. Conclusions

The interaction between immune cells and platelets in TME is a complex process, and understanding the underlying mechanisms is essential for developing effective therapies. Because of the immunogenic nature of OC, immunotherapy presents a cutting-edge and potential therapeutic approach for the treatment of OC. Even though immunotherapy is evolving quickly and has produced remarkable results, many OC patients continue to experience resistance to it.
Upon arrival, TILs are faced with very immunosuppressive TME due to the presence of an immune resistance mechanism [294,295]. Several mechanisms contribute to evasion, including the presence of immunosuppressive cells (MDSCs, TAMs, or Treg cells), expression of inhibitory molecules (PD-1L, IDO, Arg I, IL-10, B7-H3, TGF-β, PGE2,CTLA-4, and Fas ligand), decreased and/or lost TAA expression, and down-regulation of MHC [296]. Therefore, a deeper understanding of the immune TME of OC can aid researchers in identifying some key sites for innovation that will increase the clinical efficacy of cancer immunotherapy. Therefore, we focused on the mechanisms of OC cells to recruit and re-educate immune cells and how this immunosuppressive network stops immune active cells such as TILs or NK cells from mounting an anti-tumor response.
Both genomic and proteomic methods have been used to discover numerous OC serum biomarkers, which have been assessed alone or in conjunction with CA125 [297,298]. However, the ideal biomarker or combination of biomarkers has not yet been identified. Given that CA125 has been the gold standard for diagnosing OC in the last few decades and considering the advancements in technologies for biomarker discovery, it is necessary to find new markers that may improve CA125’s sensitivity using a combination of hematologic, immunological, and inflammatory markers.

Author Contributions

Conceptualization, K.A.P. and J.O.; writing—draft preparation and editing, K.A.P., J.O., and G.E.B.; writing—original draft preparation K.A.P. and J.C.-S.; writing—review and editing, K.A.P.; supervision, J.O., M.R., and M.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed]
  2. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  3. Bankhead, C.R.; Collins, C.; Stokes-Lampard, H.; Rose, P.; Wilson, S.; Clements, A.; Mant, D.; Kehoe, S.T.; Austoker, J. Identifying symptoms of ovarian cancer: A qualitative and quantitative study. BJOG Int. J. Obstet. Gynaecol. 2008, 115, 1008–1014. [Google Scholar] [CrossRef]
  4. Hamilton, W.; Peters, T.; Bankhead, C.; Sharp, D. Risk of ovarian cancer in women with symptoms in primary care: Population based case-control study. BMJ 2009, 339, b2998. [Google Scholar] [CrossRef] [PubMed]
  5. Kurnit, K.C.; Fleming, G.F.; Lengyel, E. Updates and New Options in Advanced Epithelial Ovarian Cancer Treatment. Obstet. Gynecol. 2021, 137, 108–121. [Google Scholar] [CrossRef]
  6. The Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 2011, 474, 609–615. [Google Scholar] [CrossRef]
  7. Kurman, R.J.; Shih, I.-M. The Origin and Pathogenesis of Epithelial Ovarian Cancer: A Proposed Unifying Theory. Am. J. Surg. Pathol. 2010, 34, 433–443. [Google Scholar] [CrossRef]
  8. Reid, B.M.; Permuth, J.B.; Sellers, T.A. Epidemiology of ovarian cancer: A review. Cancer Biol. Med. 2017, 14, 9–32. [Google Scholar] [CrossRef]
  9. Jayson, G.C.; Kohn, E.C.; Kitchener, H.C.; Ledermann, J.A. Ovarian cancer. Lancet 2014, 384, 1376–1388. [Google Scholar] [CrossRef]
  10. Yap, T.A.; Carden, C.P.; Kaye, S.B. Beyond chemotherapy: Targeted therapies in ovarian cancer. Nat. Rev. Cancer 2009, 9, 167–181. [Google Scholar] [CrossRef]
  11. Huang, L.; Cronin, K.A.; Johnson, K.A.; Mariotto, A.B.; Feuer, E.J. Improved survival time: What can survival cure models tell us about population-based survival improvements in late-stage colorectal, ovarian, and testicular cancer? Cancer 2008, 112, 2289–2300. [Google Scholar] [CrossRef]
  12. Braun, A.; Anders, H.-J.; Gudermann, T.; Mammadova-Bach, E. Platelet-Cancer Interplay: Molecular Mechanisms and New Therapeutic Avenues. Front. Oncol. 2021, 11, 665534. [Google Scholar] [CrossRef]
  13. Pradeep, S.; Kim, S.W.; Wu, S.Y.; Nishimura, M.; Chaluvally-Raghavan, P.; Miyake, T.; Pecot, C.V.; Kim, S.-J.; Choi, H.J.; Bischoff, F.Z.; et al. Hematogenous Metastasis of Ovarian Cancer: Rethinking Mode of Spread. Cancer Cell 2014, 26, 77–91. [Google Scholar] [CrossRef]
  14. Korniluk, A.; Koper, O.; Kemona, H.; Dymicka-Piekarska, V. From inflammation to cancer. Ir. J. Med. Sci. 2017, 186, 57–62. [Google Scholar] [CrossRef]
  15. Rasmussen, C.B.; Kjaer, S.K.; Albieri, V.; Bandera, E.V.; Doherty, J.A.; Høgdall, E.; Webb, P.M.; Jordan, S.J.; Rossing, M.A.; Wicklund, K.G.; et al. Pelvic Inflammatory Disease and the Risk of Ovarian Cancer and Borderline Ovarian Tumors: A Pooled Analysis of 13 Case-Control Studies. Am. J. Epidemiol. 2017, 185, 8–20. [Google Scholar] [CrossRef]
  16. Macciò, A.; Madeddu, C. Inflammation and ovarian cancer. Cytokine 2012, 58, 133–147. [Google Scholar] [CrossRef]
  17. Savant, S.S.; Sriramkumar, S.; O’hagan, H.M. The Role of Inflammation and Inflammatory Mediators in the Development, Progression, Metastasis, and Chemoresistance of Epithelial Ovarian Cancer. Cancers 2018, 10, 251. [Google Scholar] [CrossRef]
  18. Charbonneau, B.; Goode, E.L.; Kalli, K.R.; Knutson, K.L.; DeRycke, M.S. The Immune System in the Pathogenesis of Ovarian Cancer. Crit. Rev. Immunol. 2013, 33, 137–164. [Google Scholar] [CrossRef]
  19. Grivennikov, S.I.; Greten, F.R.; Karin, M. Immunity, inflammation, and cancer. Cell 2010, 140, 883–899. [Google Scholar] [CrossRef]
  20. Hourani, T.; Holden, J.A.; Li, W.; Lenzo, J.C.; Hadjigol, S.; O’brien-Simpson, N.M. Tumor Associated Macrophages: Origin, Recruitment, Phenotypic Diversity, and Targeting. Front. Oncol. 2021, 11, 788365. [Google Scholar] [CrossRef]
  21. Mills, C.D. Anatomy of a Discovery: M1 and M2 Macrophages. Front. Immunol. 2015, 6, 212. [Google Scholar] [CrossRef] [PubMed]
  22. Wang, H.; Yung, M.M.H.; Ngan, H.Y.S.; Chan, K.K.L.; Chan, D.W. The Impact of the Tumor Microenvironment on Macrophage Polarization in Cancer Metastatic Progression. Int. J. Mol. Sci. 2021, 22, 6560. [Google Scholar] [CrossRef] [PubMed]
  23. Sica, A. Role of tumour-associated macrophages in cancer-related inflammation. Exp. Oncol. 2010, 32, 153–158. [Google Scholar] [PubMed]
  24. Ramanathan, S.; Jagannathan, N. Tumor Associated Macrophage: A Review on the Phenotypes, Traits and Functions. Iran. J. Cancer Prev. 2014, 7, 1–8. [Google Scholar]
  25. Van Dalen, F.J.; van Stevendaal, M.H.M.E.; Fennemann, F.L.; Verdoes, M.; Ilina, O. Molecular repolarisation of tumour-associated macrophages. Molecules 2019, 24, 9. [Google Scholar] [CrossRef]
  26. Roy, A.; Li, S. Modifying the tumor microenvironment using nanoparticle therapeutics. WIREs Nanomed. Nanobiotechnol. 2016, 8, 891–908. [Google Scholar] [CrossRef]
  27. Gordon, S. Alternative activation of macrophages. Nat. Rev. Immunol. 2003, 3, 23–35. [Google Scholar] [CrossRef]
  28. Ostuni, R.; Kratochvill, F.; Murray, P.J.; Natoli, G. Macrophages and cancer: From mechanisms to therapeutic implications. Trends Immunol. 2015, 36, 229–239. [Google Scholar] [CrossRef]
  29. Xue, J.; Schmidt, S.V.; Sander, J.; Draffehn, A.; Krebs, W.; Quester, I.; De Nardo, D.; Gohel, T.D.; Emde, M.; Schmidleithner, L.; et al. Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity 2014, 40, 274–288. [Google Scholar] [CrossRef]
  30. Singhal, S.; Stadanlick, J.; Annunziata, M.J.; Rao, A.S.; Pratik, S.; Brien, S.O.; Moon, E.K.; Cantu, E.; Danet-Desnoyers, G.; Ra, H.; et al. Human tumor-associated monocytes/macrophages and their regulation of T cell responses in early-stage lung cancer. Sci. Transl. Med. 2019, 11, eaat1500. [Google Scholar] [CrossRef]
  31. Reinartz, S.; Schumann, T.; Finkernagel, F.; Wortmann, A.; Jansen, J.M.; Meissner, W.; Krause, M.; Schwörer, A.; Wagner, U.; Müller-Brüsselbach, S.; et al. Mixed-polarization phenotype of ascites-associated macrophages in human ovarian carcinoma: Correlation of CD163 expression, cytokine levels and early relapse. Int. J. Cancer 2014, 134, 32–42. [Google Scholar] [CrossRef]
  32. Thibault, B.; Castells, M.; Delord, J.-P.; Couderc, B. Ovarian cancer microenvironment: Implications for cancer dissemination and chemoresistance acquisition. Cancer Metastasis Rev. 2014, 33, 17–39. [Google Scholar] [CrossRef]
  33. Hagemann, T.; Wilson, J.; Burke, F.; Kulbe, H.; Li, N.F.; Plüddemann, A.; Charles, K.; Gordon, S.; Balkwill, F.R. Ovarian Cancer Cells Polarize Macrophages toward A Tumor-Associated Phenotype. J. Immunol. 2006, 176, 5023–5032. [Google Scholar] [CrossRef]
  34. Chambers, S.K.; Kacinski, B.M.; Ivins, C.M.; Carcangiu, M.L. Overexpression of epithelial macrophage colony-stimulating factor (CSF-1) and CSF-1 receptor: A poor prognostic factor in epithelial ovarian cancer, contrasted with a protective effect of stromal CSF-1. Clin. Cancer Res. 1997, 3, 999–1007. [Google Scholar]
  35. Cai, D.L.; Jin, L.-P. Immune Cell Population in Ovarian Tumor Microenvironment. J. Cancer 2017, 8, 2915–2923. [Google Scholar] [CrossRef]
  36. Drakes, M.L.; Stiff, P.J. Regulation of Ovarian Cancer Prognosis by Immune Cells in the Tumor Microenvironment. Cancers 2018, 10, 302. [Google Scholar] [CrossRef]
  37. Moughon, D.L.; He, H.; Schokrpur, S.; Jiang, Z.K.; Yaqoob, M.; David, J.; Lin, C.; Iruela-Arispe, M.L.; Dorigo, O.; Wu, L. Macrophage Blockade Using CSF1R Inhibitors Reverses the Vascular Leakage Underlying Malignant Ascites in Late-Stage Epithelial Ovarian Cancer. Cancer Res. 2015, 75, 4742–4752. [Google Scholar] [CrossRef]
  38. Duluc, D.; Delneste, Y.; Tan, F.; Moles, M.-P.; Grimaud, L.; Lenoir, J.; Preisser, L.; Anegon, I.; Catala, L.; Ifrah, N.; et al. Tumor-associated leukemia inhibitory factor and IL-6 skew monocyte differentiation into tumor-associated macrophage-like cells. Blood 2007, 110, 4319–4330. [Google Scholar] [CrossRef]
  39. Allavena, P.; Chieppa, M.; Bianchi, G.; Solinas, G.; Fabbri, M.; Laskarin, G.; Mantovani, A. Engagement of the Mannose Receptor by Tumoral Mucins Activates an Immune Suppressive Phenotype in Human Tumor-Associated Macrophages. Clin. Dev. Immunol. 2010, 2010, 547179. [Google Scholar] [CrossRef]
  40. He, Y.-F.; Zhang, M.-Y.; Wu, X.; Sun, X.-J.; Xu, T.; He, Q.-Z.; Di, W. High MUC2 Expression in Ovarian Cancer Is Inversely Associated with the M1/M2 Ratio of Tumor-Associated Macrophages and Patient Survival Time. PLoS ONE 2013, 8, e79769. [Google Scholar] [CrossRef]
  41. Li, H.; Fan, X.; Houghton, J. Tumor microenvironment: The role of the tumor stroma in cancer. J. Cell. Biochem. 2007, 101, 805–815. [Google Scholar] [CrossRef] [PubMed]
  42. Rodriguez, P.C.; Quiceno, D.G.; Zabaleta, J.; Ortiz, B.; Zea, A.H.; Piazuelo, M.B.; Delgado, A.; Correa, P.; Brayer, J.; Sotomayor, E.M.; et al. Arginase I Production in the Tumor Microenvironment by Mature Myeloid Cells Inhibits T-Cell Receptor Expression and Antigen-Specific T-Cell Responses. Cancer Res. 2004, 64, 5839–5849. [Google Scholar] [CrossRef] [PubMed]
  43. Kryczek, I.; Wei, S.; Zhu, G.; Myers, L.; Mottram, P.; Cheng, P.; Chen, L.; Coukos, G.; Zou, W. Relationship between B7-H4, Regulatory T Cells, and Patient Outcome in Human Ovarian Carcinoma. Cancer Res. 2007, 67, 8900–8905. [Google Scholar] [CrossRef] [PubMed]
  44. Ying, X.; Wu, Q.; Wu, X.; Zhu, Q.; Wang, X.; Jiang, L.; Chen, X.; Wang, X. Epithelial ovarian cancer-secreted exosomal miR-222-3p induces polarization of tumor-associated macrophages. Oncotarget 2016, 7, 43076–43087. [Google Scholar] [CrossRef] [PubMed]
  45. Banchereau, J.; Briere, F.; Caux, C.; Davoust, J.; Lebecque, S.; Liu, Y.; Pulendran, B.; Palucka, K. Immunobiology of dendritic cells. Annu. Rev. Immunol. 2000, 18, 767–811. [Google Scholar] [CrossRef]
  46. Timmerman, J.M.; Levy, R. Dendritic Cell Vaccine. In Encyclopedic Dictionary of Genetics, Genomics and Proteomics; Wiley: Hoboken, NJ, USA, 2004. [Google Scholar] [CrossRef]
  47. Riboldi, E.; Musso, T.; Moroni, E.; Urbinati, C.; Bernasconi, S.; Rusnati, M.; Adorini, L.; Presta, M.; Sozzani, S. Cutting Edge: Proangiogenic Properties of Alternatively. J. Immunol. 2005, 175, 2788–2792. [Google Scholar] [CrossRef]
  48. Labidi-Galy, S.I.; Sisirak, V.; Meeus, P.; Gobert, M.; Treilleux, I.; Bajard, A.; Combes, J.-D.; Faget, J.; Mithieux, F.; Cassignol, A.; et al. Quantitative and Functional Alterations of Plasmacytoid Dendritic Cells Contribute to Immune Tolerance in Ovarian Cancer. Cancer Res. 2011, 71, 5423–5434. [Google Scholar] [CrossRef]
  49. Manh, T.-P.V.; Bertho, N.; Hosmalin, A.; Schwartz-Cornil, I.; Dalod, M. Investigating Evolutionary Conservation of Dendritic Cell Subset Identity and Functions. Front. Immunol. 2015, 6, 260. [Google Scholar] [CrossRef]
  50. Volovitz, I.; Melzer, S.; Amar, S.; Bocsi, J.; Bloch, M.; Efroni, S.; Ram, Z.; Tárnok, A. Dendritic Cells in the Context of Human Tumors: Biology and Experimental Tools. Int. Rev. Immunol. 2016, 35, 116–135. [Google Scholar] [CrossRef]
  51. Strioga, M.; Schijns, V.; Powell, D.J.; Pasukoniene, V.; Dobrovolskiene, N.; Michalek, J. Dendritic cells and their role in tumor immunosurveillance. J. Endotoxin Res. 2013, 19, 98–111. [Google Scholar] [CrossRef]
  52. Braun, A.; Worbs, T.; Moschovakis, G.L.; Halle, S.; Hoffmann, K.; Bölter, J.; Münk, A.; Forster, R. Afferent lymph-derived T cells and DCs use different chemokine receptor CCR7-dependent routes for entry into the lymph node and intranodal migration. Nat. Immunol. 2011, 12, 879–887. [Google Scholar] [CrossRef] [PubMed]
  53. Comerford, I.; Harata-Lee, Y.; Bunting, M.D.; Gregor, C.; Kara, E.E.; McColl, S.R. A myriad of functions and complex regulation of the CCR7/CCL19/CCL21 chemokine axis in the adaptive immune system. Cytokine Growth Factor Rev. 2013, 24, 269–283. [Google Scholar] [CrossRef] [PubMed]
  54. Diao, J.; Zhao, J.; Winter, E.; Cattral, M.S. Tumors Suppress In Situ Proliferation of Cytotoxic T Cells by Promoting Differentiation of Gr-1+ Conventional Dendritic Cells through IL-6. J. Immunol. 2011, 186, 5058–5067. [Google Scholar] [CrossRef] [PubMed]
  55. Broz, M.L.; Binnewies, M.; Boldajipour, B.; Nelson, A.E.; Pollack, J.L.; Erle, D.J.; Barczak, A.; Rosenblum, M.D.; Daud, A.; Barber, D.L.; et al. Dissecting the Tumor Myeloid Compartment Reveals Rare Activating Antigen-Presenting Cells Critical for T Cell Immunity. Cancer Cell 2014, 26, 638–652. [Google Scholar] [CrossRef] [PubMed]
  56. Martinek, J.; Wu, T.-C.; Cadena, D.; Banchereau, J.; Palucka, K. Interplay between Dendritic Cells and Cancer Cells, 1st ed.; Elsevier: Amsterdam, The Netherlands, 2019; Volume 348. [Google Scholar] [CrossRef]
  57. Lin, H.; Wei, S.; Hurt, E.M.; Green, M.D.; Zhao, L.; Vatan, L.; Szeliga, W.; Herbst, R.; Harms, P.W.; Fecher, L.A.; et al. Host expression of PD-L1 determines efficacy of PD-L1 pathway blockade-mediated tumor regression. J. Clin. Investig. 2018, 128, 805–815. [Google Scholar] [CrossRef] [PubMed]
  58. Chen, F.; Hou, M.; Ye, F.; Lv, W.; Xie, X. Ovarian Cancer Cells Induce Peripheral Mature Dendritic Cells to Differentiate into Macrophagelike Cells In Vitro. Int. J. Gynecol. Cancer 2009, 19, 1487–1493. [Google Scholar] [CrossRef]
  59. Scarlett, U.K.; Rutkowski, M.R.; Rauwerdink, A.M.; Fields, J.; Escovar-Fadul, X.; Baird, J.; Cubillos-Ruiz, J.R.; Jacobs, A.C.; Gonzalez, J.L.; Weaver, J.; et al. Ovarian cancer progression is controlled by phenotypic changes in dendritic cells. J. Exp. Med. 2012, 209, 495–506. [Google Scholar] [CrossRef]
  60. Conrad, C.; Gregorio, J.; Wang, Y.-H.; Ito, T.; Meller, S.; Hanabuchi, S.; Anderson, S.; Atkinson, N.; Ramirez, P.T.; Liu, Y.-J.; et al. Plasmacytoid Dendritic Cells Promote Immunosuppression in Ovarian Cancer via ICOS Costimulation of Foxp3+ T-Regulatory Cells. Cancer Res. 2012, 72, 5240–5249. [Google Scholar] [CrossRef]
  61. Liefers-Visser, J.; Meijering, R.; Reyners, A.; van der Zee, A.; de Jong, S. IGF system targeted therapy: Therapeutic opportunities for ovarian cancer. Cancer Treat. Rev. 2017, 60, 90–99. [Google Scholar] [CrossRef]
  62. Huang, C.-T.; Chang, M.-C.; Chen, Y.-L.; Chen, T.-C.; Chen, C.-A.; Cheng, W.-F. Insulin-like growth factors inhibit dendritic cell-mediated anti-tumor immunity through regulating ERK1/2 phosphorylation and p38 dephosphorylation. Cancer Lett. 2015, 359, 117–126. [Google Scholar] [CrossRef]
  63. Somri-Gannam, L.; Meisel-Sharon, S.; Hantisteanu, S.; Groisman, G.; Limonad, O.; Hallak, M.; Bruchim, I. IGF1R Axis Inhibition Restores Dendritic Cell Antitumor Response in Ovarian Cancer. Transl. Oncol. 2020, 13, 100790. [Google Scholar] [CrossRef]
  64. Tanyi, J.L.; Bobisse, S.; Ophir, E.; Tuyaerts, S.; Roberti, A.; Genolet, R.; Baumgartner, P.; Stevenson, B.J.; Iseli, C.; Dangaj, D.; et al. Personalized cancer vaccine effectively mobilizes antitumor T cell immunity in ovarian cancer. Sci. Transl. Med. 2018, 10, eaao5931. [Google Scholar] [CrossRef]
  65. Krempski, J.; Karyampudi, L.; Behrens, M.D.; Erskine, C.L.; Hartmann, L.; Dong, H.; Goode, E.L.; Kalli, K.R.; Knutson, K.L. Tumor-Infiltrating Programmed Death Receptor-1+ Dendritic Cells Mediate Immune Suppression in Ovarian Cancer. J. Immunol. 2011, 186, 6905–6913. [Google Scholar] [CrossRef]
  66. Dolcetti, L.; Peranzoni, E.; Ugel, S.; Marigo, I.; Gomez, A.F.; Mesa, C.; Geilich, M.; Winkels, G.; Traggiai, E.; Casati, A.; et al. Hierarchy of immunosuppressive strength among myeloid-derived suppressor cell subsets is determined by GM-CSF. Eur. J. Immunol. 2010, 40, 22–35. [Google Scholar] [CrossRef]
  67. Atretkhany, K.-S.N.; Drutskaya, M.S. Myeloid-derived suppressor cells and proinflammatory cytokines as targets for cancer therapy. Biochemistry 2016, 81, 1274–1283. [Google Scholar] [CrossRef]
  68. Gabrilovich, D.I. Myeloid-Derived Suppressor Cells. Cancer Immunol. Res. 2017, 5, 3–8. [Google Scholar] [CrossRef]
  69. Obermajer, N.; Muthuswamy, R.; Lesnock, J.; Edwards, R.P.; Kalinski, P. Positive feedback between PGE2 and COX2 redirects the differentiation of human dendritic cells toward stable myeloid-derived suppressor cells. Blood 2011, 118, 5498–5505. [Google Scholar] [CrossRef]
  70. Wong, J.L.; Obermajer, N.; Odunsi, K.; Edwards, R.P.; Kalinski, P. Synergistic COX2 Induction by IFNγ and TNFα Self-Limits Type-1 Immunity in the Human Tumor Microenvironment. Cancer Immunol. Res. 2016, 4, 303–311. [Google Scholar] [CrossRef]
  71. Wouters, M.; Dijkgraaf, E.M.; Kuijjer, M.; Jordanova, E.S.; Hollema, H.; Welters, M.; Van Der Hoeven, J.; Daemen, T.; Kroep, J.; Nijman, H.W.; et al. Interleukin-6 receptor and its ligand interleukin-6 are opposite markers for survival and infiltration with mature myeloid cells in ovarian cancer. Oncoimmunology 2014, 3, e962397. [Google Scholar] [CrossRef]
  72. Bronte, V.; Brandau, S.; Chen, S.-H.; Colombo, M.P.; Frey, A.B.; Greten, T.F.; Mandruzzato, S.; Murray, P.J.; Ochoa, A.; Ostrand-Rosenberg, S.; et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat. Commun. 2016, 7, 12150. [Google Scholar] [CrossRef]
  73. Molon, B.; Ugel, S.; Del Pozzo, F.; Soldani, C.; Zilio, S.; Avella, D.; De Palma, A.; Mauri, P.; Monegal, A.; Rescigno, M.; et al. Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells. J. Exp. Med. 2011, 208, 1949–1962. [Google Scholar] [CrossRef] [PubMed]
  74. Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Bronte, V. Coordinated regulation of myeloid cells by tumours. Nat. Rev. Immunol. 2012, 12, 253–268. [Google Scholar] [CrossRef] [PubMed]
  75. Fujimura, T.; Mahnke, K.; Enk, A.H. Myeloid derived suppressor cells and their role in tolerance induction in cancer. J. Dermatol. Sci. 2010, 59, 1–6. [Google Scholar] [CrossRef] [PubMed]
  76. Huang, B.; Pan, P.-Y.; Li, Q.; Sato, A.I.; Levy, D.E.; Bromberg, J.; Divino, C.M.; Chen, S.-H. Gr-1+ CD115+ Immature Myeloid Suppressor Cells Mediate the Development of Tumor-Induced T Regulatory Cells and T-Cell Anergy in Tumor-Bearing Host. Cancer Res. 2006, 66, 1123–1131. [Google Scholar] [CrossRef]
  77. Hanson, E.M.; Clements, V.K.; Sinha, P.; Ilkovitch, D.; Ostrand-Rosenberg, S. Myeloid-Derived Suppressor Cells Down-Regulate L-Selectin Expression on CD4+ and CD8+ T Cells. J. Immunol. 2009, 183, 937–944. [Google Scholar] [CrossRef]
  78. Neumann, K.; Ostmann, A.; Breda, P.C.; Ochel, A.; Tacke, F.; Paust, H.-J.; Panzer, U.; Tiegs, G. The co-inhibitory molecule PD-L1 contributes to regulatory T cell-mediated protection in murine crescentic glomerulonephritis. Sci. Rep. 2019, 9, 2038. [Google Scholar] [CrossRef]
  79. Groth, C.; Hu, X.; Weber, R.; Fleming, V.; Altevogt, P.; Utikal, J.; Umansky, V. Immunosuppression mediated by myeloid-derived suppressor cells (MDSCs) during tumour progression. Br. J. Cancer 2019, 120, 16–25. [Google Scholar] [CrossRef]
  80. Juneja, V.R.; McGuire, K.A.; Manguso, R.T.; LaFleur, M.W.; Collins, N.; Haining, W.N.; Freeman, G.J.; Sharpe, A.H. PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity. J. Exp. Med. 2017, 214, 895–904. [Google Scholar] [CrossRef]
  81. Chambers, C.A.; Kuhns, M.S.; Egen, J.G.; Allison, J.P. CTLA-4-Mediated Inhibition in Regulation of T Cell Responses: Mechanisms and Manipulation in Tumor Immunotherapy. Annu. Rev. Immunol. 2001, 19, 565–594. [Google Scholar] [CrossRef]
  82. Togashi, Y.; Shitara, K.; Nishikawa, H. Regulatory T cells in cancer immunosuppression—Implications for anticancer therapy. Nat. Rev. Clin. Oncol. 2019, 16, 356–371. [Google Scholar] [CrossRef]
  83. Curiel, T.J.; Coukos, G.; Zou, L.; Alvarez, X.; Cheng, P.; Mottram, P.; Evdemon-Hogan, M.; Conejo-Garcia, J.R.; Zhang, L.; Burow, M.; et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat. Med. 2004, 10, 942–949. [Google Scholar] [CrossRef]
  84. Wolf, D.; Wolf, A.M.; Rumpold, H.; Fiegl, H.; Zeimet, A.G.; Muller-Holzner, E.; Deibl, M.; Gastl, G.; Gunsilius, E.; Marth, C. The Expression of the Regulatory T Cell-Specific Forkhead Box Transcription Factor FoxP3 Is Associated with Poor Prognosis in Ovarian Cancer. Clin. Cancer Res. 2005, 11, 8326–8331. [Google Scholar] [CrossRef] [PubMed]
  85. Tan, M.C.B.; Goedegebuure, P.S.; Belt, B.A.; Flaherty, B.; Sankpal, N.; Gillanders, W.E.; Eberlein, T.J.; Hsieh, C.-S.; Linehan, D.C. Disruption of CCR5-Dependent Homing of Regulatory T Cells Inhibits Tumor Growth in a Murine Model of Pancreatic Cancer. J. Immunol. 2009, 182, 1746–1755. [Google Scholar] [CrossRef] [PubMed]
  86. Fialová, A.; Partlová, S.; Sojka, L.; Hromádková, H.; Brtnický, T.; Fučíková, J.; Kocián, P.; Rob, L.; Bartůňková, J.; Špíšek, R. Dynamics of T-cell infiltration during the course of ovarian cancer: The gradual shift from a Th17 effector cell response to a predominant infiltration by regulatory T-cells. Int. J. Cancer 2013, 132, 1070–1079. [Google Scholar] [CrossRef] [PubMed]
  87. Jonuleit, H.; Schmitt, E.; Schuler, G.; Knop, J.; Enk, A.H. Induction of Interleukin 10—Producing, Nonproliferating Cd4+ T Cells with Regulatory Properties by Repetitive Stimulation with Allogeneic Immature Human Dendritic Cells. J. Exp. Med. 2000, 192, 1213–1222. [Google Scholar] [CrossRef]
  88. Facciabene, A.; Peng, X.; Hagemann, I.S.; Balint, K.; Barchetti, A.; Wang, L.-P.; Gimotty, P.A.; Gilks, C.B.; Lal, P.; Zhang, L.; et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and Treg cells. Nature 2011, 475, 226–230. [Google Scholar] [CrossRef]
  89. Barnett, B.; Kryczek, I.; Cheng, P.; Zou, W.; Curiel, T.J. Regulatory T Cells in Ovarian Cancer: Biology and Therapeutic Potential. Am. J. Reprod. Immunol. 2005, 54, 369–377. [Google Scholar] [CrossRef]
  90. Friedline, R.H.; Brown, D.S.; Nguyen, H.; Kornfeld, H.; Lee, J.; Zhang, Y.; Appleby, M.; Der, S.D.; Kang, J.; Chambers, C.A. CD4+ regulatory T cells require CTLA-4 for the maintenance of systemic tolerance. J. Exp. Med. 2009, 206, 421–434. [Google Scholar] [CrossRef]
  91. Deaglio, S.; Dwyer, K.M.; Gao, W.; Friedman, D.; Usheva, A.; Erat, A.; Chen, J.-F.; Enjyoji, K.; Linden, J.; Oukka, M.; et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J. Exp. Med. 2007, 204, 1257–1265. [Google Scholar] [CrossRef]
  92. Wang, R.; Wan, Q.; Kozhaya, L.; Fujii, H.; Unutmaz, D. Identification of a Regulatory T Cell Specific Cell Surface Molecule that Mediates Suppressive Signals and Induces Foxp3 Expression. PLoS ONE 2008, 3, e2705. [Google Scholar] [CrossRef]
  93. Mandapathil, M.; Szczepanski, M.J.; Szajnik, M.; Ren, J.; Lenzner, D.E.; Jackson, E.K.; Gorelik, E.; Lang, S.; Johnson, J.T.; Whiteside, T.L. Increased Ectonucleotidase Expression and Activity in Regulatory T Cells of Patients with Head and Neck Cancer. Clin. Cancer Res. 2009, 15, 6348–6357. [Google Scholar] [CrossRef]
  94. Stagg, J.; Divisekera, U.; Duret, H.; Sparwasser, T.; Teng, M.W.; Darcy, P.K.; Smyth, M.J. CD73-Deficient Mice Have Increased Antitumor Immunity and Are Resistant to Experimental Metastasis. Cancer Res. 2011, 71, 2892–2900. [Google Scholar] [CrossRef]
  95. Feng, L.; Sun, X.; Csizmadia, E.; Han, L.; Bian, S.; Murakami, T.; Wang, X.; Robson, S.C.; Wu, Y. Vascular CD39/ENTPD1 Directly Promotes Tumor Cell Growth by Scavenging Extracellular Adenosine Triphosphate. Neoplasia 2011, 13, 206–216. [Google Scholar] [CrossRef]
  96. Goding, J.W. Ecto-enzymes: Physiology meets pathology. J. Leukoc. Biol. 2000, 67, 285–311. [Google Scholar] [CrossRef]
  97. Scholzen, A.; Mittag, D.; Rogerson, S.J.; Cooke, B.M.; Plebanski, M. Plasmodium falciparum—Mediated Induction of Human CD25hiFoxp3hi CD4 T Cells Is Independent of Direct TCR Stimulation and Requires IL-2, IL-10 and TGFβ. PLoS Pathog. 2009, 5, e1000543. [Google Scholar] [CrossRef]
  98. Chen, X.; Subleski, J.J.; Hamano, R.; Howard, O.M.Z.; Wiltrout, R.H.; Oppenheim, J.J. Co-expression of TNFR2 and CD25 identifies more of the functional CD4+ FOXP3+ regulatory T cells in human peripheral blood. Eur. J. Immunol. 2010, 40, 1099–1106. [Google Scholar] [CrossRef]
  99. Mercer, F.; Kozhaya, L.; Unutmaz, D. Expression and Function of TNF and IL-1 Receptors on Human Regulatory T Cells. PLoS ONE 2010, 5, e8639. [Google Scholar] [CrossRef]
  100. Kulbe, H.; Chakravarty, P.; Leinster, D.A.; Charles, K.A.; Kwong, J.; Thompson, R.G.; Coward, J.I.; Schioppa, T.; Robinson, S.C.; Gallagher, W.M.; et al. A Dynamic Inflammatory Cytokine Network in the Human Ovarian Cancer Microenvironment. Cancer Res. 2012, 72, 66–75. [Google Scholar] [CrossRef]
  101. Govindaraj, C.; Scalzo-Inguanti, K.; Madondo, M.; Hallo, J.; Flanagan, K.; Quinn, M.; Plebanski, M. Impaired Th1 immunity in ovarian cancer patients is mediated by TNFR2+ Tregs within the tumor microenvironment. Clin. Immunol. 2013, 149, 97–110. [Google Scholar] [CrossRef]
  102. Santoiemma, P.P.; Powell, D.J., Jr. Tumor infiltrating lymphocytes in ovarian cancer. Cancer Biol. Ther. 2015, 16, 807–820. [Google Scholar] [CrossRef]
  103. Hara, H.; Kobayashi, A.; Yoshida, K.; Ohashi, M.; Ohnami, S.; Uchida, E.; Higashihara, E.; Yoshida, T.; Aoki, K. Local interferon-alpha gene therapy elicits systemic immunity in a syngeneic pancreatic cancer model in hamster. Cancer Sci. 2007, 98, 455–463. [Google Scholar] [CrossRef] [PubMed]
  104. Zhu, X.; Lang, J. Programmed Death-1 Pathway Blockade Produces a Synergistic Antitumor Effect: Combined Application in Ovarian Cancer. J. Gynecol. Oncol. 2017, 28, e64. [Google Scholar] [CrossRef] [PubMed]
  105. Wylie, B.; Macri, C.; Mintern, J.; Waithman, J. Dendritic Cells and Cancer: From Biology to Therapeutic Intervention. Cancers 2019, 11, 521. [Google Scholar] [CrossRef] [PubMed]
  106. Perez, C.R.; De Palma, M. Engineering dendritic cell vaccines to improve cancer immunotherapy. Nat. Commun. 2019, 10, 5408. [Google Scholar] [CrossRef]
  107. Thomas, S.; Prendergast, G.C. Cancer Vaccines: A Brief Overview. Methods Mol. Biol. 2016, 1403, 755–761. [Google Scholar] [CrossRef]
  108. Hoogstad-van Evert, J.S.; Bekkers, R.; Ottevanger, N.; Jansen, J.H.; Massuger, L.; Dolstra, H. Harnessing natural killer cells for the treatment of ovarian cancer. Gynecol. Oncol. 2020, 157, 810–816. [Google Scholar] [CrossRef]
  109. Morvan, M.; Lanier, L.L. NK cells and cancer: You can teach innate cells new tricks. Nat. Rev. Cancer 2016, 16, 7–19. [Google Scholar] [CrossRef]
  110. Krockenberger, M.; Dombrowski, Y.; Weidler, C.; Ossadnik, M.; Hönig, A.; Häusler, S.; Voigt, H.; Becker, J.C.; Leng, L.; Steinle, A.; et al. Macrophage Migration Inhibitory Factor Contributes to the Immune Escape of Ovarian Cancer by Down-Regulating NKG2D. J. Immunol. 2008, 180, 7338–7348. [Google Scholar] [CrossRef]
  111. Zhou, Y.; Xu, Y.; Chen, L.; Xu, B.; Wu, C.; Jiang, J. B7-H6 Expression Correlates with Cancer Progression and Patient’s Survival in Human Ovarian Cancer. Int. J. Clin. Exp. Pathol. 2015, 8, 9428–9433. [Google Scholar]
  112. Pesce, S.; Tabellini, G.; Cantoni, C.; Patrizi, O.; Coltrini, D.; Rampinelli, F.; Matta, J.; Vivier, E.; Moretta, A.; Parolini, S.; et al. B7-H6-mediated downregulation of NKp30 in NK cells contributes to ovarian carcinoma immune escape. Oncoimmunology 2015, 4, e1001224. [Google Scholar] [CrossRef]
  113. Wong, J.L.; Berk, E.; Edwards, R.P.; Kalinski, P. IL-18-Primed Helper NK Cells Collaborate with Dendritic Cells to Promote Recruitment of Effector CD8+ T Cells to the Tumor Microenvironment. Cancer Res. 2013, 73, 4653–4662. [Google Scholar] [CrossRef]
  114. Böttcher, J.P.; Bonavita, E.; Chakravarty, P.; Blees, H.; Cabeza-Cabrerizo, M.; Sammicheli, S.; Rogers, N.C.; Sahai, E.; Zelenay, S.; Sousa, C.R. NK Cells Stimulate Recruitment of cDC1 into the Tumor Microenvironment Promoting Cancer Immune Control. Cell 2018, 172, 1022–1037.e14. [Google Scholar] [CrossRef]
  115. Tan, D.S.; Agarwal, R.; Kaye, S.B. Mechanisms of Transcoelomic Metastasis in Ovarian Cancer. Lancet Oncol. 2006, 7, 925–934. [Google Scholar] [CrossRef]
  116. Eisenhauer, E. Real-world evidence in the treatment of ovarian cancer. Ann. Oncol. 2017, 28, viii61–viii65. [Google Scholar] [CrossRef]
  117. Kastelein, A.W.; Vos, L.M.C.; van Baal, J.O.A.M.; Koning, J.J.; Hira, V.V.V.; Nieuwland, R.; van Driel, W.J.; Uz, Z.; van Gulik, T.M.; van Rheenen, J.; et al. Poor perfusion of the microvasculature in peritoneal metastases of ovarian cancer. Clin. Exp. Metastasis 2020, 37, 293–304. [Google Scholar] [CrossRef]
  118. Langley, R.R.; Fidler, I.J. The seed and soil hypothesis revisited—The role of tumor-stroma interactions in metastasis to different organs. Int. J. Cancer 2011, 128 (Suppl. S8), 2527–2535. [Google Scholar] [CrossRef]
  119. Rickard, B.P.; Conrad, C.; Sorrin, A.J.; Ruhi, M.K.; Reader, J.C.; Huang, S.A.; Franco, W.; Scarcelli, G.; Polacheck, W.J.; Roque, D.M.; et al. Malignant Ascites in Ovarian Cancer: Cellular, Acellular, and Biophysical Determinants of Molecular Characteristics and Therapy Response. Cancers 2021, 13, 4318. [Google Scholar] [CrossRef]
  120. Motohara, T.; Masuda, K.; Morotti, M.; Zheng, Y.; El-Sahhar, S.; Chong, K.Y.; Wietek, N.; Alsaadi, A.; Carrami, E.M.; Hu, Z.; et al. An evolving story of the metastatic voyage of ovarian cancer cells: Cellular and molecular orchestration of the adipose-rich metastatic microenvironment. Oncogene 2019, 38, 2885–2898. [Google Scholar] [CrossRef]
  121. Barbolina, M.V. Molecular Mechanisms Regulating Organ-Specific Metastases in Epithelial Ovarian Carcinoma. Cancers 2018, 10, 444. [Google Scholar] [CrossRef]
  122. Gasparri, M.L.; Savone, D.; Besharat, R.A.; Farooqi, A.A.; Bellati, F.; Ruscito, I.; Panici, P.B.; Papadia, A. Circulating tumor cells as trigger to hematogenous spreads and potential biomarkers to predict the prognosis in ovarian cancer. Tumor Biol. 2016, 37, 71–75. [Google Scholar] [CrossRef]
  123. Yin, M.; Li, X.; Tan, S.; Zhou, H.J.; Ji, W.; Bellone, S.; Xu, X.; Zhang, H.; Santin, A.D.; Lou, G.; et al. Tumor-associated macrophages drive spheroid formation during early transcoelomic metastasis of ovarian cancer. J. Clin. Investig. 2016, 126, 4157–4173. [Google Scholar] [CrossRef] [PubMed]
  124. Zhang, Y.; Ouyang, D.; Chen, Y.H.; Xia, H. Peritoneal resident macrophages in tumor metastasis and immunotherapy. Front. Cell Dev. Biol. 2022, 10, 948952. [Google Scholar] [CrossRef] [PubMed]
  125. Wen, Z.; Liu, H.; Li, M.; Li, B.; Gao, W.; Shao, Q.; Fan, B.; Zhao, F.; Wang, Q.; Xie, Q.; et al. Increased metabolites of 5-lipoxygenase from hypoxic ovarian cancer cells promote tumor-associated macrophage infiltration. Oncogene 2014, 34, 1241–1252. [Google Scholar] [CrossRef] [PubMed]
  126. Gordon, I.O.; Freedman, R.S. Defective Antitumor Function of Monocyte-Derived Macrophages from Epithelial Ovarian Cancer Patients. Clin. Cancer Res. 2006, 12, 1515–1524. [Google Scholar] [CrossRef]
  127. Robinson-Smith, T.M.; Isaacsohn, I.; Mercer, C.A.; Zhou, M.; Van Rooijen, N.; Husseinzadeh, N.; McFarland-Mancini, M.M.; Drew, A.F. Macrophages Mediate Inflammation-Enhanced Metastasis of Ovarian Tumors in Mice. Cancer Res. 2007, 67, 5708–5716. [Google Scholar] [CrossRef]
  128. Hagemann, T.; Wilson, J.; Kulbe, H.; Li, N.F.; Leinster, D.A.; Charles, K.; Klemm, F.; Pukrop, T.; Binder, C.; Balkwill, F.R. Macrophages Induce Invasiveness of Epithelial Cancer Cells via NF-κB and JNK. J. Immunol. 2005, 175, 1197–1205. [Google Scholar] [CrossRef]
  129. Hagemann, T.; Robinson, S.C.; Thompson, R.G.; Charles, K.; Kulbe, H.; Balkwill, F.R. Ovarian cancer cell-derived migration inhibitory factor enhances tumor growth, progression, and angiogenesis. Mol. Cancer Ther. 2007, 6, 1993–2002. [Google Scholar] [CrossRef]
  130. Lane, D.; Matte, I.; Laplante, C.; Garde-Granger, P.; Carignan, A.; Bessette, P.; Rancourt, C.; Piché, A. CCL18 from ascites promotes ovarian cancer cell migration through proline-rich tyrosine kinase 2 signaling. Mol. Cancer 2016, 15, 58. [Google Scholar] [CrossRef]
  131. Wang, Q.; Tang, Y.; Yu, H.; Yin, Q.; Li, M.; Shi, L.; Zhang, W.; Li, D.; Li, L. CCL18 from tumor-cells promotes epithelial ovarian cancer metastasis via mTOR signaling pathway. Mol. Carcinog. 2016, 55, 1688–1699. [Google Scholar] [CrossRef]
  132. Neyen, C.; Plüddemann, A.; Mukhopadhyay, S.; Maniati, E.; Bossard, M.; Gordon, S.; Hagemann, T. Macrophage Scavenger Receptor A Promotes Tumor Progression in Murine Models of Ovarian and Pancreatic Cancer. J. Immunol. 2013, 190, 3798–3805. [Google Scholar] [CrossRef]
  133. Hagemann, T.; Lawrence, T.; McNeish, I.; Charles, K.A.; Kulbe, H.; Thompson, R.G.; Robinson, S.C.; Balkwill, F.R. “Re-educating” tumor-associated macrophages by targeting NF-κB. J. Exp. Med. 2008, 205, 1261–1268. [Google Scholar] [CrossRef]
  134. Browning, L.; Patel, M.R.; Horvath, E.B.; Tawara, K.; Jorcyk, C.L. IL-6 and ovarian cancer: Inflammatory cytokines in promotion of metastasis. Cancer Manag. Res. 2018, 10, 6685–6693. [Google Scholar] [CrossRef]
  135. Obermajer, N.; Muthuswamy, R.; Odunsi, K.; Edwards, R.P.; Kalinski, P. PGE2-Induced CXCL12 Production and CXCR4 Expression Controls the Accumulation of Human MDSCs in Ovarian Cancer Environment. Cancer Res. 2011, 71, 7463–7470. [Google Scholar] [CrossRef]
  136. Kajiyama, H.; Shibata, K.; Terauchi, M.; Ino, K.; Nawa, A.; Kikkawa, F. Involvement of SDF-1α/CXCR4 axis in the enhanced peritoneal metastasis of epithelial ovarian carcinoma. Int. J. Cancer 2008, 122, 91–99. [Google Scholar] [CrossRef]
  137. Wang, X.; Zhao, X.; Wang, K.; Wu, L.; Duan, T. Interaction of monocytes/macrophages with ovarian cancer cells promotes angiogenesisin vitro. Cancer Sci. 2013, 104, 516–523. [Google Scholar] [CrossRef]
  138. Zhang, W.; Tian, J.; Hao, Q. HMGB1 combining with tumor-associated macrophages enhanced lymphangiogenesis in human epithelial ovarian cancer. Tumor Biol. 2014, 35, 2175–2186. [Google Scholar] [CrossRef]
  139. Bruno, A.; Mortara, L.; Baci, D.; Noonan, D.M.; Albini, A. Myeloid Derived Suppressor Cells Interactions with Natural Killer Cells and Pro-Angiogenic Activities: Roles in Tumor Progression. Front. Immunol. 2019, 10, 771. [Google Scholar] [CrossRef]
  140. 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. 2020, 11, 1364. [Google Scholar] [CrossRef]
  141. Komura, N.; Mabuchi, S.; Shimura, K.; Yokoi, E.; Kozasa, K.; Kuroda, H.; Takahashi, R.; Sasano, T.; Kawano, M.; Matsumoto, Y.; et al. The role of myeloid-derived suppressor cells in increasing cancer stem-like cells and promoting PD-L1 expression in epithelial ovarian cancer. Cancer Immunol. Immunother. 2020, 69, 2477–2499. [Google Scholar] [CrossRef]
  142. Cui, T.X.; Kryczek, I.; Zhao, L.; Zhao, E.; Kuick, R.; Roh, M.H.; Vatan, L.; Szeliga, W.; Mao, Y.; Thomas, D.G.; et al. Myeloid-Derived Suppressor Cells Enhance Stemness of Cancer Cells by Inducing MicroRNA101 and Suppressing the Corepressor CtBP2. Immunity 2013, 39, 611–621. [Google Scholar] [CrossRef]
  143. Kassim, S.K.; El-Salahy, E.M.; Fayed, S.T.; Helal, S.A.; Helal, T.; Azzam, E.E.-D.; Khalifa, A. Vascular endothelial growth factor and interleukin-8 are associated with poor prognosis in epithelial ovarian cancer patients. Clin. Biochem. 2004, 37, 363–369. [Google Scholar] [CrossRef] [PubMed]
  144. Bouzin, C.; Brouet, A.; De Vriese, J.; DeWever, J.; Feron, O. Effects of Vascular Endothelial Growth Factor on the Lymphocyte-Endothelium Interactions: Identification of Caveolin-1 and Nitric Oxide as Control Points of Endothelial Cell Anergy1. J. Immunol. 2007, 178, 1505–1511. [Google Scholar] [CrossRef] [PubMed]
  145. Motz, G.T.; Santoro, S.P.; Wang, L.-P.; Garrabrant, T.; Lastra, R.R.; Hagemann, I.S.; Lal, P.; Feldman, M.D.; Benencia, F.; Coukos, G. Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors. Nat. Med. 2014, 20, 607–615. [Google Scholar] [CrossRef] [PubMed]
  146. Horikawa, N.; Abiko, K.; Matsumura, N.; Hamanishi, J.; Baba, T.; Yamaguchi, K.; Yoshioka, Y.; Koshiyama, M.; Konishi, I. Expression of Vascular Endothelial Growth Factor in Ovarian Cancer Inhibits Tumor Immunity through the Accumulation of Myeloid-Derived Suppressor Cells. Clin. Cancer Res. 2017, 23, 587–599. [Google Scholar] [CrossRef]
  147. Patel, S.; Fu, S.; Mastio, J.; Dominguez, G.A.; Purohit, A.; Kossenkov, A.; Lin, C.; Alicea-Torres, K.; Sehgal, M.; Nefedova, Y.; et al. Unique pattern of neutrophil migration and function during tumor progression. Nat. Immunol. 2018, 19, 1236–1247. [Google Scholar] [CrossRef]
  148. Fridlender, Z.G.; Sun, J.; Kim, S.; Kapoor, V.; Cheng, G.; Ling, L.; Worthen, G.S.; Albelda, S.M. Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell 2009, 16, 183–194. [Google Scholar] [CrossRef]
  149. Jablonska, J.; Leschner, S.; Westphal, K.; Lienenklaus, S.; Weiss, S. Neutrophils Responsive to Endogenous IFN-ß Regulate Tumor Angiogenesis and Growth in a Mouse Tumor Model. J. Clin. Invest. 2010, 120, 1151–1164. [Google Scholar] [CrossRef]
  150. Piccard, H.; Muschel, R.; Opdenakker, G. On the dual roles and polarized phenotypes of neutrophils in tumor development and progression. Crit. Rev. Oncol. 2012, 82, 296–309. [Google Scholar] [CrossRef]
  151. Singhal, S.; Bhojnagarwala, P.S.; O’Brien, S.; Moon, E.K.; Garfall, A.L.; Rao, A.S.; Quatromoni, J.G.; Stephen, T.L.; Litzky, L.; Deshpande, C.; et al. Origin and role of a subset of tumor-associated neutrophils with antigen-presenting cell features in early-stage human lung cancer. Cancer Cell 2016, 30, 120–135. [Google Scholar] [CrossRef]
  152. Cohen, C.A.; Shea, A.A.; Heffron, C.L.; Schmelz, E.M.; Roberts, P.C. The Parity-Associated Microenvironmental Niche in the Omental Fat Band Is Refractory to Ovarian Cancer Metastasis. Cancer Prev. Res. 2013, 6, 1182–1193. [Google Scholar] [CrossRef]
  153. Fruhman, G.J. Neutrophil Mobilization into Peritoneal Fluid. Blood 1960, 16, 1753–1761. [Google Scholar] [CrossRef]
  154. Buscher, K.; Wang, H.; Zhang, X.; Striewski, P.; Wirth, B.; Saggu, G.; Lütke-Enking, S.; Mayadas, T.N.; Ley, K.; Sorokin, L.; et al. Protection from septic peritonitis by rapid neutrophil recruitment through omental high endothelial venules. Nat. Commun. 2016, 7, 10828. [Google Scholar] [CrossRef]
  155. Lee, W.; Ko, S.Y.; Mohamed, M.S.; Kenny, H.A.; Lengyel, E.; Naora, H. Neutrophils facilitate ovarian cancer premetastatic niche formation in the omentum. J. Exp. Med. 2019, 216, 176–194. [Google Scholar] [CrossRef]
  156. Mishalian, I.; Bayuh, R.; Levy, L.; Zolotarov, L.; Michaeli, J.; Fridlender, Z.G. Tumor-associated neutrophils (TAN) develop pro-tumorigenic properties during tumor progression. Cancer Immunol. Immunother. 2013, 62, 1745–1756. [Google Scholar] [CrossRef]
  157. Sionov, R.V.; Fridlender, Z.G.; Granot, Z. The Multifaceted Roles Neutrophils Play in the Tumor Microenvironment. Cancer Microenviron. 2015, 8, 125–158. [Google Scholar] [CrossRef]
  158. Mantovani, A.; Cassatella, M.A.; Costantini, C.; Jaillon, S. Neutrophils in the activation and regulation of innate and adaptive immunity. Nat. Rev. Immunol. 2011, 11, 519–531. [Google Scholar] [CrossRef]
  159. Costa, S.; Bevilacqua, D.; Cassatella, M.A.; Scapini, P. Recent advances on the crosstalk between neutrophils and B or T lymphocytes. Immunology 2019, 156, 23–32. [Google Scholar] [CrossRef]
  160. Zhang, X.; Xu, W. Neutrophils diminish T-cell immunity to foster gastric cancer progression: The role of GM-CSF/PD-L1/PD-1 signaling pathway. Gut 2017, 66, 1878–1880. [Google Scholar] [CrossRef]
  161. Charles, K.A.; Kulbe, H.; Soper, R.; Escorcio-Correia, M.; Lawrence, T.; Schultheis, A.; Chakravarty, P.; Thompson, R.G.; Kollias, G.; Smyth, J.F.; et al. The tumor-promoting actions of TNF-α involve TNFR1 and IL-17 in ovarian cancer in mice and humans. J. Clin. Investig. 2009, 119, 3011–3023. [Google Scholar] [CrossRef]
  162. Emmons, T.R.; Giridharan, T.; Singel, K.L.; Khan, A.N.H.; Ricciuti, J.; Howard, K.; Toro, S.L.S.-D.; Debreceni, I.L.; Aarts, C.E.; Brouwer, M.C.; et al. Mechanisms Driving Neutrophil-Induced T-cell Immunoparalysis in Ovarian Cancer. Cancer Immunol. Res. 2021, 9, 790–810. [Google Scholar] [CrossRef]
  163. Cheng, J.-C.; Klausen, C.; Leung, P.C.K. Hydrogen Peroxide Mediates EGF-Induced Down-Regulation of E-Cadherin Expression via p38 MAPK and Snail in Human Ovarian Cancer Cells. Mol. Endocrinol. 2010, 24, 1569–1580. [Google Scholar] [CrossRef] [PubMed]
  164. Takemoto, A.; Okitaka, M.; Takagi, S.; Takami, M.; Sato, S.; Nishio, M.; Okumura, S.; Fujita, N. A critical role of platelet TGF-β release in podoplanin-mediated tumour invasion and metastasis. Sci. Rep. 2017, 7, 42186. [Google Scholar] [CrossRef] [PubMed]
  165. Guo, Y.; Cui, W.; Pei, Y.; Xu, D. Platelets promote invasion and induce epithelial to mesenchymal transition in ovarian cancer cells by TGF-β signaling pathway. Gynecol. Oncol. 2019, 153, 639–650. [Google Scholar] [CrossRef] [PubMed]
  166. Seo, E.J.; Kwon, Y.W.; Jang, I.H.; Kim, D.K.; Lee, S.I.; Choi, E.J.; Kim, K.-H.; Suh, D.-S.; Lee, J.H.; Choi, K.U.; et al. Autotaxin Regulates Maintenance of Ovarian Cancer Stem Cells through Lysophosphatidic Acid-Mediated Autocrine Mechanism. Stem Cells 2016, 34, 551–564. [Google Scholar] [CrossRef] [PubMed]
  167. Reinartz, S.; Lieber, S.; Pesek, J.; Brandt, D.T.; Asafova, A.; Finkernagel, F.; Watzer, B.; Nockher, W.A.; Nist, A.; Stiewe, T.; et al. Cell type-selective pathways and clinical associations of lysophosphatidic acid biosynthesis and signaling in the ovarian cancer microenvironment. Mol. Oncol. 2019, 13, 185–201. [Google Scholar] [CrossRef]
  168. Mills, G.B.; Moolenaar, W.H. The emerging role of lysophosphatidic acid in cancer. Nat. Rev. Cancer 2003, 3, 582–591. [Google Scholar] [CrossRef]
  169. Lin, R.J.; Afshar-Kharghan, V.; Schafer, A.I. Paraneoplastic thrombocytosis: The secrets of tumor self-promotion. Blood 2014, 124, 184–187. [Google Scholar] [CrossRef]
  170. Eder, A.M.; Sasagawa, T.; Mao, M.; Aoki, J.; Mills, G.B. Constitutive and lysophosphatidic acid (LPA)-induced LPA production: Role of phospholipase D and phospholipase A2. Clin. Cancer Res. 2000, 6, 2482–2491. [Google Scholar]
  171. Safaee, M.; Clark, A.J.; Ivan, M.E.; Oh, M.C.; Bloch, O.; Sun, M.Z.; Oh, T.; Parsa, A.T. CD97 is a multifunctional leukocyte receptor with distinct roles in human cancers. Int. J. Oncol. 2013, 43, 1343–1350. [Google Scholar] [CrossRef]
  172. Labelle, M.; Begum, S.; Hynes, R.O. Platelets guide the formation of early metastatic niches. Proc. Natl. Acad. Sci. USA 2014, 111, E3053–E3061. [Google Scholar] [CrossRef]
  173. Orellana, R.; Kato, S.; Erices, R.; Bravo, M.L.; Gonzalez, P.; Oliva, B.; Cubillos, S.; Valdivia, A.; Ibañez, C.; Brañes, J.; et al. Platelets enhance tissue factor protein and metastasis initiating cell markers, and act as chemoattractants increasing the migration of ovarian cancer cells. BMC Cancer 2015, 15, 290. [Google Scholar] [CrossRef]
  174. Haemmerle, M.; Taylor, M.L.; Gutschner, T.; Pradeep, S.; Cho, M.S.; Sheng, J.; Lyons, Y.M.; Nagaraja, A.S.; Dood, R.L.; Wen, Y.; et al. Platelets reduce anoikis and promote metastasis by activating YAP1 signaling. Nat. Commun. 2017, 8, 310. [Google Scholar] [CrossRef]
  175. Rodriguez-Martinez, A.; Simon-Saez, I.; Perales, S.; Garrido-Navas, C.; Russo, A.; de Miguel-Perez, D.; Puche-Sanz, I.; Alaminos, C.; Ceron, J.; Lorente, J.A.; et al. Exchange of cellular components between platelets and tumor cells: Impact on tumor cells behavior. Theranostics 2022, 12, 2150–2161. [Google Scholar] [CrossRef]
  176. Egan, K.; Cooke, N.; Kenny, D. Living in shear: Platelets protect cancer cells from shear induced damage. Clin. Exp. Metastasis 2014, 31, 697–704. [Google Scholar] [CrossRef]
  177. Palumbo, J.S.; Talmage, K.E.; Massari, J.V.; La Jeunesse, C.M.; Flick, M.J.; Kombrinck, K.W.; Jirousková, M.; Degen, J.L. Platelets and fibrin(ogen) increase metastatic potential by impeding natural killer cell-mediated elimination of tumor cells. Blood 2005, 105, 178–185. [Google Scholar] [CrossRef]
  178. Placke, T.; Salih, H.R.; Kopp, H.-G. GITR Ligand Provided by Thrombopoietic Cells Inhibits NK Cell Antitumor Activity. J. Immunol. 2012, 189, 154–160. [Google Scholar] [CrossRef]
  179. Kopp, H.-G.; Placke, T.; Salih, H.R. Platelet-Derived Transforming Growth Factor-β Down-Regulates NKG2D Thereby Inhibiting Natural Killer Cell Antitumor Reactivity. Cancer Res. 2009, 69, 7775–7783. [Google Scholar] [CrossRef]
  180. Jorch, S.K.; Kubes, P. An emerging role for neutrophil extracellular traps in noninfectious disease. Nat. Med. 2017, 23, 279–287. [Google Scholar] [CrossRef]
  181. Abu Saadeh, F.; Norris, L.; O’toole, S.; Gleeson, N. Venous thromboembolism in ovarian cancer: Incidence, risk factors and impact on survival. Eur. J. Obstet. Gynecol. Reprod. Biol. 2013, 170, 214–218. [Google Scholar] [CrossRef]
  182. Fotopoulou, C.; Dubois, A.; Karavas, A.N.; Trappe, R.; Aminossadati, B.; Schmalfeldt, B.; Pfisterer, J.; Sehouli, J. Incidence of Venous Thromboembolism in Patients with Ovarian Cancer Undergoing Platinum/Paclitaxel-Containing First-Line Chemotherapy: An Exploratory Analysis by the Arbeitsgemeinschaft Gynaekologische Onkologie Ovarian Cancer Study Group. J. Clin. Oncol. 2008, 26, 2683–2689. [Google Scholar] [CrossRef]
  183. Weeks, K.S.; Herbach, E.; McDonald, M.; Charlton, M.; Schweizer, M.L. Meta-Analysis of VTE Risk: Ovarian Cancer Patients by Stage, Histology, Cytoreduction, and Ascites at Diagnosis. Obstet. Gynecol. Int. 2020, 2020, 2374716. [Google Scholar] [CrossRef] [PubMed]
  184. Buergy, D.; Wenz, F.; Groden, C.; Brockmann, M.A. Tumor-platelet interaction in solid tumors. Int. J. Cancer 2012, 130, 2747–2760. [Google Scholar] [CrossRef] [PubMed]
  185. Kato, Y.; Kaneko, M.; Sata, M.; Fujita, N.; Tsuruo, T.; Osawa, M. Enhanced Expression of Aggrus (T1alpha/Podoplanin), a Platelet-Aggregation-Inducing Factor in Lung Squamous Cell Carcinoma. Tumor Biol. 2005, 26, 195–200. [Google Scholar] [CrossRef] [PubMed]
  186. Mitrugno, A.; Williams, D.; Kerrigan, S.W.; Moran, N. A Novel and Essential Role for Fc g RIIa in Cancer Cell—Induced Platelet Activation. Blood 2016, 123, 249–261. [Google Scholar] [CrossRef] [PubMed]
  187. Cho, M.S.; Noh, K.; Haemmerle, M.; Li, D.; Park, H.; Hu, Q.; Hisamatsu, T.; Mitamura, T.; Mak, S.L.C.; Kunapuli, S.; et al. Role of ADP receptors on platelets in the growth of ovarian cancer. Blood 2017, 130, 1235–1242. [Google Scholar] [CrossRef]
  188. Sakurai, M.; Matsumoto, K.; Gosho, M.; Sakata, A.; Hosokawa, Y.; Tenjimbayashi, Y.; Katoh, T.; Shikama, A.; Komiya, H.; Michikami, H.; et al. Expression of Tissue Factor in Epithelial Ovarian Carcinoma Is Involved in the Development of Venous Thromboembolism. Int. J. Gynecol. Cancer 2017, 27, 37–43. [Google Scholar] [CrossRef]
  189. Yokota, N.; Koizume, S.; Miyagi, E.; Hirahara, F.; Nakamura, Y.; Kikuchi, K.; Ruf, W.; Sakuma, Y.; Tsuchiya, E.; Miyagi, Y. Self-production of tissue factor-coagulation factor VII complex by ovarian cancer cells. Br. J. Cancer 2009, 101, 2023–2029. [Google Scholar] [CrossRef]
  190. Mackman, N. Role of Tissue Factor in Hemostasis, Thrombosis, and Vascular Development. Arter. Thromb. Vasc. Biol. 2004, 24, 1015–1022. [Google Scholar] [CrossRef]
  191. Cohen, J.G.; Prendergast, E.; Geddings, J.E.; Walts, A.E.; Agadjanian, H.; Hisada, Y.; Karlan, B.Y.; Mackman, N.; Walsh, C.S. Evaluation of venous thrombosis and tissue factor in epithelial ovarian cancer. Gynecol. Oncol. 2017, 146, 146–152. [Google Scholar] [CrossRef]
  192. Zhong, Y.-C.; Zhang, T.; Di, W.; Li, W.-P. Thrombin promotes epithelial ovarian cancer cell invasion by inducing epithelial-mesenchymal transition. J. Gynecol. Oncol. 2013, 24, 265–272. [Google Scholar] [CrossRef]
  193. Alexander, E.T.; Minton, A.R.; Peters, M.C.; van Ryn, J.; Gilmour, S.K. Thrombin inhibition and cisplatin block tumor progression in ovarian cancer by alleviating the immunosuppressive microenvironment. Oncotarget 2016, 7, 85291–85305. [Google Scholar] [CrossRef]
  194. De Candia, E. Mechanisms of platelet activation by thrombin: A short history. Thromb. Res. 2012, 129, 250–256. [Google Scholar] [CrossRef]
  195. Möhle, R.; Green, D.; Moore, M.A.S.; Nachman, R.L.; Rafii, S. Constitutive production and thrombin-induced release of vascular endothelial growth factor by human megakaryocytes and platelets. Proc. Natl. Acad. Sci. USA 1997, 94, 663–668. [Google Scholar] [CrossRef]
  196. Holmes, C.E.; Levis, J.E.; Ornstein, D.L. Activated platelets enhance ovarian cancer cell invasion in a cellular model of metastasis. Clin. Exp. Metastasis 2009, 26, 653–661. [Google Scholar] [CrossRef]
  197. Yuan, L.; Liu, X. Platelets are associated with xenograft tumor growth and the clinical malignancy of ovarian cancer through an angiogenesis-dependent mechanism. Mol. Med. Rep. 2015, 11, 2449–2458. [Google Scholar] [CrossRef]
  198. Erices, R.; Cubillos, S.; Aravena, R.; Santoro, F.; Marquez, M.; Orellana, R.; Ramírez, C.; González, P.; Fuenzalida, P.; Bravo, M.L.; et al. Diabetic concentrations of metformin inhibit platelet-mediated ovarian cancer cell progression. Oncotarget 2017, 8, 20865–20880. [Google Scholar] [CrossRef]
  199. Haemmerle, M.; Bottsford-Miller, J.; Pradeep, S.; Taylor, M.L.; Choi, H.-J.; Hansen, J.M.; Dalton, H.J.; Stone, R.L.; Cho, M.S.; Nick, A.M.; et al. FAK regulates platelet extravasation and tumor growth after antiangiogenic therapy withdrawal. J. Clin. Investig. 2016, 126, 1885–1896. [Google Scholar] [CrossRef]
  200. Kawamura, K.; Komohara, Y.; Takaishi, K.; Katabuchi, H.; Takeya, M. Detection of M2 macrophages and colony-stimulating factor 1 expression in serous and mucinous ovarian epithelial tumors. Pathol. Int. 2009, 59, 300–305. [Google Scholar] [CrossRef]
  201. Wang, X.; Deavers, M.; Patenia, R.; Bassett, R.L.; Mueller, P.; Ma, Q.; Wang, E.; Freedman, R.S. Monocyte/macrophage and T-cell infiltrates in peritoneum of patients with ovarian cancer or benign pelvic disease. J. Transl. Med. 2006, 4, 30. [Google Scholar] [CrossRef]
  202. Lan, C.; Huang, X.; Lin, S.; Huang, H.; Cai, Q.; Wan, T.; Lu, J.; Liu, J. Expression of M2-Polarized Macrophages is Associated with Poor Prognosis for Advanced Epithelial Ovarian Cancer. Technol. Cancer Res. Treat. 2013, 12, 259–267. [Google Scholar] [CrossRef]
  203. Yafei, Z.; Jun, G.; Guolan, G. Correlation between Macrophage Infiltration and Prognosis of Ovarian Cancer—A Preliminary Study. Biomed. Res. 2016, 27, 305–312. [Google Scholar]
  204. Yuan, X.; Zhang, J.; Li, D.; Mao, Y.; Mo, F.; Du, W.; Ma, X. Prognostic significance of tumor-associated macrophages in ovarian cancer: A meta-analysis. Gynecol. Oncol. 2017, 147, 181–187. [Google Scholar] [CrossRef] [PubMed]
  205. Zhang, M.; He, Y.; Sun, X.; Li, Q.; Wang, W.; Zhao, A.; Di, W. A high M1/M2 ratio of tumor-associated macrophages is associated with extended survival in ovarian cancer patients. J. Ovarian Res. 2014, 7, 19. [Google Scholar] [CrossRef] [PubMed]
  206. Le Page, C.; Marineau, A.; Bonza, P.K.; Rahimi, K.; Cyr, L.; Labouba, I.; Madore, J.; Delvoye, N.; Mes-Masson, A.-M.; Provencher, D.M.; et al. BTN3A2 Expression in Epithelial Ovarian Cancer Is Associated with Higher Tumor Infiltrating T Cells and a Better Prognosis. PLoS ONE 2012, 7, e38541. [Google Scholar] [CrossRef]
  207. Tan, Q.; Liu, H.; Xu, J.; Mo, Y.; Dai, F. Integrated analysis of tumor-associated macrophage infiltration and prognosis in ovarian cancer. Aging 2021, 13, 23210–23232. [Google Scholar] [CrossRef]
  208. Sue-A-Quan, R.; Patel, P.G.; Shakfa, N.; Nyi, M.-P.N.; Afriyie-Asante, A.; Kang, E.Y.; Köbel, M.; Koti, M. Prognostic significance of T cells, PD-L1 immune checkpoint and tumour associated macrophages in clear cell carcinoma of the ovary. Gynecol. Oncol. 2021, 162, 421–430. [Google Scholar] [CrossRef]
  209. Macciò, A.; Gramignano, G.; Cherchi, M.C.; Tanca, L.; Melis, L.; Madeddu, C. Role of M1-polarized tumor-associated macrophages in the prognosis of advanced ovarian cancer patients. Sci. Rep. 2020, 10, 6096. [Google Scholar] [CrossRef]
  210. Badmann, S.; Heublein, S.; Mayr, D.; Reischer, A.; Liao, Y.; Kolben, T.; Beyer, S.; Hester, A.; Zeder-Goess, C.; Burges, A.; et al. M2 Macrophages Infiltrating Epithelial Ovarian Cancer Express MDR1: A Feature That May Account for the Poor Prognosis. Cells 2020, 9, 1224. [Google Scholar] [CrossRef]
  211. Hensler, M.; Kasikova, L.; Fiser, K.; Rakova, J.; Skapa, P.; Laco, J.; Lanickova, T.; Pecen, L.; Truxova, I.; Vosahlikova, S.; et al. M2-like macrophages dictate clinically relevant immunosuppression in metastatic ovarian cancer. J. Immunother. Cancer 2020, 8, e000979. [Google Scholar] [CrossRef]
  212. Truxova, I.; Kasikova, L.; Hensler, M.; Skapa, P.; Laco, J.; Pecen, L.; Belicova, L.; Praznovec, I.; Halaska, M.J.; Brtnicky, T.; et al. Mature dendritic cells correlate with favorable immune infiltrate and improved prognosis in ovarian carcinoma patients. J. Immunother. Cancer 2018, 6, 139. [Google Scholar] [CrossRef]
  213. Mastelic-Gavillet, B.; Sarivalasis, A.; Lozano, L.E.; Wyss, T.; Inoges, S.; de Vries, I.J.M.; Dartiguenave, F.; Jichlinski, P.; Derrè, L.; Coukos, G.; et al. Quantitative and qualitative impairments in dendritic cell subsets of patients with ovarian or prostate cancer. Eur. J. Cancer 2020, 135, 173–182. [Google Scholar] [CrossRef]
  214. Labidi-Galy, S.I.; Treilleux, I.; Goddard-Leon, S.; Combes, J.-D.; Blay, J.-Y.; Ray-Coquard, I.; Caux, C.; Bendriss-Vermare, N. Plasmacytoid dendritic cells infiltrating ovarian cancer are associated with poor prognosis. Oncoimmunology 2012, 1, 380–382. [Google Scholar] [CrossRef]
  215. Zhang, X.; He, T.; Li, Y.; Chen, L.; Liu, H.; Wu, Y.; Guo, H. Dendritic Cell Vaccines in Ovarian Cancer. Front. Immunol. 2021, 11, 613773. [Google Scholar] [CrossRef]
  216. Salmon, H.; Idoyaga, J.; Rahman, A.; Leboeuf, M.; Remark, R.; Jordan, S.; Casanova-Acebes, M.; Khudoynazarova, M.; Agudo, J.; Tung, N.; et al. Expansion and Activation of CD103+ Dendritic Cell Progenitors at the Tumor Site Enhances Tumor Responses to Therapeutic PD-L1 and BRAF Inhibition. Immunity 2016, 44, 924–938. [Google Scholar] [CrossRef]
  217. Scarlett, U.K.; Cubillos-Ruiz, J.R.; Nesbeth, Y.C.; Martinez, D.G.; Engle, X.; Gewirtz, A.T.; Ahonen, C.L.; Conejo-Garcia, J.R. In Situ Stimulation of CD40 and Toll-like Receptor 3 Transforms Ovarian Cancer—Infiltrating Dendritic Cells from Immunosuppressive to Immunostimulatory Cells. Cancer Res. 2009, 69, 7329–7337. [Google Scholar] [CrossRef]
  218. Zhang, Z.; Huang, J.; Zhang, C.; Yang, H.; Qiu, H.; Li, J.; Liu, Y.; Qin, L.; Wang, L.; Hao, S.; et al. Infiltration of dendritic cells and T lymphocytes predicts favorable outcome in epithelial ovarian cancer. Cancer Gene Ther. 2015, 22, 198–206. [Google Scholar] [CrossRef]
  219. Baert, T.; Vankerckhoven, A.; Riva, M.; Van Hoylandt, A.; Thirion, G.; Holger, G.; Mathivet, T.; Vergote, I.; Coosemans, A. Myeloid Derived Suppressor Cells: Key Drivers of Immunosuppression in Ovarian Cancer. Front. Immunol. 2019, 10, 1273. [Google Scholar] [CrossRef]
  220. Lee, J.-M.; Botesteanu, D.-A.; Tomita, Y.; Yuno, A.; Lee, M.-J.; Kohn, E.C.; Annunziata, C.M.; Matulonis, U.; Macdonald, L.A.; Nair, J.R.; et al. Patients with BRCA mutated ovarian cancer may have fewer circulating MDSC and more peripheral CD8+ T cells compared with women with BRCA wild-type disease during the early disease course. Oncol. Lett. 2019, 18, 3914–3924. [Google Scholar] [CrossRef]
  221. Li, L.; Wang, L.; Li, J.; Fan, Z.; Yang, L.; Zhang, Z.; Zhang, C.; Yue, D.; Qin, G.; Zhang, T.; et al. Metformin-Induced Reduction of CD39 and CD73 Blocks Myeloid-Derived Suppressor Cell Activity in Patients with Ovarian Cancer. Cancer Res. 2018, 78, 1779–1791. [Google Scholar] [CrossRef]
  222. Okła, K.; Czerwonka, A.; Wawruszak, A.; Bobiński, M.; Bilska, M.; Tarkowski, R.; Bednarek, W.; Wertel, I.; Kotarski, J. Clinical Relevance and Immunosuppressive Pattern of Circulating and Infiltrating Subsets of Myeloid-Derived Suppressor Cells (MDSCs) in Epithelial Ovarian Cancer. Front. Immunol. 2019, 10, 691. [Google Scholar] [CrossRef]
  223. Wu, L.; Deng, Z.; Peng, Y.; Han, L.; Liu, J.; Wang, L.; Li, B.; Zhao, J.; Jiao, S.; Wei, H. Ascites-derived IL-6 and IL-10 synergistically expand CD14+ HLA-DR-/low myeloid-derived suppressor cells in ovarian cancer patients. Oncotarget 2017, 8, 76843–76856. [Google Scholar] [CrossRef]
  224. Santegoets, S.J.A.M.; de Groot, A.F.; Dijkgraaf, E.M.; Carnaz Simões, A.M.; van der Noord, V.E.; van Ham, J.J.; Welters, M.J.P.; Kroep, J.R.; van der Burg, S.H. The blood mMDSC to DC ratio is a sensitive and easy to assess independent predictive factor for epithelial ovarian cancer survival. OncoImmunology 2018, 7, e1465166. [Google Scholar] [CrossRef]
  225. Okła, K.; Rajtak, A.; Czerwonka, A.; Bobiński, M.; Wawruszak, A.; Tarkowski, R.; Bednarek, W.; Szumiło, J.; Kotarski, J. Accumulation of blood-circulating PD-L1-expressing M-MDSCs and monocytes/macrophages in pretreatment ovarian cancer patients is associated with soluble PD-L1. J. Transl. Med. 2020, 18, 220. [Google Scholar] [CrossRef] [PubMed]
  226. Taki, M.; Abiko, K.; Baba, T.; Hamanishi, J.; Yamaguchi, K.; Murakami, R.; Yamanoi, K.; Horikawa, N.; Hosoe, Y.; Nakamura, E.; et al. Snail promotes ovarian cancer progression by recruiting myeloid-derived suppressor cells via CXCR2 ligand upregulation. Nat. Commun. 2018, 9, 1685. [Google Scholar] [CrossRef]
  227. Sato, E.; Olson, S.H.; Ahn, J.; Bundy, B.; Nishikawa, H.; Qian, F.; Jungbluth, A.A.; Frosina, D.; Gnjatic, S.; Ambrosone, C.; et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc. Natl. Acad. Sci. USA 2005, 102, 18538–18543. [Google Scholar] [CrossRef] [PubMed]
  228. James, F.R.; Jiminez-Linan, M.; Alsop, J.; Mack, M.; Song, H.; Brenton, J.D.; Pharoah, P.D.P.; Ali, H.R. Association between tumour infiltrating lymphocytes, histotype and clinical outcome in epithelial ovarian cancer. BMC Cancer 2017, 17, 657. [Google Scholar] [CrossRef] [PubMed]
  229. Bösmüller, H.-C.; Wagner, P.; Peper, J.K.; Schuster, H.; Pham, D.L.; Greif, K.; Beschorner, C.; Rammensee, H.-G.; Stevanović, S.; Fend, F.; et al. Combined Immunoscore of CD103 and CD3 Identifies Long-Term Survivors in High-Grade Serous Ovarian Cancer. Int. J. Gynecol. Cancer 2016, 26, 671–679. [Google Scholar] [CrossRef]
  230. Hamanishi, J.; Mandai, M.; Iwasaki, M.; Okazaki, T.; Tanaka, Y.; Yamaguchi, K.; Higuchi, T.; Yagi, H.; Takakura, K.; Minato, N.; et al. Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proc. Natl. Acad. Sci. USA 2007, 104, 3360–3365. [Google Scholar] [CrossRef]
  231. Clarke, B.; Tinker, A.V.; Lee, C.H.; Subramanian, S.; van de Rijn, M.; Turbin, D.; Kalloger, S.; Han, G.; Ceballos, K.; Cadungog, M.G.; et al. Intraepithelial T cells and prognosis in ovarian carcinoma: Novel associations with stage, tumor type, and BRCA1 loss. Mod. Pathol. 2009, 22, 393–402. [Google Scholar] [CrossRef]
  232. Hao, J.; Yu, H.; Zhang, T.; An, R.; Xue, Y. Prognostic impact of tumor-infiltrating lymphocytes in high grade serous ovarian cancer: A systematic review and meta-analysis. Ther. Adv. Med. Oncol. 2020, 12, 1758835920967241. [Google Scholar] [CrossRef]
  233. Leffers, N.; Gooden, M.J.M.; de Jong, R.A.; Hoogeboom, B.-N.; ten Hoor, K.A.; Hollema, H.; Boezen, H.M.; van der Zee, A.G.J.; Daemen, T.; Nijman, H.W. Prognostic significance of tumor-infiltrating T-lymphocytes in primary and metastatic lesions of advanced stage ovarian cancer. Cancer Immunol. Immunother. 2009, 58, 449–459. [Google Scholar] [CrossRef]
  234. Hermans, C.; Anz, D.; Engel, J.; Kirchner, T.; Endres, S.; Mayr, D. Analysis of FoxP3+ T-Regulatory Cells and CD8+ T-Cells in Ovarian Carcinoma: Location and Tumor Infiltration Patterns Are Key Prognostic Markers. PLoS ONE 2014, 9, e111757. [Google Scholar] [CrossRef]
  235. Zhou, J.; Li, X.; Wu, X.; Zhang, T.; Zhu, Q.; Wang, X.; Wang, H.; Wang, K.; Lin, Y.; Wang, X. Exosomes Released from Tumor-Associated Macrophages Transfer miRNAs That Induce a Treg/Th17 Cell Imbalance in Epithelial Ovarian Cancer. Cancer Immunol. Res. 2018, 6, 1578–1592. [Google Scholar] [CrossRef]
  236. Milne, K.; Köbel, M.; Kalloger, S.E.; Barnes, R.O.; Gao, D.; Gilks, C.B.; Watson, P.; Nelson, B.H. Systematic Analysis of Immune Infiltrates in High-Grade Serous Ovarian Cancer Reveals CD20, FoxP3 and TIA-1 as Positive Prognostic Factors. PLoS ONE 2009, 4, e6412. [Google Scholar] [CrossRef]
  237. Shang, B.; Liu, Y.; Jiang, S.-J.; Liu, Y. Prognostic value of tumor-infiltrating FoxP3+ regulatory T cells in cancers: A systematic review and meta-analysis. Sci. Rep. 2015, 5, 15179. [Google Scholar] [CrossRef] [PubMed]
  238. Santoiemma, P.P.; Reyes, C.; Wang, L.-P.; McLane, M.W.; Feldman, M.D.; Tanyi, J.L.; Powell, D.J. Systematic evaluation of multiple immune markers reveals prognostic factors in ovarian cancer. Gynecol. Oncol. 2016, 143, 120–127. [Google Scholar] [CrossRef] [PubMed]
  239. Nielsen, J.S.; Sahota, R.A.; Milne, K.; Kost, S.E.; Nesslinger, N.J.; Watson, P.H.; Nelson, B.H. CD20+ Tumor-Infiltrating Lymphocytes Have an Atypical CD27− Memory Phenotype and Together with CD8+ T Cells Promote Favorable Prognosis in Ovarian Cancer. Clin. Cancer Res. 2012, 18, 3281–3292. [Google Scholar] [CrossRef]
  240. Kroeger, D.R.; Milne, K.; Nelson, B.H. Tumor-Infiltrating Plasma Cells Are Associated with Tertiary Lymphoid Structures, Cytolytic T-Cell Responses, and Superior Prognosis in Ovarian Cancer. Clin. Cancer Res. 2016, 22, 3005–3015. [Google Scholar] [CrossRef] [PubMed]
  241. Lundgren, S.; Berntsson, J.; Nodin, B.; Micke, P.; Jirström, K. Prognostic impact of tumour-associated B cells and plasma cells in epithelial ovarian cancer. J. Ovarian Res. 2016, 9, 21. [Google Scholar] [CrossRef]
  242. Alwosaibai, K.; Aalmri, S.; Mashhour, M.; Ghandorah, S.; Alshangiti, A.; Azam, F.; Selwi, W.; Gharaibeh, L.; Alatawi, Y.; Alruwaii, Z.; et al. PD-L1 is highly expressed in ovarian cancer and associated with cancer stem cells populations expressing CD44 and other stem cell markers. BMC Cancer 2023, 23, 13. [Google Scholar] [CrossRef]
  243. Buderath, P.; Mairinger, F.; Mairinger, E.; Böhm, K.; Mach, P.; Schmid, K.W.; Kimmig, R.; Kasimir-Bauer, S.; Bankfalvi, A.; Westerwick, D.; et al. Prognostic significance of PD-1 and PD-L1 positive tumor-infiltrating immune cells in ovarian carcinoma. Int. J. Gynecol. Cancer 2019, 29, 1389–1395. [Google Scholar] [CrossRef]
  244. Wang, L. Prognostic effect of programmed death-ligand 1 (PD-L1) in ovarian cancer: A systematic review, meta-analysis and bioinformatics study. J. Ovarian Res. 2019, 12, 37. [Google Scholar] [CrossRef]
  245. Henriksen, J.R.; Donskov, F.; Waldstrøm, M.; Jakobsen, A.; Hjortkjaer, M.; Petersen, C.B.; Steffensen, K.D. Favorable prognostic impact of Natural Killer cells and T cells in high-grade serous ovarian carcinoma. Acta Oncol. 2020, 59, 652–659. [Google Scholar] [CrossRef]
  246. Zhu, Y.; Zhang, Z.; Jiang, Z.; Liu, Y.; Zhou, J. CD38 Predicts Favorable Prognosis by Enhancing Immune Infiltration and Antitumor Immunity in the Epithelial Ovarian Cancer Microenvironment. Front. Genet. 2020, 11, 369. [Google Scholar] [CrossRef]
  247. Fucikova, J.; Rakova, J.; Hensler, M.; Kasikova, L.; Belicova, L.; Hladikova, K.; Truxova, I.; Skapa, P.; Laco, J.; Pecen, L.; et al. TIM-3 Dictates Functional Orientation of the Immune Infiltrate in Ovarian Cancer. Clin. Cancer Res. 2019, 25, 4820–4831. [Google Scholar] [CrossRef]
  248. Li, J.; Wang, J.; Chen, R.; Bai, Y.; Lu, X. The prognostic value of tumor-infiltrating T lymphocytes in ovarian cancer. Oncotarget 2017, 8, 15621–15631. [Google Scholar] [CrossRef]
  249. Wang, Q.; Lou, W.; Di, W.; Wu, X. Prognostic value of tumor PD-L1 expression combined with CD8+ tumor infiltrating lymphocytes in high grade serous ovarian cancer. Int. Immunopharmacol. 2017, 52, 7–14. [Google Scholar] [CrossRef]
  250. Montfort, A.; Pearce, O.; Maniati, E.; Vincent, B.G.; Bixby, L.; Böhm, S.; Dowe, T.; Wilkes, E.H.; Chakravarty, P.; Thompson, R.; et al. A Strong B-Cell Response Is Part of the Immune Landscape in Human High-Grade Serous Ovarian Metastases. Clin. Cancer Res. 2017, 23, 250–262. [Google Scholar] [CrossRef] [PubMed]
  251. Darb-Esfahani, S.; Kunze, C.A.; Kulbe, H.; Sehouli, J.; Wienert, S.; Lindner, J.; Budczies, J.; Bockmayr, M.; Dietel, M.; Denkert, C.; et al. Prognostic impact of programmed cell death-1 (PD-1) and PD-ligand 1 (PD-L1) expression in cancer cells and tumor-infiltrating lymphocytes in ovarian high grade serous carcinoma. Oncotarget 2016, 7, 1486–1499. [Google Scholar] [CrossRef]
  252. Knutson, K.L.; Maurer, M.J.; Preston, C.C.; Moysich, K.B.; Goergen, K.; Hawthorne, K.M.; Cunningham, J.M.; Odunsi, K.; Hartmann, L.C.; Kalli, K.R.; et al. Regulatory T cells, inherited variation, and clinical outcome in epithelial ovarian cancer. Cancer Immunol. Immunother. 2015, 64, 1495–1504. [Google Scholar] [CrossRef]
  253. Webb, J.R.; Milne, K.; Watson, P.; Deleeuw, R.J.; Nelson, B.H. Tumor-Infiltrating Lymphocytes Expressing the Tissue Resident Memory Marker CD103 Are Associated with Increased Survival in High-Grade Serous Ovarian Cancer. Clin. Cancer Res. 2014, 20, 434–444. [Google Scholar] [CrossRef]
  254. Iglesia, M.D.; Vincent, B.G.; Parker, J.S.; Hoadley, K.A.; Carey, L.A.; Perou, C.M.; Serody, J.S. Prognostic B-cell Signatures Using mRNA-Seq in Patients with Subtype-Specific Breast and Ovarian Cancer. Clin. Cancer Res. 2014, 20, 3818–3829. [Google Scholar] [CrossRef] [PubMed]
  255. Bachmayr-Heyda, A.; Aust, S.; Heinze, G.; Polterauer, S.; Grimm, C.; Braicu, E.I.; Sehouli, J.; Lambrechts, S.; Vergote, I.; Mahner, S.; et al. Prognostic impact of tumor infiltrating CD8+ T cells in association with cell proliferation in ovarian cancer patients—A study of the OVCAD consortium. BMC Cancer 2013, 13, 422. [Google Scholar] [CrossRef]
  256. Hwang, W.-T.; Adams, S.F.; Tahirovic, E.; Hagemann, I.S.; Coukos, G. Prognostic significance of tumor-infiltrating T cells in ovarian cancer: A meta-analysis. Gynecol. Oncol. 2012, 124, 192–198. [Google Scholar] [CrossRef]
  257. Callahan, M.J.; Nagymanyoki, Z.; Bonome, T.; Johnson, M.E.; Litkouhi, B.; Sullivan, E.H.; Hirsch, M.S.; Matulonis, U.A.; Liu, J.; Birrer, M.J.; et al. Increased HLA-DMB Expression in the Tumor Epithelium Is Associated with Increased CTL Infiltration and Improved Prognosis in Advanced-Stage Serous Ovarian Cancer. Clin. Cancer Res. 2010, 14, 7667–7673. [Google Scholar] [CrossRef] [PubMed]
  258. Barnett, J.C.; Bean, S.M.; Whitaker, R.S.; Kondoh, E.; Baba, T.; Fujii, S.; Marks, J.R.; Dressman, H.K.; Murphy, S.K.; Berchuck, A. Ovarian cancer tumor infiltrating T-regulatory (Treg) cells are associated with a metastatic phenotype. Gynecol. Oncol. 2010, 116, 556–562. [Google Scholar] [CrossRef] [PubMed]
  259. Stumpf, M.P.H.; Hasenburg, A.; Riener, M.-O.; Jutting, U.; Wang, C.; Shen, Y.; Orlowskavolk, M.; Fisch, P.; Wang, Z.; Gitsch, G.; et al. Intraepithelial CD8-positive T lymphocytes predict survival for patients with serous stage III ovarian carcinomas: Relevance of clonal selection of T lymphocytes. Br. J. Cancer 2009, 101, 1513–1521. [Google Scholar] [CrossRef]
  260. Hoogstad-van Evert, J.S.; Maas, R.J.; Van Der Meer, J.; Cany, J.; Van Der Steen, S.; Jansen, J.H.; Miller, J.S.; Bekkers, R.; Hobo, W.; Massuger, L.; et al. Peritoneal NK cells are responsive to IL-15 and percentages are correlated with outcome in advanced ovarian cancer patients. Oncotarget 2018, 9, 34810–34820. [Google Scholar] [CrossRef]
  261. Henriksen, J.R.; Nederby, L.; Donskov, F.; Waldstrøm, M.; Adimi, P.; Jakobsen, A.; Steffensen, K.D. Blood natural killer cells during treatment in recurrent ovarian cancer. Acta Oncol. 2020, 59, 1365–1373. [Google Scholar] [CrossRef]
  262. Komura, N.; Mabuchi, S.; Yokoi, E.; Kozasa, K.; Kuroda, H.; Sasano, T.; Matsumoto, Y.; Kimura, T. Comparison of clinical utility between neutrophil count and neutrophil-lymphocyte ratio in patients with ovarian cancer: A single institutional experience and a literature review. Int. J. Clin. Oncol. 2018, 23, 104–113. [Google Scholar] [CrossRef]
  263. Bishara, S.; Griffin, M.; Cargill, A.; Bali, A.; Gore, M.; Kaye, S.; Shepherd, J.; Van Trappen, P. Pre-treatment white blood cell subtypes as prognostic indicators in ovarian cancer. Eur. J. Obstet. Gynecol. Reprod. Biol. 2008, 138, 71–75. [Google Scholar] [CrossRef]
  264. Cho, H.; Hur, H.W.; Kim, S.W.; Kim, S.H.; Kim, J.H.; Kim, Y.T.; Lee, K. Pre-treatment neutrophil to lymphocyte ratio is elevated in epithelial ovarian cancer and predicts survival after treatment. Cancer Immunol. Immunother. 2009, 58, 15–23. [Google Scholar] [CrossRef]
  265. Badora-Rybicka, A.; Nowara, E.; Starzyczny-Słota, D. Neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio before chemotherapy as potential prognostic factors in patients with newly diagnosed epithelial ovarian cancer. ESMO Open 2016, 1, e000039. [Google Scholar] [CrossRef] [PubMed]
  266. Wang, Y.-Q.; Jin, C.; Zheng, H.-M.; Zhou, K.; Shi, B.-B.; Zhang, Q.; Zheng, F.-Y.; Lin, F. A novel prognostic inflammation score predicts outcomes in patients with ovarian cancer. Clin. Chim. Acta 2016, 456, 163–169. [Google Scholar] [CrossRef]
  267. Van Nguyen, J.M.; Ferguson, S.E.; Bernardini, M.Q.; May, T.; LaFramboise, S.; Hogen, L.; Bouchard-Fortier, G. Preoperative neutrophil-to-lymphocyte ratio predicts 30 day postoperative morbidity and survival after primary surgery for ovarian cancer. Int. J. Gynecol. Cancer 2020, 30, 1378–1383. [Google Scholar] [CrossRef]
  268. Nakamura, K.; Nagasaka, T.; Nishida, T.; Haruma, T.; Ogawa, C.; Kusumoto, T.; Seki, N.; Hiramatsu, Y. Neutrophil to lymphocyte ratio in the pre-treatment phase of final-line chemotherapy predicts the outcome of patients with recurrent ovarian cancer. Oncol. Lett. 2016, 11, 3975–3981. [Google Scholar] [CrossRef]
  269. Yildirim, M.; Cendek, B.D.; Avsar, A.F. Differentiation between benign and malignant ovarian masses in the preoperative period using neutrophil-to-lymphocyte and platelet-to-lymphocyte ratios. Mol. Clin. Oncol. 2015, 3, 317–321. [Google Scholar] [CrossRef]
  270. Yildirim, M.A.; Seckin, K.D.; Togrul, C.; Baser, E.; Karsli, M.F.; Gungor, T.; Gulerman, H.C. Roles of Neutrophil/Lymphocyte and Platelet/Lymphocyte Ratios in the Early Diagnosis of Malignant Ovarian Masses. Asian Pac. J. Cancer Prev. 2014, 15, 6881–6885. [Google Scholar] [CrossRef]
  271. Abu-Shawer, O.; Hirmas, N.; Alhouri, A.; Massad, A.; Alsibai, B.; Sultan, H.; Hammo, H.; Souleiman, M.; Shebli, Y.; Al-Hussaini, M. Hematologic markers of distant metastases and poor prognosis in gynecological cancers. BMC Cancer 2019, 19, 141. [Google Scholar] [CrossRef] [PubMed]
  272. Forget, P.; Khalifa, C.; Defour, J.-P.; Latinne, D.; Van Pel, M.-C.; De Kock, M. What is the normal value of the neutrophil-to-lymphocyte ratio? BMC Res. Notes 2017, 10, 12. [Google Scholar] [CrossRef]
  273. Kim, J.G.; Kim, S.I.; Song, S.H.; Gu, J.-Y.; Lee, M.; Kim, H.K. Diagnostic and prognostic role of circulating neutrophil extracellular trap markers and prekallikrein in patients with high-grade serous ovarian cancer. Front. Oncol. 2022, 12, 992056. [Google Scholar] [CrossRef]
  274. Muqaku, B.; Pils, D.; Mader, J.C.; Aust, S.; Mangold, A.; Muqaku, L.; Slany, A.; Del Favero, G.; Gerner, C. Neutrophil Extracellular Trap Formation Correlates with Favorable Overall Survival in High Grade Ovarian Cancer. Cancers 2020, 12, 505. [Google Scholar] [CrossRef]
  275. Marchetti, C.; D’indinosante, M.; Bottoni, C.; Di Ilio, C.; Di Berardino, S.; Costantini, B.; Minucci, A.; Vertechy, L.; Scambia, G.; Fagotti, A. NLR and BRCA mutational status in patients with high grade serous advanced ovarian cancer. Sci. Rep. 2021, 11, 11125. [Google Scholar] [CrossRef]
  276. He, Y.; Li, T.; Liu, J.; Ou, Q.; Zhou, J. Early onset neutropenia: A useful predictor of chemosensitivity and favorable prognosis in patients with serous ovarian cancer. BMC Cancer 2020, 20, 116. [Google Scholar] [CrossRef]
  277. Singel, K.; Grzankowski, K.S.; Khan, A.N.M.N.H.; Grimm, M.J.; D’auria, A.C.; Morrell, K.; Eng, K.H.; Hylander, B.; Mayor, P.C.; Emmons, T.R.; et al. Mitochondrial DNA in the tumour microenvironment activates neutrophils and is associated with worse outcomes in patients with advanced epithelial ovarian cancer. Br. J. Cancer 2019, 120, 207–217. [Google Scholar] [CrossRef]
  278. Posabella, A.; Köhn, P.; Lalos, A.; Wilhelm, A.; Mechera, R.; Soysal, S.; Muenst, S.; Güth, U.; Stadlmann, S.; Terracciano, L.; et al. High density of CD66b in primary high-grade ovarian cancer independently predicts response to chemotherapy. J. Cancer Res. Clin. Oncol. 2020, 146, 127–136. [Google Scholar] [CrossRef]
  279. Chen, G.; Zhu, L.; Yang, Y.; Long, Y.; Li, X.; Wang, Y. Prognostic Role of Neutrophil to Lymphocyte Ratio in Ovarian Cancer: A Meta-Analysis. Technol. Cancer Res. Treat. 2018, 17, 1533033818791500. [Google Scholar] [CrossRef]
  280. Huang, Q.-T.; Zhou, L.; Zeng, W.-J.; Ma, Q.-Q.; Wang, W.; Zhong, M.; Yu, Y.-H. Prognostic Significance of Neutrophil-To-Lymphocyte Ratio in Ovarian Cancer: A Systematic Review and Meta-Analysis of Observational Studies. Cell. Physiol. Biochem. 2017, 41, 2411–2418. [Google Scholar] [CrossRef]
  281. Allensworth, S.; Langstraat, C.; Martin, J.; Lemens, M.; McGree, M.; Weaver, A.; Dowdy, S.; Podratz, K.; Bakkum-Gamez, J. Evaluating the prognostic significance of preoperative thrombocytosis in epithelial ovarian cancer. Gynecol. Oncol. 2013, 130, 499–504. [Google Scholar] [CrossRef] [PubMed]
  282. Ye, Q.; Cheng, J.; Ye, M.; Liu, D.; Zhang, Y. Association of pretreatment thrombocytosis with prognosis in ovarian cancer: A systematic review and meta-analysis. J. Gynecol. Oncol. 2019, 30, e5. [Google Scholar] [CrossRef] [PubMed]
  283. Feng, Z.; Wen, H.; Bi, R.; Duan, Y.; Yang, W.; Wu, X. Thrombocytosis and hyperfibrinogenemia are predictive factors of clinical outcomes in high-grade serous ovarian cancer patients. BMC Cancer 2016, 16, 43. [Google Scholar] [CrossRef] [PubMed]
  284. Lee, M.; Kim, S.W.; Nam, E.J.; Yim, G.W.; Kim, S.; Kim, Y.T. The impact of pretreatment thrombocytosis and persistent thrombocytosis after adjuvant chemotherapy in patients with advanced epithelial ovarian cancer. Gynecol. Oncol. 2011, 122, 238–241. [Google Scholar] [CrossRef]
  285. Asher, V.; Lee, J.; Innamaa, A.; Bali, A. Preoperative platelet lymphocyte ratio as an independent prognostic marker in ovarian cancer. Clin. Transl. Oncol. 2011, 13, 499–503. [Google Scholar] [CrossRef]
  286. Ceran, M.U.; Tasdemir, U.; Colak, E.; Güngör, T. Can complete blood count inflammatory parameters in epithelial ovarian cancer contribute to prognosis?—A survival analysis. J. Ovarian Res. 2019, 12, 16. [Google Scholar] [CrossRef]
  287. Winarno, G.N.A.; Pasaribu, M.; Susanto, H.; Nisa, A.S.; Harsono, A.B.; Yuseran, H.; Suardi, D.; Trianasari, N. The Platelet to Lymphocyte and Neutrophil to Lymphocyte Ratios in Predicting Response to Platinum-Based Chemotherapy for Epithelial Ovarian Cancer. Asian Pac. J. Cancer Prev. 2021, 22, 1561–1566. [Google Scholar] [CrossRef]
  288. Eo, W.K.; Chang, H.J.; Kwon, S.H.; Koh, S.B.; Kim, Y.O.; Ji, Y.I.; Kim, H.-B.; Lee, J.Y.; Suh, D.S.; Kim, K.H.; et al. The Lymphocyte-Monocyte Ratio Predicts Patient Survival and Aggressiveness of Ovarian Cancer. J. Cancer 2016, 7, 289–296. [Google Scholar] [CrossRef]
  289. Polat, M.; Senol, T.; Ozkaya, E.; Pakay, G.O.; Cikman, M.S.; Konukcu, B.; Ozten, M.A.; Karateke, A. Neutrophil to lymphocyte and platelet to lymphocyte ratios increase in ovarian tumors in the presence of frank stromal invasion. Clin. Transl. Oncol. 2016, 18, 457–463. [Google Scholar] [CrossRef]
  290. Wang, N.; Li, C.; Yang, Y.; Guan, Y.; Wang, F.; Wang, Y.; Zhao, W. The Use of Platelet/Lymphocyte Ratio and Cancer Antigen 125 Combined with Magnetic Resonance Diffusion-Weighted Imaging in Diagnosis of Recurrent Ovarian Cancer and Neuropathic Pain. World Neurosurg. 2021, 149, 502–510. [Google Scholar] [CrossRef]
  291. Kim, M.-S.; Baek, S.H.; Noh, J.J.; Shim, J.I.; Kang, J.H.; Jeong, S.Y.; Choi, C.H.; Kim, T.-J.; Lee, J.-W.; Lee, Y.-Y. Role of reactive thrombocytosis after primary cytoreductive surgery in advanced ovarian cancer. Front. Oncol. 2022, 12, 926878. [Google Scholar] [CrossRef]
  292. Canzler, U.; Lück, H.-J.; Neuser, P.; Sehouli, J.; Burges, A.; Harter, P.; Schmalfeldt, B.; Aminossadati, B.; Mahner, S.; Kommoss, S.; et al. Prognostic role of thrombocytosis in recurrent ovarian cancer: A pooled analysis of the AGO Study Group. Arch. Gynecol. Obstet. 2020, 301, 1267–1274. [Google Scholar] [CrossRef]
  293. Ma, X.; Wang, Y.; Sheng, H.; Tian, W.; Qi, Z.; Teng, F.; Xue, F. Prognostic significance of thrombocytosis, platelet parameters and aggregation rates in epithelial ovarian cancer. J. Obstet. Gynaecol. Res. 2014, 40, 178–183. [Google Scholar] [CrossRef] [PubMed]
  294. Kandalaft, L.E.; Motz, G.T.; Duraiswamy, J.; Coukos, G. Tumor immune surveillance and ovarian cancer. Cancer Metastasis Rev. 2011, 30, 141–151. [Google Scholar] [CrossRef] [PubMed]
  295. Yigit, R.; Massuger, L.F.; Figdor, C.; Torensma, R. Ovarian cancer creates a suppressive microenvironment to escape immune elimination. Gynecol. Oncol. 2010, 117, 366–372. [Google Scholar] [CrossRef]
  296. Vesely, M.D.; Kershaw, M.H.; Schreiber, R.D.; Smyth, M.J. Natural Innate and Adaptive Immunity to Cancer. Annu. Rev. Immunol. 2011, 29, 235–271. [Google Scholar] [CrossRef]
  297. Rosen, D.G.; Wang, L.; Atkinson, J.N.; Yu, Y.; Lu, K.H.; Diamandis, E.P.; Hellstrom, I.; Mok, S.C.; Liu, J.; Bast, R.C. Potential markers that complement expression of CA125 in epithelial ovarian cancer. Gynecol. Oncol. 2005, 99, 267–277. [Google Scholar] [CrossRef]
  298. Terry, K.L.; Sluss, P.M.; Skates, S.J.; Mok, S.C.; Ye, B.; Vitonis, A.F.; Cramer, D.W. Blood and Urine Markers for Ovarian Cancer: A Comprehensive Review. Dis. Markers 2004, 20, 53–70. [Google Scholar] [CrossRef]
Figure 1. In the tumor microenvironment (TME), cells have specific components and roles. These cells actively interact with one another through the molecules they secrete (cytokines and chemokines) and receptors they express forming an evolving microenvironment. Cell components in TME can be divided into cancer cells, immune cells, and platelets. Different cell components can localize at distinct locations along the spectrum from anti-tumor (tumor infiltrating lymphocytes (TIL), mature dendritic cells (mature DC), M1 tumor-associated macrophages (M1 TAMs), natural killers cells (NK cells), and N1 tumor-associated-neutrophils (N1 TANs)) to pro-tumor effect (T regulatory cells (Treg cells), platelets, immature dendritic cells (immature DC), M2 tumor-associated macrophages (M2 TAMs), N2 tumor-associated-neutrophils (N2 TANs), and myeloid-derived suppressor cells (MDSC)), and the same group of cells may also be re-polarized depending on the signals in the TME. A single tumor site’s ability to advance or retreat is influenced by TME’s intricate cellular and the molecular regulatory network as a whole.
Figure 1. In the tumor microenvironment (TME), cells have specific components and roles. These cells actively interact with one another through the molecules they secrete (cytokines and chemokines) and receptors they express forming an evolving microenvironment. Cell components in TME can be divided into cancer cells, immune cells, and platelets. Different cell components can localize at distinct locations along the spectrum from anti-tumor (tumor infiltrating lymphocytes (TIL), mature dendritic cells (mature DC), M1 tumor-associated macrophages (M1 TAMs), natural killers cells (NK cells), and N1 tumor-associated-neutrophils (N1 TANs)) to pro-tumor effect (T regulatory cells (Treg cells), platelets, immature dendritic cells (immature DC), M2 tumor-associated macrophages (M2 TAMs), N2 tumor-associated-neutrophils (N2 TANs), and myeloid-derived suppressor cells (MDSC)), and the same group of cells may also be re-polarized depending on the signals in the TME. A single tumor site’s ability to advance or retreat is influenced by TME’s intricate cellular and the molecular regulatory network as a whole.
Ijms 24 09279 g001
Table 5. Characteristics of studies on NK cells and ovarian cancer prognostic significance.
Table 5. Characteristics of studies on NK cells and ovarian cancer prognostic significance.
Author/YearHistologyStageMethodImpactFindings
Webb et al., 2014 [253]EOCI-IIIIHC, FCBeneficialHigh density of CD103+ NK cells correlated with favorable disease outcome.
Henriksen et al., 2020 [261]EOCn.aFCDetrimentalA decreased level of NK cell count in recurrent metastasis during chemotherapy is associated with unfavorable prognostic impact.
Henriksen et al., 2020 [245]HGSOCI-IVIHCBeneficialHigh density of CD57+ NK cells correlated with favorable OS.
Krockenberger et al., 2008 [110]EOCn.aIHC, IFDetrimentalHigh density of MIF inhibits NK cells and correlated with poor prognosis
Hoogstad-van Evert et al., 2018 [260]HGSOCIII-IVFCBeneficialHigh density of CD103+ NK cells correlated with favorable disease outcome.
EOC various subtypes; HGSOC, high-grade serous ovarian cancer; IHC, immunohistochemistry; FC, flow cytometry; IF, immunofluorescence; n.a, not available.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pankowska, K.A.; Będkowska, G.E.; Chociej-Stypułkowska, J.; Rusak, M.; Dąbrowska, M.; Osada, J. Crosstalk of Immune Cells and Platelets in an Ovarian Cancer Microenvironment and Their Prognostic Significance. Int. J. Mol. Sci. 2023, 24, 9279. https://doi.org/10.3390/ijms24119279

AMA Style

Pankowska KA, Będkowska GE, Chociej-Stypułkowska J, Rusak M, Dąbrowska M, Osada J. Crosstalk of Immune Cells and Platelets in an Ovarian Cancer Microenvironment and Their Prognostic Significance. International Journal of Molecular Sciences. 2023; 24(11):9279. https://doi.org/10.3390/ijms24119279

Chicago/Turabian Style

Pankowska, Katarzyna Aneta, Grażyna Ewa Będkowska, Joanna Chociej-Stypułkowska, Małgorzata Rusak, Milena Dąbrowska, and Joanna Osada. 2023. "Crosstalk of Immune Cells and Platelets in an Ovarian Cancer Microenvironment and Their Prognostic Significance" International Journal of Molecular Sciences 24, no. 11: 9279. https://doi.org/10.3390/ijms24119279

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

Article Metrics

Back to TopTop