Next Article in Journal
Green Tea Catechins: Nature’s Way of Preventing and Treating Cancer
Next Article in Special Issue
Triaging HPV-Positive Cervical Samples with p16 and Ki-67 Dual Stained Cytology within an Organized Screening Program—A Prospective Observational Study from Western Norway
Previous Article in Journal
PSP-GNM: Predicting Protein Stability Changes upon Point Mutations with a Gaussian Network Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immune Tumor Microenvironment in Ovarian Cancer Ascites

by
Diana Luísa Almeida-Nunes
1,2,
Ana Mendes-Frias
3,4,
Ricardo Silvestre
3,4,
Ricardo Jorge Dinis-Oliveira
2,5,6,7 and
Sara Ricardo
1,2,8,*
1
Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S), Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), 4200-135 Porto, Portugal
2
TOXRUN—Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, 4585-116 Gandra, Portugal
3
Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
4
ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
5
UCIBIO-REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal
6
Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, 4099-002 Porto, Portugal
7
MTG Research and Development Lab, 4200-604 Porto, Portugal
8
Faculty of Medicine, University of Porto (FMUP), 4099-002 Porto, Portugal
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(18), 10692; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms231810692
Submission received: 10 August 2022 / Revised: 26 August 2022 / Accepted: 6 September 2022 / Published: 14 September 2022
(This article belongs to the Special Issue Advances in Gynecological Cancers 2.0)

Abstract

:
Ovarian cancer (OC) has a specific type of metastasis, via transcoelomic, and most of the patients are diagnosed at advanced stages with multiple tumors spread within the peritoneal cavity. The role of Malignant Ascites (MA) is to serve as a transporter of tumor cells from the primary location to the peritoneal wall or to the surface of the peritoneal organs. MA comprise cellular components with tumor and non-tumor cells and acellular components, creating a unique microenvironment capable of modifying the tumor behavior. These microenvironment factors influence tumor cell proliferation, progression, chemoresistance, and immune evasion, suggesting that MA play an active role in OC progression. Tumor cells induce a complex immune suppression that neutralizes antitumor immunity, leading to disease progression and treatment failure, provoking a tumor-promoting environment. In this review, we will focus on the High-Grade Serous Carcinoma (HGSC) microenvironment with special attention to the tumor microenvironment immunology.

1. Introduction

Ovarian cancer (OC) is one of the most frequent gynecologic cancers, being the seventh most-common malignant tumor and the eighth cause of cancer death in women around the world [1]. This malignancy is characterized by the rapid growth and spread of multiple intraperitoneal tumors [2]. The World Health Organization classifies ovarian tumors as epithelial, which represents almost 90% of the cases, germ cell (3%), and sex cord-stromal (2%) origins [3]. Epithelial Ovarian Carcinoma (EOC) has five main subtypes based on its histopathology, immune, and molecular profile: high-grade serous carcinoma (HGSC) comprising 70% of total EOC, low-grade serous carcinoma (LGSC), endometrioid carcinoma (EC), clear cell carcinoma (CCC), and mucinous carcinoma (MC) (Figure 1) [3]. Despite the differences between the subtypes, these tumors are treated as a single entity, relying on cytoreductive surgery and platinum-taxane combination chemotherapy. The response rate to first-line therapy is about 80–90%, but most patients relapse and develop chemotherapy resistance, leading to a 5-year survival rate below 25% [4,5]. Serous carcinomas are tumors with a more aggressive behavior, e.g., HGSC spreads rapidly throughout the pelvis, while endometrioid and mucinous carcinomas are typically confined to the ovary being mostly low-grade lesions [6]. The histologic types of OC are highly associated with the clinical response. Late-stage serous and clear cell carcinomas, both have similar 5-year survival rates (20–30%); however, their response to first-line chemotherapy is different: Serous tumors are highly responsive, while clear cell tumors are especially resistant [7]. However, the survival rate of patients with serous tumors relies on chemoresistance upon recurrence which is very frequent.
OC has a lower prevalence in contrast to breast cancer but is three times more lethal due to the lack of specific symptoms and the absence of good screening tools, resulting in diagnosis at advanced stages [8]. OC patients present high recurrence rates, mostly due to the incomplete resection of tumors in cytoreductive surgery [9]. In addition, although most patients are platinum-responsive to first-line therapy, most of them develop chemoresistant recurrences [9]. These facts contribute to worse clinical outcomes; approximately 15% of OC patients die from the disease within the first year, and only 25% survive over 5 years from the date of diagnosis [10]. These different outcomes trigger the need for a more profound exploration of tumor and host characteristics. The anti-tumoral immune response in OC patients could help to disclose these different outcomes. OC is an immune reactive malignancy in which tumor cells establish a complex multilayered immune suppression network that effectively neutralizes most attempts to increase antitumor immunity [10]. This leads to a lack of responses to immune-based treatments in these patients.
In this review, we present a summary of the immunological populations and their role in OC, including the subsets of adaptive and innate immune response, the mechanisms of immune escape, and the effect of key cytokines, chemokines, and growth factors in the tumor immune microenvironment (TIME).

2. High-Grade Serous Carcinoma

HGSC is the most common ovarian malignancy, representing approximately 70% of ovarian carcinoma [3]. It is recognized to be of epithelial origin, arising from the fallopian tube epithelium through a series of precursor lesions that target the secretory cell [7,11]. This tumor type is generally recognized by the lack of architecture and sheets of malignant cells with enlarged and dysmorphic nuclei, and the high frequency of the tumor suppressor p53 protein (TP53) mutation [12]. The TP53 mutation occurs exclusively in secretory cells that show strong expression of this gene and evidence of DNA damage but are not proliferative. The progression to a serous tubal intraepithelial carcinoma involves the gain of nuclear pleomorphism and mitoses and the loss of cell polarity [7,8].
The median age of patients diagnosed with this type of tumor is 56 years (ranging from 45 to 65 years) [3]. The available screening techniques are unsuccessful for early detection and just a minority of OC patients are diagnosed with tumors limited to one ovary (<5%). Typically, HGSC presents at diagnosis with bilateral ovarian involvement and diffuse and extensive peritoneal carcinomatosis, particularly with omental involvement, characterized by the presence of malignant ascites (MA) [13,14]. The spread of multiple intra-abdominal tumors is often associated with signs of intestinal obstruction, including nausea, vomiting, persistent bloating, and abdominal pain. Ultrasound, magnetic resonance imaging, and computerized tomography have no specified role in preoperative tumor staging. Thus, laparotomy and the surgical exploration of the abdominal cavity are used as standard approaches to defining the tumor stage [3]. Although specific biomarkers for OC are not defined, women with deleterious germline BRCA1 or BRCA2 mutations have a 30–70% risk of developing HGSC [3,15].
Despite these features at diagnosis, HGSC patients are usually highly sensitive to first-line therapy (platinum-taxane) and only a small subgroup (<10%) is refractory to this chemotherapy [16]. Nevertheless, even after a period of initial clinical remission, most patients suffer from a relapse of the disease (approximately 75%) in 3 years that are typically incurable [16,17]. Some of these patients are refractory to first-line therapy due to acquired chemoresistance, while the majority suffer remission with the same treatment. This fact shows a potential mechanism of therapy failure different from intrinsic or acquired resistance [18].

3. Malignant Ascites—Tumor Microenvironment in Ovarian Cancer

Ascites can occur in different diseases, such as cirrhosis, pancreatitis, nephritis, heart failure, and cancer [19]. MA are the pathological accumulation of fluid in the peritoneal cavity that can be found in many neoplasms of peritoneal organs (ovarian, endometrial, pancreatic, gastric, colorectal, and liver) [20]. This inflammatory condition is a consequence of a disruption in the balance of fluid production and reabsorption [21], being triggered by increased vascular permeability, peritoneal lymphatic obstruction, and high levels of fluid production [22]. The presence of MA is often indicative of tumor cells in the peritoneal cavity or peritoneal carcinomatosis [23].
Several studies associate OC growth and metastization with its intrinsic tumor microenvironment [24,25,26]. The late-stage diagnosis (stage III/IV) implies the presence of metastasis in the pelvic and peritoneal cavities which are coupled with the accumulation of a large volume of peritoneal fluid [27]. Unveiling the mechanisms behind this liquid metastatic microenvironment is crucial for the improvement of new attempts to abrogate the tumor peritoneal spread and improve OC management.
The role of MAs is to facilitate the spread of tumor cells to other pelvic and peritoneal organs, serving as a transporter [28]. This transcoelomic dissemination process is fundamental for the adhesion of cancer cells to the omentum and serous membranes lining the peritoneal organs, leading to the implantation of metastatic tumors in the peritoneal cavity, not being typical the invasion of the lamina propria [29]. OC cells disseminate into peritoneal sites such as the hepatic hile, omentum, spleen, and uterus, among others. MA comprise not only tumor cells, but also many other non-tumor cells (Figure 2), which create a unique microenvironment capable of modifying the neoplastic properties of tumor cells [30]. This exudative fluid is composed of a cellular and an acellular fraction. The cellular counterpart is composed of highly tumorigenic cancer cells [31,32], innate and adaptive immune cells [10,22], and other non-tumor cells. The acellular fraction contains tumor-promoting soluble factors (angiogenic and growth factors), bioactive lipids, cytokines, and extracellular vesicles [33]. All of these factors contribute to tumor cell proliferation, progression, chemoresistance, and immune evasion [34], suggesting that MA play an active role in the development and progression of OC, especially in HGSC [35]. Stromal cells can regulate the extracellular matrix (ECM) composition and produce molecules that could attract ovarian carcinoma cells to bind to the ECM [36,37]. HGSC is typically highly vascularized, which correlates with a poor prognosis [30,38]. Ascites-associated cancer cells appear as single cells or multicellular spheroids and are responsible for peritoneal dissemination and disease relapse [39]. The multicellular spheroids are crucial mediators of metastasis, and their survival in the MA environment is essential for peritoneal dissemination because this type of cellular organization allows OC cells to resist anoikis and apoptosis induced by chemotherapeutic agents [27].

3.1. The Tumor Immune Microenvironment

OC cells and the TIME components maintain an important crosstalk, which is involved in reprogramming both innate and adaptive immune responses and promoting tumor growth and metastasis. The tumor microenvironment communication consists of an indirect cytokine-mediated interaction and a direct cell–cell interface between cancer cells and stromal cells. As a consequence of this communication, several pathways function as potent regulators contributing to the aggressive and metastatic footprints of HGSC [16]. MA contain an environment of pro-inflammatory factors that contribute to the release of mucin 16 (MUC16), a well-known glycoprotein involved in OC tumorigenesis and metastasis [40,41,42,43,44,45,46,47,48]. Pro-inflammatory cytokines and chemokines are the key element of MA and modulate HGSC in paracrine and autocrine manners [49]. This inflammatory response towards cancer cells supports the infiltration of neutrophils, which promotes cancer progression via the secretion of transforming growth factor-β (TGF-β), tumor necrosis factor (TNF)-α, metalloproteinase 9 (MMP9), reactive oxygen species (ROS), and nitric oxide (NO) [50]. The production of ROS and NO inhibits T cell infiltration and activation, which leads to T cell apoptosis [50]. In connection with this, a higher neutrophil-to-lymphocyte ratio (NLR) is correlated with decreased overall survival in OC patients [51,52].
In the OC context, MA promote an immunosuppressive microenvironment with cellular immune populations that do not reflect the populations present in the patient’s blood or tumor [53]. The immune cells are classified into two categories: anti-tumoral (cause tumor cell death) and pro-tumoral (tumor-promoting cells) (Figure 3). The anti-tumoral cells include cytotoxic T lymphocytes (CD8+) and activated helper T cells (CD4+). The pro-tumoral cells are myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs, especially pro-tumoral phenotype), lymphocyte T helper cells (Th2 subtype), and T regulatory cells (Tregs) [16]. In fact, the presence of myeloid cells, such as MDSCs and immature dendritic cells (DCs), is usually found in tumors [54,55]. There are several studies that strongly support the theory that promoting anti-tumoral responses could be crucial for controlling the tumor progression of HGSC [56].

3.1.1. Innate Immune Cells in the Ovarian Cancer Tumor Immune Microenvironment

  • Myeloid-Derived Suppressor Cells (MDSCs)
Myeloid cells are frequently observed in the stroma of tumors [57] and, in the OC context, they present an increased capacity to block local and systemic immune activation [58]. MDSCs are composed of a heterogeneous population of immature myeloid cells (macrophages, DCs, and granulocytes at initial stages of differentiation [59]) that grow in pathologic conditions and have a high potential to suppress T cells [60,61,62]. These cells are important inducers of tumor immune evasion and impaired immunity by upregulating arginase-1 and generating NO and ROS [58]. MDSCs also deplete cysteine, induce Tregs, inhibit T-cell activation and proliferation, reduce the cytolytic ability of NK cells, and trigger a pro-tumoral phenotype in macrophages [63]. In ten pre-clinical models of tumorigenesis, MDSC subgroups were associated with immune suppression [64]. The contribution of MDSCs who express the combination markers (Lin-CD45+CD33+) was studied in a cohort of patients with HGSC [65], and the results showed that 37% of non-neoplastic cells were MDSCs in the TIME, being also responsible for inhibiting T cell immunity, by blocking both T cell proliferation and effector function. In this study, the authors also showed that an increased tumor of MDSCs is negatively correlated with CD8+ tumor-infiltrate lymphocytes (TILS) and overall survival in advanced OC [66]. Curiously, the same populations of MDSCs in patients’ blood have distinct functions that they have in MA, suggesting that they support metastasis and a cancer stem cell phenotype in MA. Mechanistically, it is proved that tumor-resident MDSCs increase cancer cell stemness through the upregulation of microRNA101, which targets the co-repressor gene C-terminal binding protein-2 (CtBP2) 3′-UTR region and interferes with its binding at NANOG, OCT4/3, and SOX2 promoters in primary OC cells [65]. Additionally, the concentration of MDSCs is correlated with poor patient prognosis and elevated levels of IL-6 and IL-10 [66,67], in addition to VEGF expression and the production of adenosine by OC cells that induce MDSCs recruitment, inhibiting local immunity [66,68]. This evidence supports immunotherapeutic strategies targeting MDSCs that could help to improve antitumoral responses. For example, it is possible to block MDSC suppressor functions by decreasing the expression of CD39 and CD73 using metformin, a drug for type 2 diabetes. This blockade will promote HGSC clinical benefits by improving antitumoral T cell responses that were inhibited by MDSCs in the TIME [69]. This shows a strategy to reduce tumor progression by targeting immature myeloid cells and their crosstalk with other immune cells and cancer cells. Several drugs targeting MDSCs or tumor-associated macrophages (TAMs) have been described [69] and they include inhibitors of immune suppression function (sildenafil, triterpenoids, COX-2 inhibitors, nitric oxide inducers), antibodies that stimulate the depletion of MDSCs and/or TAMs, blockers of recruitment (by targeting chemokines and their receptors) or MDSCs proliferation, promoters of MDSCs apoptosis, TAM reprogramming factors, and inducers of immature myeloid cells differentiation (such as retinoic acid or vitamin D3) [70]. TAMs and MDSCs promote resistance to targeted immunotherapy, so repressing these populations could increase the success rate of checkpoint blockade inhibitors such as nivolumab and pembrolizumab [71].
  • Macrophages
Monocytes develop from myeloid cells found in the bone marrow and after maturation, they circulate in the bloodstream and can migrate into tissues where they differentiate into macrophages [72]. The macrophages can be polarized in separate ways, express specific surface markers, and acquire distinct functional states, depending on stimulating factors such as cytokines and other signals [29]. Activated macrophages are divided into “M1” and “M2” types based on in vitro phenotypes. Classically activated macrophages, also known as M1-like, are polarized by pro-inflammatory stimuli such as interferon (IFN)-γ and lipopolysaccharides (LPS), and they release TNFα, IL-1β, IL-6, IL-12, and IL-23, exhibiting anti-bactericidal, immunostimulatory, and antitumoral activities [29,72]. Alternatively activated macrophages, or M2-like, are polarized by anti-inflammatory stimuli such as IL-4, IL-10, and IL-13 and they release IL-6, IL-10, IL-13, TGF-β, VEGF-A, arginase, normally related with wound healing, cellular proliferation, the resolution of inflammation, immunosuppression, tumor invasion, tumor growth, angiogenesis, and metastasis [29,72]. Hence, M1-like macrophages inhibit tumor growth and M2-like are pro-tumoral [72]. Hagemann et al. described that OC cells have the capability to regulate the macrophage phenotype. In fact, OC cells can differentiate macrophages into a TAM phenotype [73]. The work of Duluc et al. demonstrated that this process is caused by the actions of the leukemia inhibitory factor (LIF), IL-6, and the macrophage colony-stimulating factor (M-CSF) [74]. TAMs present in the omentum predominantly have an anti-inflammatory phenotype to facilitate tumor progression [75] by secreting cytokines such as IL-6 and IL-8 [76]. Yin et al., [77] showed that TAMs were localized within spheroid centers and secreted EGF, by the signaling pathway EGF–EGFR [78], causing an upregulation of integrins and VEGF signaling by the activation of the NF-κB and JNK signaling pathways [79], supporting both tumor cell proliferation and migration. Macrophages have a dual effect on tumor cells: on the one hand, they can increase cancer cells’ invasion potential by TNFα and NF-κB pathways; on the other hand, they can facilitate the OC dissemination of tumors in the peritoneum by combination with VEGF, proteases, and secreted growth factors [80].
Chemotherapy also modulates macrophage activity, promoting an inflammatory environment, which paradoxically promotes tumor growth, mediated by the release of bioactive lipids from macrophages, which stimulate cyclooxygenase (COX) pathways [81]. Reader et al. [82] showed that chemotherapy-resistant cancer cells that overexpress class III β-tubulin in response to taxanes are originated by the inhibition of EP4 receptors, leading to a downstream product of COX enzymes, Prostaglandin E2 (PGE2). EP4 is overexpressed in various EOC histotypes [83]. The inflammatory stimuli of the environment in MA activate the upregulation of class III β-tubulin [84], being associated with a more aggressive biologic OC behavior [23,84]. Macrophages in MA are distinguished by the expression of surface markers: CD163+ is associated with a recurrence-free survival [22]; CD163 is a scavenger receptor that affects hemoglobin–haptoglobin complexes but also interacts with erythroblasts and may be distorted to a pro-tumoral phenotype [85]. The macrophage-derived chemokine (MDC=CCL22) is secreted by the macrophages and OC cells of MA, which attract Tregs to the tumor, suppressing T cell immunity and enhancing tumor growth [86]. The B7-H4 is a costimulatory molecule that decreases the proliferation and cytokine production of T cells and is expressed by a subpopulation of OC stromal macrophages, so their presence correlates with the number of tumor-infiltrating Tregs, which negatively regulate T cell immunity, leading to a poor outcome in OC [87]. Recapitulated, TAMs could be considered markers of poor prognosis since there is a clear association between the abundance of TAMs and tumor progression [74].
  • Neutrophils
Neutrophils are leukocytes specialized in phagocytosis and defense against invading microorganisms. Although their antibacterial functions are well described, there is a rising interest in their role in the cancer context. In fact, intratumoral neutrophils can be separated into anti-tumor, “N1-like”, and pro-tumor, “N2-like” [88]. Based on the tumoral context, tumor-associated neutrophils (TANs) can present a different phenotype and show either N1-like or N2-like phenotypes [89]. Although there are no studies showing a specific function of TANs in OC progression, Lee et al. demonstrated that OC cells release IL-8, leading to decreased tumor growth which could be in part attributed to the recruitment of neutrophils [90]. In another study, Klink et al. demonstrated that the direct interactions between neutrophils and OC cells stimulated increased ROS production, adhesion ability, and the upregulation of CD11b/CD18 expression in neutrophils from OC patients compared with neutrophils from healthy woman volunteers [91]. The NLR has been pointed to as an indirect measurement of inflammatory status and several studies showed that elevated NLR is a prognostic factor associated with an increase in disease recurrence in several cancers [92,93,94,95,96]. A study by Cho et al. evaluated the prognostic significance of NLR in patients with OC compared with patients with benign gynecological tumors and healthy controls [93], and showed that OC patients with high preoperative NLR had a decreased overall survival compared to patients with low NLR [93], meaning that neutrophils have a potential immune deregulating role in OC.
  • Dendritic cells
DCs are professional antigen-presenting cells (APCs) that link innate and adaptive immunity and are critical for the induction of protective immune responses [30]. DCs are classified into two subtypes according to their lineage: plasmacytoid DCs (pDCs) from the lymphoid lineage, expressing CD123, CD45RA, CD8, and ILT3, and myeloid DCs (mDCs) from the myeloid lineage, expressing CD11c and CD33 [54]. Under inactive conditions, these cells wander in the body in an immature form and are responsible for detecting phagocyte pathogens. DCs are activated by their pathogen-associated molecular pattern (PAMP) receptors or danger-associated molecular pattern (DAMP) receptors, and then they mature and migrate to the lymph nodes to activate CD4+ and CD8+ T lymphocytes [54]. Although tumors produce danger signals, they are useless in inducing DC maturation, leading to a reduced number of functional cells available for effective T-cell activation [97]. Wei et al. showed that tumor-associated pDCs (TApDCs) can modify ovarian tumor immunity by inducing immunosuppressive CD8+ T lymphocytes [98]. Similarly, Curiel et al. showed that ovarian tumors can reject mDCs, which have angiogenesis inhibition properties, and attract pDCs, which enhance angiogenesis via TNFα and IL-8 secretion [99]. In OC, some TApDCs (CD11+) acquire endothelial and pericyte characteristics and participate in the preservation of tumor vasculature. In fact, the depletion of these cells results in vascular apoptosis, tumor necrosis, and an increased result of chemotherapies and anti-tumor immunity [100]. Labidi-Galy et al. found phenotypic and functional differences between TApDCs and pDCs in advanced OC, supporting the theory that pDCs exhibit pro-inflammatory properties, whereas TApDCs have strong immunosuppressive characteristics and correlate with early relapse and a poor outcome [101,102]. MAs are enriched with pDCs but not mDCs [103], which, curiously, is not correlated with the survival of HGSC patients [103], suggesting deficits in functionality. This lack of functionality could be correlated with the expression of PGE2 and its receptors (EP2 and EP4) by DCS, which comprises Toll-like receptor-mediated DC activation [104], suggesting one mechanism by which the inflammatory environment of MAs may lead to DC malfunction.
  • NK cells
NK cells are key effectors in cancer immunosurveillance, recognizing and spontaneously killing virus-infected cells, cancer cells, and pathogens [105]. NK cells secrete pro-inflammatory cytokines and chemokines such as IFNγ, TNFα, IL-6, granulocyte-macrophage colony-stimulating factor (GM-CSF), and C-C Motif Chemokine Ligand (CCL5), promoting antitumoral innate and adaptive responses in the TIME [106]. Ovarian carcinoma effusions were analyzed and show the presence of NK cells in advanced stages (IV), predicting a worse overall survival [107]. Nevertheless, it has been reported that NK cells along with effector CD8+ T cells have a positive antitumoral role [108], and the activity of circulating NK cells was related to a significant progression-free survival of OC patients [109]. Like many other cancers, ovarian carcinoma tumors express the NK cell receptor ligand ULBP2, which is an indicator of poor prognosis and could promote T-cell dysfunction in the TIME [110]. Vazquez et al. [111] found that an increase in cytolyzing cancer targets of CD56bright NK cells is not correlated with an increase in producing cytokines. Contrarywise to this, CD16+ NK cells are associated with cytotoxic responses but are significantly reduced in HGSC MA [111]. A high density of NK cells is found in MAs but often without functionality [112]: in these cases, IL-18 and TGF-β decrease CD16 expression in NKs, impeding antibody-dependent cellular cytotoxicity [113,114]. A reduced number of NK cells in MAs also correlates with chemoresistance [53]. Essentially, NK cells are activated or not, according to the equilibrium between inhibitory and activating signals through different NK receptors [114]. NK cells also express estrogen receptors and programmed death protein (PD)-1 [115], suggesting additional mechanisms by which hormonal modulation and checkpoint inhibition may affect OC. Nham et al. realize a study with an artificial APC-based ex vivo expansion technique to generate cytotoxic expanded NK cells for use in an autologous model of immunotherapy [116]. In this study, they acquire NK cells from the MAs of OC patients that upregulated the surface expression of activating receptors (NKG2D, NKp30, NKp44), producing anti-tumor cytokines in the presence of OC cells and mediating direct tumor cytotoxicity against ascites-derived, primary OC cells obtained from autologous patients. This discovery shows a possibility to create cytotoxic NK cells from the MA of OC patients, which shows a hopeful immunotherapeutic target for the second-line treatment of OC [116].

3.1.2. Adaptative Immune Cells in the Ovarian Cancer Tumor Immune Microenvironment

  • Tumor Infiltrating Lymphocytes (TILs)
TILs are a type of white blood cells, which include T cells or B cells localized in the tumor and grouped in islets (intraepithelial) and in the peritumoral space (stromal) [117,118]. TILs can be found in primary tumors and omental metastases [119,120,121,122,123,124,125,126,127] and their presence has been correlated with a good prognosis, attributable to its role in the control of tumor growth by activating anti-tumor immune response [121]. Antitumoral responses were mostly characterized by the secretion of TNFα and GM-CSF [125]. Zhang and colleagues showed that intraepithelial CD3+ TILs can be found in >50% of advanced-stage EOC with their presence correlating with a five-year overall survival rate of 38% in contrast to 4.5% in patients whose tumors do not have T cells [128]. Even after the debulking and platinum-based chemotherapy, the presence of intraepithelial CD3+ TILs increased the five-year overall survival rate (>70%) in comparison to patients whose tumors do not contain TILS (11%) [129]. T cell-rich tumors are correlated with better overall survival (OS) and were associated with the increased expression of IL-2, IFN-γ, and lymphocyte-attracting chemokines within the tumor such as CXCL9 [118], CCL21, and CCL22 [130]. On the contrary, tumors without TILS were associated with an increased level of VEGF, an angiogenic regulatory factor in the TIME associated with early recurrence and short survival [131]. An investigation of the composition of TILS in patients with OC at different stages by Fialova et al. showed that early stages were characterized by a strong Th17 immune response followed by Th1 recruitment for stage II [129]. Among infiltrating T cells, CD8+ T cells are associated with a better prognosis, while CD4+ T cells expressing the transcription factor FOXP3 (a marker of Tregs) suppress the beneficial effects of CD8+ T cells in the TIME [88,111,132,133,134,135,136]. Helios+ (a hematopoietic-specific transcription factor involved in the regulation of lymphocyte development) activated Tregs, and high quantities of myeloid dendritic cells and monocytes/macrophages were detected in the advanced stages III and IV [133]. Another study proved that the intratumoral accumulation of CXCR3 ligands, such as CXCL9 and CXCL10, predicts doubled overall survival in advanced HGSC [137]. This study also identified PGE2 as a negative regulator of chemokine secretion that contributes to tumor progression by blocking TILs recruitment in OC [137]. Other researchers showed that reduced EOC patient survival is related to the expression of both COX-1 and COX-2, which are negatively correlated with intraepithelial CD8+ TILs [138]. Some studies showed that improved disease-specific survival for EOC patients is correlated with the presence of both intraepithelial CD4+ and CD8+ T cells [139,140]. However, other studies show that this helpful characteristic is attributed only to intraepithelial CD8+ TILS [141]. In 2012, a meta-analysis of 10 studies gathering 1815 OC patients proved the prognostic value of intraepithelial CD8+ TILs in EOC specimens regardless of the tumor grade, stage, or histologic subtype [129]. The presence of this specific TILs population in cancer tissues suggests that they spontaneously activated antitumoral responses to control tumor outgrowth [129]. The presence of tumor-reactive antibodies and T cells in the peripheral blood of advanced-stage EOC patients [142,143,144] and oligoclonal tumor-reactive T cells, isolated from blood, MA, and tumors, show the antitumoral response activated by TILs [145,146,147,148]. On the other hand, the lack of intraepithelial TILs is significantly associated with poor survival among EOC patients [129]. At present, immunotherapies aiming to increase the effector functions of pre-existing antitumoral CD8+ TILs and triggering effector T cell-trafficking to the TIME are the big goals of cancer immunotherapy [118].
  • CD8+ T Lymphocytes
CD8+ T lymphocytes, also known as cytotoxic T lymphocytes, are specialized in killing virus-infected cells and tumor cells. They secrete perforin which creates pores in the plasma membrane of target cells, as well as granzyme, a serine protease that activates caspases and leads to cell apoptosis.
After antigen presentation, naïve CD8+ T cells are activated and differentiate into multipotent memory stem cells, which progressively differentiate into memory T-cell (TM) subpopulations and eventually effector CD8+ T cells [149]. This sequential differentiation process is supported by a stepwise loss of plasticity, proliferative potential, and capacity for homing into lymphoid organs. In parallel, these cells acquire cytotoxicity, the production of the proinflammatory cytokines IFNγ and TNFα, tropism for inflamed tissues, and eventually a senescent phenotype [149,150]. TM cells are divided into central memory (TCM) and effector memory (TEM) cells, according to different homing and functional properties [151]. TCM cells express lymph node homing receptors (CCR7 and CD62L), produce lower levels of IFNγ in response to antigen presentation, and differentiate into TEM cells upon secondary stimulation. In contrast, TEM cells express receptors for chemotaxis into inflamed tissue (CXCR3) and rapidly secrete higher levels of effector cytokine IFNγ after memory stimulation with an antigen [151]. Besides these specificities, CD8+ T cells change in their dependence on glycolysis and oxidative phosphorylation as their energy source [149,152,153]. Activated CD8+ T cells rewire their metabolism to achieve the energetic and anabolic requirements to support their rapid proliferation and effector function, so they increase glucose and glutamine uptake, aerobic glycolysis, oxidative phosphorylation, and glutaminolysis, while they suppress fatty acid oxidation. Moreover, the tricarboxylic acid cycle is also used as a source of intermediates for nucleotide, protein, and lipid synthesis [50,154,155]. Several studies report that the presence of intra-tumoral T cells in ovarian carcinoma decreases the rate of recurrences and prolongs patient survival [18,34,131,156], indicating that T cells contribute to a more efficient eradication of tumor cells. In addition, the OC environment damages the anti-tumor function of CD8+ T cells through the inhibition of signaling pathways, including checkpoints triggered by cytotoxic T lymphocyte antigen 4 (CTLA-4), programmed cell death protein 1 (PD1), lymphocyte activation gene 3 protein (LAG-3), and T-cell immunoglobulin and mucin-domain containing protein 3 (TIM-3) [157,158,159,160]. These suppressive signals are delivered by ligands expressed on antigen-presenting cells, tumor cells, and tumor-infiltrating immune cells. The induced suppression can be efficiently blocked by antibodies, including anti-CTLA-4, anti-PD1, and anti-PD-L1 [161,162]; nevertheless, the specific contribution of immune checkpoints to OC progression is doubtful. The checkpoint blockade by anti-CTLA4 antibody [157] or anti-PD1 antibody [159] showed limited clinical efficacy and it is already known that the over-expression of PD-L1 in MA tumor cells is associated with CD8+ T cells malfunction and reduction in the anti-tumoral response [163]. The expression of PD1 is also a characteristic of T cell exhaustion, detected by low levels of secretion of cytokines by T-cells and its loss of cytotoxicity, which allows its elimination from the TIME [164]. The suppressive effect of MA involves a reduced T cell receptor (TCR) signaling and the activation of the transcription factors NF-κB and NFAT, which are crucial for T cell activation, resulting in the inhibition of signal transduction upstream of phospholipase C-γ (PLCγ) [165]. Nevertheless, the infiltrating CD8+ T cells could maintain their functionality because it was proven that the increased levels of IFNγ in tumor tissue of patients with favorable clinical outcomes are maintained. Further, MA play an essential role in T cell biology, thus favorable (i.e., recruitment of T cells into the tumor) and unfavorable (i.e., inhibition of activation-associated signal transduction pathways) aspects [166]. The utilization of these cytotoxic cells through improved immune therapies could be a way to improve the prognosis of OC patients [30,166].
  • CD4+ T cells
T helper (Th) cells are CD4+ naïve T lymphocytes activated by antigen-presenting cells (APCs). CD4+ and CD8+ T cells can specifically recognize tumor-associated antigens from cancer cells. CD4+ T cells provide cytokine support for CD8+ T cell proliferation and expansion to destroy cancer cells and trigger antitumoral responses [118]. In the context of OC, in this review, we will focus only on these four subtypes of T helper cells: Th1, Th2, Th17, and Tregs, which are defined by the cytokine environment and interactions with APCs. These different subtypes stimulate the immune system, even though they have differences in their cytokine production and target cells. Th1 cells are associated with proinflammatory responses important to killing intracellular parasites and spreading autoimmune response by secreting IL-2, IL-3, IFNγ, TNFα, and GM-CSF [167]. They are also responsible for the activation of CD8+ T cells or anti-tumoral macrophages. On the contrary, Th2 cells are associated with anti-inflammatory response, immunoglobulin E, and eosinophilic responses by IL-4, IL-5, and IL-13 secretion [167] and the activation of TAMS and, therefore, are associated with tumor progression [168]. There is a direct link between Th2 cells and OC because these cells produce specific cytokines (e.g., IL-4) present in MA and are correlated with a poor prognosis in OC [167]. Th17 cells secrete the cytokine IL-17 that has both effects, anti-tumoral and pro-tumoral, and shows strong interactions with Tregs [169], and the chemokines CXCL9 and CXCL10 that recruit TILs [170]. Miyahara et al. demonstrated that OC tumors have Th17 cells and OC cells, and tumor-associated APCs secrete cytokines that could be responsible for the increase in Th17 cells [171]. However, Fialova et al., observed a high amount of Th17 cells only in the early stages of OC [129], so they decrease with the advancing stage and appear to be inversely related to Tregs [172,173]. Furthermore, Leveque et al. proved that IL-2 can trigger the conversion of OC-associated CD4+ Tregs into Th17 cells [169]. Recent data showed that a low density of IL-17-producing cells is related to the increased ratio of CD4/CD8 T cells within MA and is associated with compromised survival [174]. Although the presence of Th17 cells has been confirmed in OC, the specific effects of these cells in tumor progression wait to be revealed.
Tregs cells are a suppressor T cell population responsible for the immune tolerance to self-antigens and the regulation of the immune system [30]. These cells can inhibit the activity of effector T cells due to their competition for IL-2, thus causing a decrease in the levels of this T cell growth factor by the production of immunosuppressive factors (such as IL-10 or TGF-β) or through cell-to-cell contact [175]. Further, in the TIME, Tregs can promote angiogenesis via VEGF production [172]. Tregs have also been associated with the immune paralysis of DCs, which contribute to immunosuppression [58,62]. Regarding the accumulation of Tregs in solid tumors and MA, there is evidence of the direct migration of naturally occurring Tregs mediated by the expression of chemokines receptors (CCR4, CCR5, and CXCR1 [176]) by the TIME, and it has been demonstrated that the levels of circulating Tregs are increased in OC patients [177,178]. Tregs are characterized by the expression of CD4, CD25, and FOXP3 and can be attracted by CCL22 secreted by OC cells or TAMs through interaction with CCR4 [88,179] and other chemokine receptors, such as CXCR3 or CCR10 [144,175]. The chemokine CCR10 ligand (CCL28) produced by tumor cells under hypoxic conditions contributes to Tregs infiltration [180]. Importantly, higher levels of activated Tregs were found in MA when compared with blood [181], meaning that tumor-infiltrating Tregs in MA produce higher levels of IL-10, express higher levels of the activation marker CD69, and proliferate at a higher extent than circulating Tregs from the same patients [181,182]. The number of Tregs also correlates with the proportion of epithelial cell adhesion molecule (EpCAM)+ cancer-derived epithelial cells in MA [181], and they increase with stage [183]. Peng et al. reviewed the different populations of Tregs and their potential clinical applications in OC [181]. It is now clear that OC cells modulate the phenotype of immune cells, corroborating Alvero et al.’s findings showing the existence of two subpopulations of OC cells with different cytokine profiles: cancer stem cells and differentiated cancer cells [182]. The differentiated cancer cells can increase the production of Tregs, leading to a tolerant microenvironment, inhibiting an immune response, being correlated with poor survival in OC patients [88]. Since Tregs are strongly influenced by microenvironmental regulation, a strategy of reprogramming these cells could be an alternative for OC treatment to be explored.

3.1.3. Cytokines Present in Malignant Ascites

Cytokines mediate key interactions between immune and non-immune cells in TIME. MA have a highly immunosuppressive microenvironment [153] and it was demonstrated in the presence of both the pro-tumorigenic and anti-tumorigenic factors of TIME [41,42,184,185]. The anti-tumorigenic cytokines are secreted by Th1 cells, such as IL-2, IL-3, INFγ, TNFα, CCL4, and CXCL10, and the pro-tumorigenic cytokines are secreted by Th2 cells, including IL-4, IL-5, IL-9, IL-10, IL-13, IL-15, CCL2, and VEGF [156,186]. Figure 4 shows some effects of cytokines in the tumor microenvironment. These cytokines cumulatively contribute to creating a pro-inflammatory and immunosuppressive tumor microenvironment [187].
As mentioned, several interleukins are clearly associated with a Th1 or Th2 response, so significantly lower levels of IL-2 in MAs are consistent with a reduced Th1 response [185], because IL-2 is associated with T cell and NK cell growth factor but blocks T cell responses by the maintenance of Treg cells and induction of activation-induced cell death [188].
Xie and colleagues found that decreased levels of IL-7 (associated with carcinogenesis, immunosuppression, and epithelial–mesenchymal transition [189,190]) appear to have similar increasing effects as IL-2 in MAs compared to serum [189]. IL-7 is also important for lymphocyte survival and is associated with the induction of anti-tumor T-cell response and decreased levels of IL-17 [191,192].
IL-15 (activates lymphocytes to produce IFNγ [191]) binds to the IL-2 receptor and stimulates both antigen-independent expansion and the long-term survival of anti-tumor CD8+ T cells [193].
Some studies evaluated cytokine concentrations in MA and found increased levels of inhibitor cytokines such as IL-6 and IL-10 but, in some cases, also found elevated levels of stimulatory cytokines such as IL-1β and TNFα compared to the normal control [184]. IL-1β promotes inflammation-induced carcinogenesis but also recruits antineoplastic immune cells that may block metastatic outgrowth [194,195]. TNFα produces and maintains a network of other mediators that promote tumor growth and peritoneal spread by stimulating the release of IL-6 and other chemokines such as CCL2 and CXCL12, macrophage migration-inhibitory factor (MIF), and VEGF. All these factors may act in an autocrine/paracrine manner to promote the colonization of the peritoneum and neovascularization of developing tumor implants [45].
The cytokines IL-6 and IL-10 are frequently analyzed due to their correlation with poor prognosis and response to therapy [42,46]. IL-6 is associated with MA formation in OC patients being involved in the upregulation of VEGF expression which leads to increased vascular permeability [185], promotes tumor growth, mediates cytokine release, and is associated with cachexia (extreme weight loss and muscle wasting) in cancer patients [196,197,198]. IL-10 promotes cytotoxicity but inhibits anti-tumor responses [199,200]. This cytokine is produced at high levels by MDSCs and plays a role in creating a tumor-permissive microenvironment [201,202], so its blockage improves MDSC-mediated immunosuppression and improves survival because this function is not redundant with other immunosuppressive molecules [202]. The blockage promoted by IL-10 increases cytotoxic T cell function in the peritoneal cavity and limits tumor spread [84]. In MAs are found high levels of IL-4, IL-10, TGF-β, and VEGF [182,183]. IL-4, IL-10, and TGF-β can affect phagocyte function, silencing macrophages, and DC activity [188]. Importantly, TGF-β can be also produced by ovarian cancer cells and is a powerful immunosuppressor within the tumor microenvironment, affecting NK and dendritic cell activity, cytokine production, and T-cell function [203]. The increased secretion of TGF-β within the tumor microenvironment recruits Tregs via the expression of FoxP3 [204], which ultimately results in diminished cytotoxic T-lymphocytes [205].
The overexpression of the proangiogenic chemokine IL-8 has been associated with poor outcomes in OC, enhancing tumor progression [206] by promoting tumor implant neovascularization [184] and attracting neutrophils facilitating a suppressive environment [207].
The chemokines are also important to the recruitment of immune cells into the tumor microenvironment, having distinct effects on tumor progression. The chemokines CXCL10 and CCL4 are found elevated in MAs, with CXCL10 being associated with increased anti-tumor response in a mouse central nervous system model [208] and the intratumoral expression of CCL4 correlated with the inhibition of colorectal tumor growth and tumor-specific CD8+ T cells response [209].
The neutralization of the chemokine CCL2 results in reduced tumor burden in a mouse prostate cancer model [210]. On the other hand, CCL5 is a chemokine that appears to induce the activation of NK cells and enhances anti-tumor immunity in a mouse model [211]. PDGF regulates cell growth and division with a significant role in angiogenesis, inducing VEGF production in OC cells, which leads to the correlation between PDGF-BB and VEGF expression in MAs [212]. The overexpression of VEGF has an important role in OC progression and blocking this pathway appears to improve survival [213,214].
Evaluating the influence of chemokines in OC, we could conclude that the overexpression of VEGF and CCL2 and the reduction in CCL5 in MA may provide a facilitating pathway for tumor dissemination in the peritoneal cavity [215].
Giuntoli L. Robert and colleagues compared the levels of 27 cytokines and chemokines in MAs and the serum of OC patients and found significant differences between these two samples. They found statistically significant elevated levels of IL-6, IL-8, IL-10, IL-15, CXCL10, CCL2, CCL4, and VEGF and significantly reduced levels of IL-2, IL-5, IL-7, IL-17, platelet-derived growth factor (PDGF)-BB, and CCL5 in MA compared to serum, concluding that MAs are an inflammatory microenvironment [153].
In conclusion, different cytokines and chemokines are associated with the prognosis in OC. IL-2, IL-5, IL-7, and CCL5 are associated with a better prognosis, and IL-6, IL-8, IL-10, CCL2, and VEGF are associated with a worse prognosis. All of them are putative biomarker candidates in the OC context.

4. Concluding Remarks and Future Perspectives

The majority of OC patients are diagnosed at an advanced stage (stage III/IV) with metastasis within the pelvic and peritoneal cavities, accumulating large volumes of MA comprising a mixture of tumor and non-tumor cells. In this metastatic niche, MA play an essential role in the OC dissemination process and are present in patients at several stages of the disease progression. This liquid tumor microenvironment is rich in immune cells that interact with tumor cells and this crosstalk has a high impact on tumor progression. Thus, it is crucial to understand the mechanisms that support tumor dissemination in this liquid metastatic microenvironment, with particular attention given to the role of immune cells in this environment.
In this review, we showed that some cytokines and chemokines are associated with good prognosis in OC including IL-2, IL-5, IL-7, and CCL5. On the contrary, IL-6, IL-8, IL-10, CCL2, and VEGF are associated with a worse prognosis in these patients. The immune cells also play an important role in MA, including cells from innate and adaptive immune systems. Many of these cells (TAMs, NK cells, MSDCs, and Tregs) are dysregulated in the MA and are associated with immune suppression, chemoresistance, and worse overall survival of the patients.
Currently, several studies consider that targeting the immune metabolism could be an alternative treatment in OC patients. The current principle of immune metabolism considers that cancer cells are inserted in a context of multiple cell types, including the immune type. In fact, several metabolic drugs used in other disease contexts are being tested to target OC cell metabolism [50]. These compounds showed to have an immunotherapeutic effect when used alone or in combination with other drugs such as immune checkpoint inhibitors [50]. The understanding of these processes and the mechanisms responsible for their perturbation in individual patients could be a way to improve OC management.

Author Contributions

D.L.A.-N. performed the writing—review and editing, and A.M.-F., R.S., R.J.D.-O. and S.R. critically reviewed, visualized, and supervised the work. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Fundacão para a Ciência e a Tecnologia/Ministério da Ciência, Tecnologia e Ensino Superior and European Union through a PhD fellowship (2021.05081.BD) co-sponsored by Fundo Social Europeu (FSE) through Programa Operacional Regional Norte (Norte 2020).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The authors confirm that the materials included in this chapter do not violate copyright laws. Where relevant, appropriate permissions have been obtained from the original copyright holder(s), and all original sources have been appropriately acknowledged or referenced.

Acknowledgments

This work was developed at i3S/IPATIMUP, an Associate Laboratory of the Portuguese Ministry of Science, Technology and Higher Education, and partially supported by Fundacão para a Ciência e a Tecnologia/Ministério da Ciência, Tecnologia e Ensino Superior. Diana Nunes acknowledges to Fundacão para a Ciência e a Tecnologia/Ministério da Ciência, Tecnologia e Ensino Superior and European Union for financial support through a PhD fellowship (2021.05081.BD) co-sponsored by Fundo Social Europeu (FSE) through Programa Operacional Regional Norte (Norte 2020). The authors would like to acknowledge the editorial support, namely the constructive review of the manuscript and raised comments.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Gaona-Luviano, P.; Medina-Gaona, L.A.; Magaña-Pérez, K. Epidemiology of Ovarian Cancer. Chin. Clin. Oncol. 2020, 9, 47. [Google Scholar] [CrossRef] [PubMed]
  2. Kim, S.; Kim, B.; Song, Y.S. Ascites Modulates Cancer Cell Behavior, Contributing to Tumor Heterogeneity in Ovarian Cancer. Cancer Sci. 2016, 107, 1173–1178. [Google Scholar] [CrossRef] [PubMed]
  3. de Leo, A.; Santini, D.; Ceccarelli, C.; Santandrea, G.; Palicelli, A.; Acquaviva, G.; Chiarucci, F.; Rosini, F.; Ravegnini, G.; Pession, A.; et al. What Is New on Ovarian Carcinoma: Integrated Morphologic and Molecular Analysis Following the New 2020 World Health Organization Classification of Female Genital Tumors. Diagnostics 2021, 11, 697. [Google Scholar] [CrossRef] [PubMed]
  4. Herzog, T.J. Recurrent Ovarian Cancer: How Important Is It to Treat to Disease Progression? Clin. Cancer Res. 2004, 10, 7439–7449. [Google Scholar] [CrossRef]
  5. Herzog, T.J.; Pothuri, B. Ovarian Cancer: A Focus on Management of Recurrent Disease. Nat. Clin. Pract. Oncol. 2006, 3, 604–611. [Google Scholar] [CrossRef]
  6. Shih, I.-M.; Kurman, R.J. Ovarian Tumorigenesis: A Proposed Model Based on Morphological and Molecular Genetic Analysis. Am. J. Pathol. 2004, 164, 1511–1518. [Google Scholar] [CrossRef]
  7. Karst, A.M.; Drapkin, R. Ovarian Cancer Pathogenesis: A Model in Evolution. J. Oncol. 2010, 2010, 932371. [Google Scholar] [CrossRef]
  8. Momenimovahed, Z.; Tiznobaik, A.; Taheri, S.; Salehiniya, H. Ovarian Cancer in the World: Epidemiology and Risk Factors. Int. J. Women’s Health 2019, 11, 287–299. [Google Scholar] [CrossRef]
  9. Zhang, J.; Li, X.; Ji, Z.-H.; Ma, R.; Bai, W.-P.; Li, Y. Cytoreductive Surgery plus Hyperthermic Intraperitoneal Chemotherapy Improves Survival with Acceptable Safety for Advanced Ovarian Cancer: A Clinical Study of 100 Patients. BioMed Res. Int. 2021, 2021, 1–12. [Google Scholar] [CrossRef]
  10. Preston, C.C.; Goode, E.L.; Hartmann, L.C.; Kalli, K.R.; Knutson, K.L. Immunity and Immune Suppression in Human Ovarian Cancer. Immunotherapy 2011, 3, 539–556. [Google Scholar] [CrossRef] [Green Version]
  11. Kroeger, P.T.J.; Drapkin, R. Pathogenesis and Heterogeneity of Ovarian Cancer. Curr. Opin. Obstet. Gynecol. 2017, 29, 26–34. [Google Scholar] [CrossRef]
  12. Kohn, E.C.; Ivy, S.P. Whence High-Grade Serous Ovarian Cancer. Am. Soc. Clin. Oncol. Educ. Book 2017, 37, 443–448. [Google Scholar] [CrossRef] [PubMed]
  13. Prat, J.; Mutch, D.G. Pathology of Cancers of the Female Genital Tract Including Molecular Pathology. Int. J. Gynecol. Obstet. 2018, 143, 93–108. [Google Scholar] [CrossRef] [PubMed]
  14. Prat, J.; Belhadj, H.; Berek, J.; Bermudez, A.; Bhatla, N.; Cain, J.; Denny, L.; Fujiwara, K.; Hacker, N.; Åvall-Lundqvist, E.; et al. Staging Classification for Cancer of the Ovary, Fallopian Tube, and Peritoneum. Int. J. Gynecol. Obstet. 2015, 126, 171–174. [Google Scholar] [CrossRef] [PubMed]
  15. Girolimetti, G.; Perrone, A.M.; Santini, D.; Barbieri, E.; Guerra, F.; Ferrari, S.; Zamagni, C.; de Iaco, P.; Gasparre, G.; Turchetti, D. BRCA-Associated Ovarian Cancer: From Molecular Genetics to Risk Management. BioMed Res. Int. 2014, 2014, 787143. [Google Scholar] [CrossRef]
  16. Nwani, N.G.; Sima, L.E.; Nieves-Neira, W.; Matei, D. Targeting the Microenvironment in High Grade Serous Ovarian Cancer. Cancers 2018, 10, 266. [Google Scholar] [CrossRef]
  17. Colombo, N.; Peiretti, M.; Parma, G.; Lapresa, M.; Mancari, R.; Carinelli, S.; Sessa, C.; Castiglione, M. Newly Diagnosed and Relapsed Epithelial Ovarian Carcinoma: ESMO Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up. Ann. Oncol. 2010, 21 (Suppl. S5), v23–v30. [Google Scholar] [CrossRef]
  18. Worzfeld, T.; Pogge von Strandmann, E.; Huber, M.; Adhikary, T.; Wagner, U.; Reinartz, S.; Müller, R. The Unique Molecular and Cellular Microenvironment of Ovarian Cancer. Front. Oncol. 2017, 7, 24. [Google Scholar] [CrossRef]
  19. Cavazzoni, E.; Bugiantella, W.; Graziosi, L.; Franceschini, M.S.; Donini, A. Malignant ascites: Pathophysiology and treatment. Int. J. Clin. Oncol. 2013, 18, 1–9. [Google Scholar] [CrossRef]
  20. Chiejina, M.; Kudaravalli, P.; Samant, H. Ascites. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 8 May 2022. [Google Scholar]
  21. Kipps, E.; Tan, D.S.P.; Kaye, S.B. Meeting the challenge of ascites in ovarian cancer: New avenues for therapy and research. Nature Reviews. Cancer 2013, 13, 273–282. [Google Scholar] [CrossRef] [Green Version]
  22. Lengyel, E. Ovarian Cancer Development and Metastasis. Am. J. Pathol. 2010, 177, 1053–1064. [Google Scholar] [CrossRef] [PubMed]
  23. 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]
  24. Castells, M.; Thibault, B.; Delord, J.-P.; Couderc, B. Implication of Tumor Microenvironment in Chemoresistance: Tumor-Associated Stromal Cells Protect Tumor Cells from Cell Death. Int. J. Mol. Sci. 2012, 13, 9545–9571. [Google Scholar] [CrossRef] [PubMed]
  25. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  26. Hanahan, D.; Coussens, L.M. Accessories to the Crime: Functions of Cells Recruited to the Tumor Microenvironment. Cancer Cell 2012, 21, 309–322. [Google Scholar] [CrossRef]
  27. Matte, I.; Legault, C.M.; Garde-Granger, P.; Laplante, C.; Bessette, P.; Rancourt, C.; Piché, A. Mesothelial Cells Interact with Tumor Cells for the Formation of Ovarian Cancer Multicellular Spheroids in Peritoneal Effusions. Clin. Exp. Metastasis 2016, 33, 839–852. [Google Scholar] [CrossRef] [PubMed]
  28. Steinkamp, M.P.; Winner, K.K.; Davies, S.; Muller, C.; Zhang, Y.; Hoffman, R.M.; Shirinifard, A.; Moses, M.; Jiang, Y.; Wilson, B.S. Ovarian Tumor Attachment, Invasion, and Vascularization Reflect Unique Microenvironments in the Peritoneum: Insights from Xenograft and Mathematical Models. Front. Oncol. 2013, 3, 97. [Google Scholar] [CrossRef]
  29. 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]
  30. Latifi, A.; Luwor, R.B.; Bilandzic, M.; Nazaretian, S.; Stenvers, K.; Pyman, J.; Zhu, H.; Thompson, E.W.; Quinn, M.A.; Findlay, J.K.; et al. Isolation and Characterization of Tumor Cells from the Ascites of Ovarian Cancer Patients: Molecular Phenotype of Chemoresistant Ovarian Tumors. PLoS ONE 2012, 7, e46858. [Google Scholar] [CrossRef]
  31. Reinartz, S.; Schumann, T.; Finkernagel, F.; Wortmann, A.; Jansen, J.M.; Meissner, W.; Krause, M.; Schwörer, A.M.; 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. 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] [PubMed]
  33. Vaughan, S.; Coward, J.I.; Bast, R.C.J.; Berchuck, A.; Berek, J.S.; Brenton, J.D.; Coukos, G.; Crum, C.C.; Drapkin, R.; Etemadmoghadam, D.; et al. Rethinking Ovarian Cancer: Recommendations for Improving Outcomes. Nat. Rev. Cancer 2011, 11, 719–725. [Google Scholar] [CrossRef] [PubMed]
  34. Cohen, M.; Pierredon, S.; Wuillemin, C.; Delie, F.; Petignat, P. Acellular Fraction of Ovarian Cancer Ascites Induce Apoptosis by Activating JNK and Inducing BRCA1, Fas and FasL Expression in Ovarian Cancer Cells. Oncoscience 2014, 1, 262–271. [Google Scholar] [CrossRef] [PubMed]
  35. Rieppi, M.; Vergani, V.; Gatto, C.; Zanetta, G.; Allavena, P.; Taraboletti, G.; Giavazzi, R. Mesothelial Cells Induce the Motility of Human Ovarian Carcinoma Cells. Int. J. Cancer 1999, 80, 303–307. [Google Scholar] [CrossRef]
  36. Touboul, C.; Lis, R.; al Farsi, H.; Raynaud, C.M.; Warfa, M.; Althawadi, H.; Mery, E.; Mirshahi, M.; Rafii, A. Mesenchymal Stem Cells Enhance Ovarian Cancer Cell Infiltration through IL6 Secretion in an Amniochorionic Membrane Based 3D Model. J. Transl. Med. 2013, 11, 28. [Google Scholar] [CrossRef] [PubMed]
  37. Duncan, T.J.; Al-Attar, A.; Rolland, P.; Scott, I.v; Deen, S.; Liu, D.T.Y.; Spendlove, I.; Durrant, L.G. Vascular Endothelial Growth Factor Expression in Ovarian Cancer: A Model for Targeted Use of Novel Therapies? Clin. Cancer Res. 2008, 14, 3030–3035. [Google Scholar] [CrossRef] [PubMed]
  38. Liao, J.; Qian, F.; Tchabo, N.; Mhawech-Fauceglia, P.; Beck, A.; Qian, Z.; Wang, X.; Huss, W.J.; Lele, S.B.; Morrison, C.D.; et al. Ovarian Cancer Spheroid Cells with Stem Cell-like Properties Contribute to Tumor Generation, Metastasis and Chemotherapy Resistance through Hypoxia-Resistant Metabolism. PLoS ONE 2014, 9, e84941. [Google Scholar] [CrossRef]
  39. Thériault, C.; Pinard, M.; Comamala, M.; Migneault, M.; Beaudin, J.; Matte, I.; Boivin, M.; Piché, A.; Rancourt, C. MUC16 (CA125) Regulates Epithelial Ovarian Cancer Cell Growth, Tumorigenesis and Metastasis. Gynecol. Oncol. 2011, 121, 434–443. [Google Scholar] [CrossRef]
  40. Lane, D.; Matte, I.; Rancourt, C.; Piché, A. Prognostic Significance of IL-6 and IL-8 Ascites Levels in Ovarian Cancer Patients. BMC Cancer 2011, 11, 210. [Google Scholar] [CrossRef]
  41. Matte, I.; Lane, D.; Laplante, C.; Rancourt, C.; Piché, A. Profiling of Cytokines in Human Epithelial Ovarian Cancer Ascites. Am J. Cancer Res. 2012, 2, 566–580. [Google Scholar]
  42. Mills, G.B.; May, C.; McGill, M.; Roifman, C.M.; Mellors, A. A Putative New Growth Factor in Ascitic Fluid from Ovarian Cancer Patients: Identification, Characterization, and Mechanism of Action. Cancer Res. 1988, 48, 1066–1071. [Google Scholar] [PubMed]
  43. Freedman, R.S.; Deavers, M.; Liu, J.; Wang, E. Peritoneal Inflammation—A Microenvironment for Epithelial Ovarian Cancer (EOC). J. Transl. Med. 2004, 2, 23. [Google Scholar] [CrossRef] [PubMed]
  44. Liu, F.; Kong, X.; Dou, Q.; Ye, J.; Xu, D.; Shang, H.; Xu, K.; Song, Y. Evaluation of Tumor Markers for the Differential Diagnosis of Benign and Malignant Ascites. Ann. Hepatol. 2014, 13, 357–363. [Google Scholar] [CrossRef]
  45. Lane, D.; Matte, I.; Garde-Granger, P.; Laplante, C.; Carignan, A.; Rancourt, C.; Piché, A. Inflammation-Regulating Factors in Ascites as Predictive Biomarkers of Drug Resistance and Progression-Free Survival in Serous Epithelial Ovarian Cancers. BMC Cancer 2015, 15, 492. [Google Scholar] [CrossRef]
  46. Matte, I.; Garde-Granger, P.; Bessette, P.; Piché, A. Ascites from Ovarian Cancer Patients Stimulates MUC16 Mucin Expression and Secretion in Human Peritoneal Mesothelial Cells through an Akt-Dependent Pathway. BMC Cancer 2019, 19, 406. [Google Scholar] [CrossRef]
  47. Jia, D.; Nagaoka, Y.; Katsumata, M.; Orsulic, S. Inflammation Is a Key Contributor to Ovarian Cancer Cell Seeding. Sci. Rep. 2018, 8, 12394. [Google Scholar] [CrossRef]
  48. 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]
  49. Riera-Domingo, C.; Audigé, A.; Granja, S.; Cheng, W.C.; Ho, P.C.; Baltazar, F.; Stockmann, C.; Mazzone, M. Immunity, Hypoxia, and Metabolism–the Ménage à Trois of Cancer: Implications for Immunotherapy. Physiol. Rev. 2020, 100, 1–102. [Google Scholar] [CrossRef]
  50. Yin, X.; Wu, L.; Yang, H.; Yang, H. Prognostic Significance of Neutrophil-Lymphocyte Ratio (NLR) in Patients with Ovarian Cancer: A Systematic Review and Meta-Analysis. Medicine 2019, 98, e17475. [Google Scholar] [CrossRef]
  51. Vergote, I.; Tropé, C.G.; Amant, F.; Kristensen, G.B.; Ehlen, T.; Johnson, N.; Verheijen, R.H.M.; van der Burg, M.E.L.; Lacave, A.J.; Panici, P.B.; et al. Neoadjuvant Chemotherapy or Primary Surgery in Stage IIIC or IV Ovarian Cancer. N. Engl. J. Med. 2010, 363, 943–953. [Google Scholar] [CrossRef]
  52. Bamias, A.; Tsiatas, M.L.; Kafantari, E.; Liakou, C.; Rodolakis, A.; Voulgaris, Z.; Vlahos, G.; Papageorgiou, T.; Tsitsilonis, O.; Bamia, C.; et al. Significant Differences of Lymphocytes Isolated from Ascites of Patients with Ovarian Cancer Compared to Blood and Tumor Lymphocytes. Association of CD3+CD56+ Cells with Platinum Resistance. Gynecol. Oncol. 2007, 106, 75–81. [Google Scholar] [CrossRef] [PubMed]
  53. Fricke, I.; Gabrilovich, D.I. Dendritic Cells and Tumor Microenvironment: A Dangerous Liaison. Immunol. Investig. 2006, 35, 459–483. [Google Scholar] [CrossRef]
  54. Qu, P.; Boelte, K.C.; Lin, P.C. Negative Regulation of Myeloid-Derived Suppressor Cells in Cancer. Immunol. Investig. 2012, 41, 562–580. [Google Scholar] [CrossRef] [PubMed]
  55. 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] [PubMed]
  56. Noy, R.; Pollard, J.W. Tumor-Associated Macrophages: From Mechanisms to Therapy. Immunity 2014, 41, 49–61. [Google Scholar] [CrossRef] [PubMed]
  57. Gabrilovich, D.I.; Nagaraj, S. Myeloid-Derived Suppressor Cells as Regulators of the Immune System. Nat. Rev. Immunol. 2009, 9, 162–174. [Google Scholar] [CrossRef]
  58. Ostrand-Rosenberg, S. Myeloid-Derived Suppressor Cells: More Mechanisms for Inhibiting Antitumor Immunity. Cancer Immunol. Immunother. 2010, 59, 1593–1600. [Google Scholar] [CrossRef]
  59. Almand, B.; Clark, J.I.; Nikitina, E.; van Beynen, J.; English, N.R.; Knight, S.C.; Carbone, D.P.; Gabrilovich, D.I. Increased Production of Immature Myeloid Cells in Cancer Patients: A Mechanism of Immunosuppression in Cancer. J. Immunol. 2001, 166, 678–689. [Google Scholar] [CrossRef]
  60. Bronte, V.; Serafini, P.; Apolloni, E.; Zanovello, P. Tumor-Induced Immune Dysfunctions Caused by Myeloid Suppressor Cells. J. Immunother. 2001, 24, 431–446. [Google Scholar] [CrossRef]
  61. Youn, J.-I.; Gabrilovich, D.I. The Biology of Myeloid-Derived Suppressor Cells: The Blessing and the Curse of Morphological and Functional Heterogeneity. Eur. J. Immunol. 2010, 40, 2969–2975. [Google Scholar] [CrossRef]
  62. Condello, S.; Sima, L.; Ivan, C.; Cardenas, H.; Schiltz, G.; Mishra, R.K.; Matei, D. Tissue Tranglutaminase Regulates Interactions between Ovarian Cancer Stem Cells and the Tumor Niche. Cancer Res. 2018, 78, 2990–3001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Youn, J.-I.; Nagaraj, S.; Collazo, M.; Gabrilovich, D.I. Subsets of Myeloid-Derived Suppressor Cells in Tumor-Bearing Mice. J. Immunol. 2008, 181, 5791–5802. [Google Scholar] [CrossRef] [PubMed]
  64. 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] [PubMed]
  65. 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]
  66. 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]
  67. Montalbán Del Barrio, I.; Penski, C.; Schlahsa, L.; Stein, R.G.; Diessner, J.; Wöckel, A.; Dietl, J.; Lutz, M.B.; Mittelbronn, M.; Wischhusen, J.; et al. Adenosine-Generating Ovarian Cancer Cells Attract Myeloid Cells Which Differentiate into Adenosine-Generating Tumor Associated Macrophages–A Self-Amplifying, CD39- and CD73-Dependent Mechanism for Tumor Immune Escape. J. Immunother. Cancer 2016, 4, 49. [Google Scholar] [CrossRef]
  68. 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]
  69. de Sanctis, F.; Bronte, V.; Ugel, S. Tumor-Induced Myeloid-Derived Suppressor Cells. Microbiol. Spectr. 2016, 4. [Google Scholar] [CrossRef]
  70. Engblom, C.; Pfirschke, C.; Pittet, M.J. The Role of Myeloid Cells in Cancer Therapies. Nat. Rev. Cancer 2016, 16, 447–462. [Google Scholar] [CrossRef]
  71. Orecchioni, M.; Ghosheh, Y.; Pramod, A.B.; Ley, K. Macrophage Polarization: Different Gene Signatures in M1(LPS+) vs. Classically and M2(LPS-) vs. Alternatively Activated Macrophages. Front. Immunol. 2019, 10, 1084. [Google Scholar] [CrossRef]
  72. Flerin, N.C.; Pinioti, S.; Menga, A.; Castegna, A.; Mazzone, M. Impact of Immunometabolism on Cancer Metastasis: A Focus on T Cells and Macrophages. Cold Spring Harb. Perspect. Med. 2020, 10, 1–32. [Google Scholar] [CrossRef] [PubMed]
  73. 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] [PubMed]
  74. 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]
  75. 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]
  76. Wang, X.; Deavers, M.; Patenia, R.; Bassett, R.L.J.; 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] [PubMed]
  77. 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]
  78. 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-Kappa B and JNK. J. Immunol. 2005, 175, 1197–1205. [Google Scholar] [CrossRef]
  79. 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]
  80. Sweat, R.S.; Stapor, P.C.; Murfee, W.L. Relationships between Lymphangiogenesis and Angiogenesis during Inflammation in Rat Mesentery Microvascular Networks. Lymphat. Res. Biol. 2012, 10, 198–207. [Google Scholar] [CrossRef]
  81. Gartung, A.; Yang, J.; Sukhatme, V.P.; Bielenberg, D.R.; Fernandes, D.; Chang, J.; Schmidt, B.A.; Hwang, S.H.; Zurakowski, D.; Huang, S.; et al. Suppression of Chemotherapy-Induced Cytokine/Lipid Mediator Surge and Ovarian Cancer by a Dual COX-2/SEH Inhibitor. Proc. Natl. Acad. Sci. USA 2019, 116, 1698–1703. [Google Scholar] [CrossRef]
  82. Reader, J.; Harper, A.K.; Legesse, T.; Staats, P.N.; Goloubeva, O.; Rao, G.G.; Fulton, A.; Roque, D.M. EP4 and Class III β-Tubulin Expression in Uterine Smooth Muscle Tumors: Implications for Prognosis and Treatment. Cancers 2019, 11, 1590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Roque, D.M.; Bellone, S.; Buza, N.; Romani, C.; Cocco, E.; Bignotti, E.; Ravaggi, A.; Rutherford, T.J.; Schwartz, P.E.; Pecorelli, S.; et al. Class III β-Tubulin Overexpression in Ovarian Clear Cell and Serous Carcinoma as a Maker for Poor Overall Survival after Platinum/Taxane Chemotherapy and Sensitivity to Patupilone. Am. J. Obs. Gynecol. 2013, 209, 62.e1-9. [Google Scholar] [CrossRef] [PubMed]
  84. Roque, D.M.; Buza, N.; Glasgow, M.; Bellone, S.; Bortolomai, I.; Gasparrini, S.; Cocco, E.; Ratner, E.; Silasi, D.-A.; Azodi, M.; et al. Class III β-Tubulin Overexpression within the Tumor Microenvironment Is a Prognostic Biomarker for Poor Overall Survival in Ovarian Cancer Patients Treated with Neoadjuvant Carboplatin/Paclitaxel. Clin. Exp. Metastasis 2014, 31, 101–110. [Google Scholar] [CrossRef] [PubMed]
  85. 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]
  86. 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]
  87. Kryczek, I.; Zou, L.; Rodriguez, P.; Zhu, G.; Wei, S.; Mottram, P.; Brumlik, M.; Cheng, P.; Curiel, T.; Myers, L.; et al. B7-H4 Expression Identifies a Novel Suppressive Macrophage Population in Human Ovarian Carcinoma. J. Exp. Med. 2006, 203, 871–881. [Google Scholar] [CrossRef]
  88. Fridlender, Z.G.; Albelda, S.M. Tumor-Associated Neutrophils: Friend or Foe? Carcinogenesis 2012, 33, 949–955. [Google Scholar] [CrossRef] [PubMed]
  89. Piccard, H.; Muschel, R.J.; Opdenakker, G. On the Dual Roles and Polarized Phenotypes of Neutrophils in Tumor Development and Progression. Crit. Rev. Oncol. Hematol. 2012, 82, 296–309. [Google Scholar] [CrossRef] [PubMed]
  90. Lee, L.F.; Hellendall, R.P.; Wang, Y.; Haskill, J.S.; Mukaida, N.; Matsushima, K.; Ting, J.P. IL-8 Reduced Tumorigenicity of Human Ovarian Cancer in Vivo Due to Neutrophil Infiltration. J. Immunol. 2000, 164, 2769–2775. [Google Scholar] [CrossRef]
  91. Klink, M.; Jastrzembska, K.; Nowak, M.; Bednarska, K.; Szpakowski, M.; Szyllo, K.; Sulowska, Z. Ovarian Cancer Cells Modulate Human Blood Neutrophils Response to Activation in Vitro. Scand. J. Immunol. 2008, 68, 328–336. [Google Scholar] [CrossRef]
  92. An, X.; Ding, P.-R.; Li, Y.-H.; Wang, F.-H.; Shi, Y.-X.; Wang, Z.-Q.; He, Y.-J.; Xu, R.-H.; Jiang, W.-Q. Elevated Neutrophil to Lymphocyte Ratio Predicts Survival in Advanced Pancreatic Cancer. Biomarkers 2010, 15, 516–522. [Google Scholar] [CrossRef] [PubMed]
  93. 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] [PubMed]
  94. Ding, P.-R.; An, X.; Zhang, R.-X.; Fang, Y.-J.; Li, L.-R.; Chen, G.; Wu, X.-J.; Lu, Z.-H.; Lin, J.-Z.; Kong, L.-H.; et al. Elevated Preoperative Neutrophil to Lymphocyte Ratio Predicts Risk of Recurrence Following Curative Resection for Stage IIA Colon Cancer. Int. J. Colorectal. Dis. 2010, 25, 1427–1433. [Google Scholar] [CrossRef]
  95. Kim, H.S.; Han, K.H.; Chung, H.H.; Kim, J.W.; Park, N.H.; Song, Y.S.; Kang, S.B. Neutrophil to Lymphocyte Ratio for Preoperative Diagnosis of Uterine Sarcomas: A Case-Matched Comparison. Eur. J. Surg. Oncol. 2010, 36, 691–698. [Google Scholar] [CrossRef] [PubMed]
  96. Shimada, H.; Takiguchi, N.; Kainuma, O.; Soda, H.; Ikeda, A.; Cho, A.; Miyazaki, A.; Gunji, H.; Yamamoto, H.; Nagata, M. High Preoperative Neutrophil-Lymphocyte Ratio Predicts Poor Survival in Patients with Gastric Cancer. Gastric. Cancer 2010, 13, 170–176. [Google Scholar] [CrossRef] [PubMed]
  97. Chen, F.; Hou, M.; Ye, F.; Lv, W.; Xie, X. Ovarian Cancer Cells Induce Peripheral Mature Dendritic Cells to Differentiate into Macrophage like Cells in Vitro. Int. J. Gynecol. Cancer 2009, 19, 1487–1493. [Google Scholar] [CrossRef] [PubMed]
  98. Wei, S.; Kryczek, I.; Zou, L.; Daniel, B.; Cheng, P.; Mottram, P.; Curiel, T.; Lange, A.; Zou, W. Plasmacytoid Dendritic Cells Induce CD8+ Regulatory T Cells in Human Ovarian Carcinoma. Cancer Res. 2005, 65, 5020–5026. [Google Scholar] [CrossRef] [PubMed]
  99. Curiel, T.J.; Cheng, P.; Mottram, P.; Alvarez, X.; Moons, L.; Evdemon-Hogan, M.; Wei, S.; Zou, L.; Kryczek, I.; Hoyle, G.; et al. Dendritic Cell Subsets Differentially Regulate Angiogenesis in Human Ovarian Cancer. Cancer Res. 2004, 64, 5535–5538. [Google Scholar] [CrossRef]
  100. Huarte, E.; Cubillos-Ruiz, J.R.; Nesbeth, Y.C.; Scarlett, U.K.; Martinez, D.G.; Buckanovich, R.J.; Benencia, F.; Stan, R.v; Keler, T.; Sarobe, P.; et al. Depletion of Dendritic Cells Delays Ovarian Cancer Progression by Boosting Antitumor Immunity. Cancer Res. 2008, 68, 7684–7691. [Google Scholar] [CrossRef]
  101. 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]
  102. 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] [PubMed] [Green Version]
  103. Wefers, C.; Duiveman-de Boer, T.; Yigit, R.; Zusterzeel, P.L.M.; van Altena, A.M.; Massuger, L.F.A.G.; de Vries, I.J.M. Survival of Ovarian Cancer Patients Is Independent of the Presence of DC and T Cell Subsets in Ascites. Front. Immunol. 2018, 9, 3156. [Google Scholar] [CrossRef] [PubMed]
  104. Brencicova, E.; Jagger, A.L.; Evans, H.G.; Georgouli, M.; Laios, A.; Attard Montalto, S.; Mehra, G.; Spencer, J.; Ahmed, A.A.; Raju-Kankipati, S.; et al. Interleukin-10 and Prostaglandin E2 Have Complementary but Distinct Suppressive Effects on Toll-like Receptor-Mediated Dendritic Cell Activation in Ovarian Carcinoma. PLoS ONE 2017, 12, e0175712. [Google Scholar] [CrossRef]
  105. Guillerey, C.; Huntington, N.D.; Smyth, M.J. Targeting Natural Killer Cells in Cancer Immunotherapy. Nat. Immunol. 2016, 17, 1025–1036. [Google Scholar] [CrossRef] [PubMed]
  106. Sungur, C.M.; Murphy, W.J. Positive and Negative Regulation by NK Cells in Cancer. Crit. Rev. Oncog. 2014, 19, 57–66. [Google Scholar] [CrossRef] [PubMed]
  107. Dong, H.P.; Elstrand, M.B.; Holth, A.; Silins, I.; Berner, A.; Trope, C.G.; Davidson, B.; Risberg, B. NK- and B-Cell Infiltration Correlates with Worse Outcome in Metastatic Ovarian Carcinoma. Am. J. Clin. Pathol. 2006, 125, 451–458. [Google Scholar] [CrossRef] [PubMed]
  108. 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]
  109. Garzetti, G.G.; Cignitti, M.; Ciavattini, A.; Fabris, N.; Romanini, C. Natural Killer Cell Activity and Progression-Free Survival in Ovarian Cancer. Gynecol. Obs. Investig. 1993, 35, 118–120. [Google Scholar] [CrossRef]
  110. Li, K.; Mandai, M.; Hamanishi, J.; Matsumura, N.; Suzuki, A.; Yagi, H.; Yamaguchi, K.; Baba, T.; Fujii, S.; Konishi, I. Clinical Significance of the NKG2D Ligands, MICA/B and ULBP2 in Ovarian Cancer: High Expression of ULBP2 Is an Indicator of Poor Prognosis. Cancer Immunol. Immunother. 2009, 58, 641–652. [Google Scholar] [CrossRef]
  111. Vazquez, J.; Chavarria, M.; Lopez, G.E.; Felder, M.A.; Kapur, A.; Romo Chavez, A.; Karst, N.; Barroilhet, L.; Patankar, M.S.; Stanic, A.K. Identification of Unique Clusters of T, Dendritic, and Innate Lymphoid Cells in the Peritoneal Fluid of Ovarian Cancer Patients. Am. J. Reprod. Immunol. 2020, 84, e13284. [Google Scholar] [CrossRef]
  112. Lai, P.; Rabinowich, H.; Crowley-Nowick, P.A.; Bell, M.C.; Mantovani, G.; Whiteside, T.L. Alterations in Expression and Function of Signal-Transducing Proteins in Tumor-Associated T and Natural Killer Cells in Patients with Ovarian Carcinoma. Clin. Cancer Res. 1996, 2, 161–173. [Google Scholar] [PubMed]
  113. Castriconi, R.; Cantoni, C.; della Chiesa, M.; Vitale, M.; Marcenaro, E.; Conte, R.; Biassoni, R.; Bottino, C.; Moretta, L.; Moretta, A. Transforming Growth Factor Beta 1 Inhibits Expression of NKp30 and NKG2D Receptors: Consequences for the NK-Mediated Killing of Dendritic Cells. Proc. Natl. Acad. Sci. USA 2003, 100, 4120–4125. [Google Scholar] [CrossRef] [PubMed]
  114. Greppi, M.; Tabellini, G.; Patrizi, O.; Candiani, S.; Decensi, A.; Parolini, S.; Sivori, S.; Pesce, S.; Paleari, L.; Marcenaro, E. Strengthening the AntiTumor NK Cell Function for the Treatment of Ovarian Cancer. Int. J. Mol. Sci. 2019, 20, 890. [Google Scholar] [CrossRef]
  115. Rodriguez, G.; Galpin, K.; McCloskey, C.; Vanderhyden, B. The Tumor Microenvironment of Epithelial Ovarian Cancer and Its Influence on Response to Immunotherapy. Cancers 2018, 10, 242. [Google Scholar] [CrossRef] [PubMed]
  116. Nham, T.; Poznanski, S.M.; Fan, I.Y.; Shenouda, M.M.; Chew, M.V.; Lee, A.J.; Vahedi, F.; Karimi, Y.; Butcher, M.; Lee, D.A.; et al. Ex Vivo-Expanded NK Cells from Blood and Ascites of Ovarian Cancer Patients Are Cytotoxic against Autologous Primary Ovarian Cancer Cells. Cancer Immunol. Immunother. 2018, 67, 575–587. [Google Scholar] [CrossRef]
  117. Santoiemma, P.P.; Powell, D.J.J. Tumor Infiltrating Lymphocytes in Ovarian Cancer. Cancer Biol. Ther. 2015, 16, 807–820. [Google Scholar] [CrossRef]
  118. Wang, W.; Zou, W.; Liu, J.R. Tumor-Infiltrating T Cells in Epithelial Ovarian Cancer: Predictors of Prognosis and Biological Basis of Immunotherapy. Gynecol. Oncol. 2018, 151, 1–3. [Google Scholar] [CrossRef]
  119. Stumpf, M.; Hasenburg, A.; Riener, M.-O.; Jütting, U.; Wang, C.; Shen, Y.; Orlowska-Volk, 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]
  120. 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]
  121. Raspollini, M.R.; Castiglione, F.; Rossi Degl’innocenti, D.; Amunni, G.; Villanucci, A.; Garbini, F.; Baroni, G.; Taddei, G.L. Tumour-Infiltrating Gamma/Delta T-Lymphocytes Are Correlated with a Brief Disease-Free Interval in Advanced Ovarian Serous Carcinoma. Ann. Oncol. 2005, 16, 590–596. [Google Scholar] [CrossRef]
  122. Tomsová, M.; Melichar, B.; Sedláková, I.; Steiner, I. Prognostic Significance of CD3+ Tumor-Infiltrating Lymphocytes in Ovarian Carcinoma. Gynecol. Oncol. 2008, 108, 415–420. [Google Scholar] [CrossRef] [PubMed]
  123. 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] [PubMed]
  124. 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 (T(Reg)) Cells Are Associated with a Metastatic Phenotype. Gynecol. Oncol. 2010, 116, 556–562. [Google Scholar] [CrossRef] [PubMed]
  125. Hamanishi, J.; Mandai, M.; Abiko, K.; Matsumura, N.; Baba, T.; Yoshioka, Y.; Kosaka, K.; Konishi, I. The Comprehensive Assessment of Local Immune Status of Ovarian Cancer by the Clustering of Multiple Immune Factors. Clin. Immunol. 2011, 141, 338–347. [Google Scholar] [CrossRef] [PubMed]
  126. 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]
  127. Dadmarz, R.D.; Ordoubadi, A.; Mixon, A.; Thompson, C.O.; Barracchini, K.C.; Hijazi, Y.M.; Steller, M.A.; Rosenberg, S.A.; Schwartzentruber, D.J. Tumor-Infiltrating Lymphocytes from Human Ovarian Cancer Patients Recognize Autologous Tumor in an MHC Class II-Restricted Fashion. Cancer J. Sci. Am. 1996, 2, 263–272. [Google Scholar]
  128. Zhang, L.; Conejo-Garcia, J.R.; Katsaros, D.; Gimotty, P.A.; Massobrio, M.; Regnani, G.; Makrigiannakis, A.; Gray, H.; Schlienger, K.; Liebman, M.N.; et al. Intratumoral T Cells, Recurrence, and Survival in Epithelial Ovarian Cancer. N. Engl. J. Med. 2003, 348, 203–213. [Google Scholar] [CrossRef]
  129. Fialová, A.; Partlová, S.; Sojka, L.; Hromádková, H.; Brtnický, T.; Fučíková, J.; Kocián, P.; Rob, L.; Bartůňková, J.; Spíšek, 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]
  130. 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]
  131. Chang, D.-K.; Peterson, E.; Sun, J.; Goudie, C.; Drapkin, R.I.; Liu, J.F.; Matulonis, U.; Zhu, Q.; Marasco, W.A. Anti-CCR4 Monoclonal Antibody Enhances Antitumor Immunity by Modulating Tumor-Infiltrating Tregs in an Ovarian Cancer Xenograft Humanized Mouse Model. Oncoimmunology 2016, 5, e1090075. [Google Scholar] [CrossRef]
  132. Komdeur, F.L.; Wouters, M.C.A.; Workel, H.H.; Tijans, A.M.; Terwindt, A.L.J.; Brunekreeft, K.L.; Plat, A.; Klip, H.G.; Eggink, F.A.; Leffers, N.; et al. CD103+ Intraepithelial T Cells in High-Grade Serous Ovarian Cancer Are Phenotypically Diverse TCRαβ+ CD8αβ+ T Cells That Can Be Targeted for Cancer Immunotherapy. Oncotarget 2016, 7, 75130–75144. [Google Scholar] [CrossRef] [PubMed]
  133. Preston, C.C.; Maurer, M.J.; Oberg, A.L.; Visscher, D.W.; Kalli, K.R.; Hartmann, L.C.; Goode, E.L.; Knutson, K.L. The Ratios of CD8+ T Cells to CD4+CD25+ FOXP3+ and FOXP3- T Cells Correlate with Poor Clinical Outcome in Human Serous Ovarian Cancer. PLoS ONE 2013, 8, e80063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Bronger, H.; Singer, J.; Windmüller, C.; Reuning, U.; Zech, D.; Delbridge, C.; Dorn, J.; Kiechle, M.; Schmalfeldt, B.; Schmitt, M.; et al. CXCL9 and CXCL10 Predict Survival and Are Regulated by Cyclooxygenase Inhibition in Advanced Serous Ovarian Cancer. Br. J. Cancer 2016, 115, 553–563. [Google Scholar] [CrossRef]
  135. Liu, M.; Matsumura, N.; Mandai, M.; Li, K.; Yagi, H.; Baba, T.; Suzuki, A.; Hamanishi, J.; Fukuhara, K.; Konishi, I. Classification Using Hierarchical Clustering of Tumor-Infiltrating Immune Cells Identifies Poor Prognostic Ovarian Cancers with High Levels of COX Expression. Mod. Pathol. 2009, 22, 373–384. [Google Scholar] [CrossRef]
  136. Han, L.Y.; Fletcher, M.S.; Urbauer, D.L.; Mueller, P.; Landen, C.N.; Kamat, A.A.; Lin, Y.G.; Merritt, W.M.; Spannuth, W.A.; Deavers, M.T.; et al. HLA Class I Antigen Processing Machinery Component Expression and Intratumoral T-Cell Infiltrate as Independent Prognostic Markers in Ovarian Carcinoma. Clin. Cancer Res. 2008, 14, 3372–3379. [Google Scholar] [CrossRef] [PubMed]
  137. 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]
  138. 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]
  139. Taylor, D.D.; Atay, S.; Metzinger, D.S.; Gercel-Taylor, C. Characterization of Humoral Responses of Ovarian Cancer Patients: Antibody Subclasses and Antigenic Components. Gynecol. Oncol. 2010, 116, 213–221. [Google Scholar] [CrossRef]
  140. Shi, J.-X.; Qin, J.-J.; Ye, H.; Wang, P.; Wang, K.-J.; Zhang, J.-Y. Tumor Associated Antigens or Anti-TAA Autoantibodies as Biomarkers in the Diagnosis of Ovarian Cancer: A Systematic Review with Meta-Analysis. Expert Rev. Mol. Diagn. 2015, 15, 829–852. [Google Scholar] [CrossRef]
  141. Taylor, D.D.; Gercel-Taylor, C.; Parker, L.P. Patient-Derived Tumor-Reactive Antibodies as Diagnostic Markers for Ovarian Cancer. Gynecol. Oncol. 2009, 115, 112–120. [Google Scholar] [CrossRef]
  142. Jang, M.; Yew, P.-Y.; Hasegawa, K.; Ikeda, Y.; Fujiwara, K.; Fleming, G.F.; Nakamura, Y.; Park, J.-H. Characterization of T Cell Repertoire of Blood, Tumor, and Ascites in Ovarian Cancer Patients Using Next Generation Sequencing. Oncoimmunology 2015, 4, e1030561. [Google Scholar] [CrossRef] [PubMed]
  143. Martin, S.D.; Wick, D.A.; Nielsen, J.S.; Little, N.; Holt, R.A.; Nelson, B.H. A Library-Based Screening Method Identifies Neoantigen-Reactive T Cells in Peripheral Blood Prior to Relapse of Ovarian Cancer. Oncoimmunology 2017, 7, e1371895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Landskron, J.; Helland, Ø.; Torgersen, K.M.; Aandahl, E.M.; Gjertsen, B.T.; Bjørge, L.; Taskén, K. Activated Regulatory and Memory T-Cells Accumulate in Malignant Ascites from Ovarian Carcinoma Patients. Cancer Immunol. Immunother. 2015, 64, 337–347. [Google Scholar] [CrossRef]
  145. Lukesova, S.; Vroblova, V.; Tosner, J.; Kopecky, J.; Sedlakova, I.; Čermáková, E.; Vokurkova, D.; Kopecky, O. Comparative Study of Various Subpopulations of Cytotoxic Cells in Blood and Ascites from Patients with Ovarian Carcinoma. Contemp. Oncol. 2015, 19, 290–299. [Google Scholar] [CrossRef]
  146. Gattinoni, L.; Speiser, D.E.; Lichterfeld, M.; Bonini, C. T Memory Stem Cells in Health and Disease. Nat. Med. 2017, 23, 18–27. [Google Scholar] [CrossRef]
  147. Hamann, D.; Baars, P.A.; Rep, M.H.; Hooibrink, B.; Kerkhof-Garde, S.R.; Klein, M.R.; van Lier, R.A. Phenotypic and Functional Separation of Memory and Effector Human CD8+ T Cells. J. Exp. Med. 1997, 186, 1407–1418. [Google Scholar] [CrossRef]
  148. Sallusto, F.; Lenig, D.; Förster, R.; Lipp, M.; Lanzavecchia, A. Two Subsets of Memory T Lymphocytes with Distinct Homing Potentials and Effector Functions. Nature 1999, 401, 708–712. [Google Scholar] [CrossRef]
  149. Chang, C.-H.; Pearce, E.L. Emerging Concepts of T Cell Metabolism as a Target of Immunotherapy. Nat. Immunol. 2016, 17, 364–368. [Google Scholar] [CrossRef]
  150. Gubser, P.M.; Bantug, G.R.; Razik, L.; Fischer, M.; Dimeloe, S.; Hoenger, G.; Durovic, B.; Jauch, A.; Hess, C. Rapid Effector Function of Memory CD8+ T Cells Requires an Immediate-Early Glycolytic Switch. Nat. Immunol. 2013, 14, 1064–1072. [Google Scholar] [CrossRef]
  151. Sukumar, M.; Kishton, R.J.; Restifo, N.P. Metabolic Reprograming of Anti-Tumor Immunity. Curr. Opin. Immunol. 2017, 46, 14–22. [Google Scholar] [CrossRef]
  152. Sukumar, M.; Liu, J.; Ji, Y.; Subramanian, M.; Crompton, J.G.; Yu, Z.; Roychoudhuri, R.; Palmer, D.C.; Muranski, P.; Karoly, E.D.; et al. Inhibiting Glycolytic Metabolism Enhances CD8+ T Cell Memory and Antitumor Function. J. Clin. Investig. 2013, 123, 4479–4488. [Google Scholar] [CrossRef] [PubMed]
  153. Giuntoli, R.L.; Webb, T.J.; Zoso, A.; Rogers, O.; Diaz-Montes, T.P.; Bristow, R.E.; Oelke, M. Ovarian Cancer-Associated Ascites Demonstrates Altered Immune Environment-2009. Anticancer Res. 2009, 29, 2875–2884. [Google Scholar] [PubMed]
  154. Hodi, F.S.; Butler, M.; Oble, D.A.; Seiden, M.v; Haluska, F.G.; Kruse, A.; Macrae, S.; Nelson, M.; Canning, C.; Lowy, I.; et al. Immunologic and Clinical Effects of Antibody Blockade of Cytotoxic T Lymphocyte-Associated Antigen 4 in Previously Vaccinated Cancer Patients. Proc. Natl. Acad. Sci. USA 2008, 105, 3005–3010. [Google Scholar] [CrossRef]
  155. Matsuzaki, J.; Gnjatic, S.; Mhawech-Fauceglia, P.; Beck, A.; Miller, A.; Tsuji, T.; Eppolito, C.; Qian, F.; Lele, S.; Shrikant, P.; et al. Tumor-Infiltrating NY-ESO-1-Specific CD8+ T Cells Are Negatively Regulated by LAG-3 and PD-1 in Human Ovarian Cancer. Proc. Natl. Acad. Sci. USA 2010, 107, 7875–7880. [Google Scholar] [CrossRef] [PubMed]
  156. Hamanishi, J.; Mandai, M.; Ikeda, T.; Minami, M.; Kawaguchi, A.; Murayama, T.; Kanai, M.; Mori, Y.; Matsumoto, S.; Chikuma, S.; et al. Safety and Antitumor Activity of Anti-PD-1 Antibody, Nivolumab, in Patients With Platinum-Resistant Ovarian Cancer. J. Clin. Oncol. 2015, 33, 4015–4022. [Google Scholar] [CrossRef] [PubMed]
  157. Huang, R.-Y.; Eppolito, C.; Lele, S.; Shrikant, P.; Matsuzaki, J.; Odunsi, K. LAG3 and PD1 Co-Inhibitory Molecules Collaborate to Limit CD8+ T Cell Signaling and Dampen Antitumor Immunity in a Murine Ovarian Cancer Model. Oncotarget 2015, 6, 27359–27377. [Google Scholar] [CrossRef]
  158. Chen, D.S.; Mellman, I. Elements of Cancer Immunity and the Cancer-Immune Set Point. Nature 2017, 541, 321–330. [Google Scholar] [CrossRef]
  159. Zou, W.; Wolchok, J.D.; Chen, L. PD-L1 (B7-H1) and PD-1 Pathway Blockade for Cancer Therapy: Mechanisms, Response Biomarkers, and Combinations. Sci. Transl. Med. 2016, 8, 328rv4. [Google Scholar] [CrossRef]
  160. Abiko, K.; Mandai, M.; Hamanishi, J.; Yoshioka, Y.; Matsumura, N.; Baba, T.; Yamaguchi, K.; Murakami, R.; Yamamoto, A.; Kharma, B.; et al. PD-L1 on Tumor Cells Is Induced in Ascites and Promotes Peritoneal Dissemination of Ovarian Cancer through CTL Dysfunction. Clin. Cancer Res. 2013, 19, 1363–1374. [Google Scholar] [CrossRef]
  161. Jiang, Y.; Li, Y.; Zhu, B. T-Cell Exhaustion in the Tumor Microenvironment. Cell Death Dis. 2015, 6, 1–9. [Google Scholar] [CrossRef]
  162. Simpson-Abelson, M.R.; Loyall, J.L.; Lehman, H.K.; Barnas, J.L.; Minderman, H.; O’Loughlin, K.L.; Wallace, P.K.; George, T.C.; Peng, P.; Kelleher, R.J.J.; et al. Human Ovarian Tumor Ascites Fluids Rapidly and Reversibly Inhibit T Cell Receptor-Induced NF-ΚB and NFAT Signaling in Tumor-Associated T Cells. Cancer Immun. 2013, 13, 14. [Google Scholar] [PubMed]
  163. Lieber, S.; Reinartz, S.; Raifer, H.; Finkernagel, F.; Dreyer, T.; Bronger, H.; Jansen, J.M.; Wagner, U.; Worzfeld, T.; Müller, R.; et al. Prognosis of Ovarian Cancer Is Associated with Effector Memory CD8 + T Cell Accumulation in Ascites, CXCL9 Levels and Activation-Triggered Signal Transduction in T Cells. OncoImmunology 2018, 7, e1424672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Berger, A. Th1 and Th2 Responses: What Are They? BMJ 2000, 321, 424. [Google Scholar] [CrossRef]
  165. DeNardo, D.G.; Barreto, J.B.; Andreu, P.; Vasquez, L.; Tawfik, D.; Kolhatkar, N.; Coussens, L.M. CD4(+) T Cells Regulate Pulmonary Metastasis of Mammary Carcinomas by Enhancing Protumor Properties of Macrophages. Cancer Cell 2009, 16, 91–102. [Google Scholar] [CrossRef] [PubMed]
  166. Gavalas, N.G.; Karadimou, A.; Dimopoulos, M.A.; Bamias, A. Immune Response in Ovarian Cancer: How Is the Immune System Involved in Prognosis and Therapy: Potential for Treatment Utilization. Clin. Dev. Immunol. 2010, 2010, 791603. [Google Scholar] [CrossRef]
  167. Bettelli, E.; Korn, T.; Oukka, M.; Kuchroo, V.K. Induction and Effector Functions of T(H)17 Cells. Nature 2008, 453, 1051–1057. [Google Scholar] [CrossRef]
  168. Kryczek, I.; Banerjee, M.; Cheng, P.; Vatan, L.; Szeliga, W.; Wei, S.; Huang, E.; Finlayson, E.; Simeone, D.; Welling, T.H.; et al. Phenotype, Distribution, Generation, and Functional and Clinical Relevance of Th17 Cells in the Human Tumor Environments. Blood 2009, 114, 1141–1149. [Google Scholar] [CrossRef]
  169. Leveque, L.; Deknuydt, F.; Bioley, G.; Old, L.J.; Matsuzaki, J.; Odunsi, K.; Ayyoub, M.; Valmori, D. Interleukin 2-Mediated Conversion of Ovarian Cancer-Associated CD4+ Regulatory T Cells into Proinflammatory Interleukin 17-Producing Helper T Cells. J. Immunother. 2009, 32, 101–108. [Google Scholar] [CrossRef]
  170. de Rezende, L.C.D.; Silva, I.V.; Rangel, L.B.A.; Guimarães, M.C.C. Regulatory T Cell as a Target for Cancer Therapy. Arch. Immunol. Ther. Exp. 2010, 58, 179–190. [Google Scholar] [CrossRef]
  171. Miyahara, Y.; Odunsi, K.; Chen, W.; Peng, G.; Matsuzaki, J.; Wang, R.-F. Generation and Regulation of Human CD4+ IL-17-Producing T Cells in Ovarian Cancer. Proc. Natl. Acad. Sci. USA 2008, 105, 15505–15510. [Google Scholar] [CrossRef]
  172. 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 T(Reg) Cells. Nature 2011, 475, 226–230. [Google Scholar] [CrossRef]
  173. Ding, Y.; Xu, J.; Bromberg, J.S. Regulatory T Cell Migration during an Immune Response. Trends Immunol. 2012, 33, 174–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Brtnický, T.; Fialová, A.; Laštovička, J.; Rob, L.; Špíšek, R. Clinical Relevance of Regulatory T Cells Monitoring in the Peripheral Blood of Ovarian Cancer Patients. Hum. Immunol. 2015, 76, 187–191. [Google Scholar] [CrossRef]
  175. Sawan, S.; Burt, D.J.; Stern, P.L.; Holland, C.; Elkord, E. Circulating Regulatory T Cells in Endometrial Cancer: A Role for Age and Menopausal Status. Immunol. Investig. 2011, 40, 62–75. [Google Scholar] [CrossRef] [PubMed]
  176. Wertel, I.; Surówka, J.; Polak, G.; Barczyński, B.; Bednarek, W.; Jakubowicz-Gil, J.; Bojarska-Junak, A.; Kotarski, J. Macrophage-Derived Chemokine CCL22 and Regulatory T Cells in Ovarian Cancer Patients. Tumour Biol. 2015, 36, 4811–4817. [Google Scholar] [CrossRef]
  177. Redjimi, N.; Raffin, C.; Raimbaud, I.; Pignon, P.; Matsuzaki, J.; Odunsi, K.; Valmori, D.; Ayyoub, M. CXCR3+ T Regulatory Cells Selectively Accumulate in Human Ovarian Carcinomas to Limit Type I Immunity. Cancer Res. 2012, 72, 4351–4360. [Google Scholar] [CrossRef]
  178. Singh, M.; Loftus, T.; Webb, E.; Benencia, F. Minireview: Regulatory T Cells and Ovarian Cancer. Immunol. Investig. 2016, 45, 712–720. [Google Scholar] [CrossRef]
  179. Bu, M.; Shen, Y.; Seeger, W.L.; An, S.; Qi, R.; Sanderson, J.A.; Cai, Y. Ovarian Carcinoma-Infiltrating Regulatory T Cells Were More Potent Suppressors of CD8(+) T Cell Inflammation than Their Peripheral Counterparts, a Function Dependent on TIM3 Expression. Tumour Biol. 2016, 37, 3949–3956. [Google Scholar] [CrossRef]
  180. Chen, Y.-L.; Chang, M.-C.; Chen, C.-A.; Lin, H.-W.; Cheng, W.-F.; Chien, C.-L. Depletion of Regulatory T Lymphocytes Reverses the Imbalance between Pro- and Anti-Tumor Immunities via Enhancing Antigen-Specific T Cell Immune Responses. PLoS ONE 2012, 7, e47190. [Google Scholar] [CrossRef]
  181. Peng, D.-J.; Liu, R.; Zou, W. Regulatory T Cells in Human Ovarian Cancer. J. Oncol. 2012, 2012, 345164. [Google Scholar] [CrossRef]
  182. Alvero, A.B.; Montagna, M.K.; Craveiro, V.; Liu, L.; Mor, G. Distinct Subpopulations of Epithelial Ovarian Cancer Cells Can Differentially Induce Macrophages and T Regulatory Cells toward a Pro-Tumor Phenotype. Am. J. Reprod. Immunol. 2012, 67, 256–265. [Google Scholar] [CrossRef] [PubMed]
  183. Yigit, R.; Figdor, C.G.; Zusterzeel, P.L.M.; Pots, J.M.; Torensma, R.; Massuger, L.F.A.G. Cytokine Analysis as a Tool to Understand Tumour-Host Interaction in Ovarian Cancer. Eur. J. Cancer 2011, 47, 1883–1889. [Google Scholar] [CrossRef] [PubMed]
  184. Mesiano, S.; Ferrara, N.; Jaffe, R.B. Role of Vascular Endothelial Growth Factor in Ovarian Cancer: Inhibition of Ascites Formation by Immunoneutralization. Am. J. Pathol. 1998, 153, 1249–1256. [Google Scholar] [CrossRef]
  185. Briukhovetska, D.; Dörr, J.; Endres, S.; Libby, P.; Dinarello, C.A.; Kobold, S. Interleukins in Cancer: From Biology to Therapy. Nat. Rev. Cancer 2021, 21, 481–499. [Google Scholar] [CrossRef]
  186. Rabinowich, H.; Suminami, Y.; Reichert, T.E.; Crowley-Nowick, P.; Bell, M.; Edwards, R.; Whiteside, T.L. Expression of Cytokine Genes or Proteins and Signaling Molecules in Lymphocytes Associated with Human Ovarian Carcinoma. Int. J. Cancer 1996, 68, 276–284. [Google Scholar] [CrossRef]
  187. Waldmann, T.A. Cytokines in Cancer Immunotherapy. Cold Spring Harb. Perspect. Biol. 2018, 10, a028472. [Google Scholar] [CrossRef]
  188. Damoiseaux, J. The IL-2 - IL-2 Receptor Pathway in Health and Disease: The Role of the Soluble IL-2 Receptor. Clin. Immunol. 2020, 218, 108515. [Google Scholar] [CrossRef]
  189. Xie, X.; Ye, D.; Chen, H.; Lu, W.; Cheng, B.; Zhong, H. Interleukin-7 and Suppression of Local Peritoneal Immunity in Ovarian Carcinoma. Int. J. Gynaecol. Obstet. 2004, 85, 151–158. [Google Scholar] [CrossRef]
  190. Zhao, J.; Chen, X.; Herjan, T.; Li, X. The Role of Interleukin-17 in Tumor Development and Progression. J. Exp. Med. 2019, 217, e20190297. [Google Scholar] [CrossRef]
  191. Vitiello, G.A.; Miller, G. Targeting the Interleukin-17 Immune Axis for Cancer Immunotherapy. J. Exp. Med. 2020, 217. [Google Scholar] [CrossRef]
  192. Hirahara, N.; Nio, Y.; Sasaki, S.; Minari, Y.; Takamura, M.; Iguchi, C.; Dong, M.; Yamasawa, K.; Tamura, K. Inoculation of Human Interleukin-17 Gene-Transfected Meth-A Fibrosarcoma Cells Induces T Cell-Dependent Tumor-Specific Immunity in Mice. Oncology 2001, 61, 79–89. [Google Scholar] [CrossRef] [PubMed]
  193. Chen, C.K.; Wu, M.Y.; Chao, K.H.; Ho, H.N.; Sheu, B.C.; Huang, S.C. T Lymphocytes and Cytokine Production in Ascitic Fluid of Ovarian Malignancies. J. Formos. Med. Assoc. 1999, 98, 24–30. [Google Scholar] [PubMed]
  194. Mantovani, A.; Dinarello, C.A.; Molgora, M.; Garlanda, C. Interleukin-1 and Related Cytokines in the Regulation of Inflammation and Immunity. Immunity 2019, 50, 778–795. [Google Scholar] [CrossRef] [PubMed]
  195. Gottschlich, A.; Endres, S.; Kobold, S. Therapeutic Strategies for Targeting IL-1 in Cancer. Cancers 2021, 13, 477. [Google Scholar] [CrossRef]
  196. Jones, S.A.; Jenkins, B.J. Recent Insights into Targeting the IL-6 Cytokine Family in Inflammatory Diseases and Cancer. Nat. Rev. Immunol. 2018, 18, 773–789. [Google Scholar] [CrossRef]
  197. Hirano, T. IL-6 in Inflammation, Autoimmunity and Cancer. Int. Immunol. 2021, 33, 127–148. [Google Scholar] [CrossRef]
  198. Rose-John, S. Interleukin-6 Family Cytokines. Cold Spring Harb. Perspect. Biol. 2018, 10, a028415. [Google Scholar] [CrossRef]
  199. Mustea, A.; Könsgen, D.; Braicu, E.I.; Pirvulescu, C.; Sun, P.; Sofroni, D.; Lichtenegger, W.; Sehouli, J. Expression of IL-10 in Patients with Ovarian Carcinoma. Anticancer Res. 2006, 26, 1715–1718. [Google Scholar]
  200. Ouyang, W.; O’Garra, A. IL-10 Family Cytokines IL-10 and IL-22: From Basic Science to Clinical Translation. Immunity 2019, 50, 871–891. [Google Scholar] [CrossRef]
  201. Naing, A.; Infante, J.R.; Papadopoulos, K.P.; Chan, I.H.; Shen, C.; Ratti, N.P.; Rojo, B.; Autio, K.A.; Wong, D.J.; Patel, M.R.; et al. PEGylated IL-10 (Pegilodecakin) Induces Systemic Immune Activation, CD8(+) T Cell Invigoration and Polyclonal T Cell Expansion in Cancer Patients. Cancer Cell 2018, 34, 775–791.e3. [Google Scholar] [CrossRef]
  202. Hart, K.M.; Byrne, K.T.; Molloy, M.J.; Usherwood, E.M.; Berwin, B. IL-10 Immunomodulation of Myeloid Cells Regulates a Murine Model of Ovarian Cancer. Front. Immunol. 2011, 2, 29. [Google Scholar] [CrossRef] [PubMed]
  203. Chen, W.; Wahl, S.M. TGF-beta: The missing link in CD4+CD25+ regulatory T cell-mediated immunosuppression. Cytokine Growth Factor Rev. 2003, 14, 85–89. [Google Scholar] [CrossRef]
  204. Mulé, J.J.; Schwarz, S.L.; Roberts, A.B.; Sporn, M.B.; Rosenberg, S.A. Transforming growth factor-beta inhibits the in vitro generation of lymphokine-activated killer cells and cytotoxic T cells. Cancer Immunol. Immunother. 1988, 26, 95–100. [Google Scholar] [CrossRef]
  205. Kao, J.Y.; Gong, Y.; Chen, C.M.; Zheng, Q.D.; Chen, J.J. Tumor-derived TGF-beta reduces the efficacy of dendritic cell/tumor fusion vaccine. J. Immunol. 2003, 170, 3806–3811. [Google Scholar] [CrossRef] [PubMed]
  206. Merritt, W.M.; Lin, Y.G.; Spannuth, W.A.; Fletcher, M.S.; Kamat, A.A.; Han, L.Y.; Landen, C.N.; Jennings, N.; de Geest, K.; Langley, R.R.; et al. Effect of Interleukin-8 Gene Silencing with Liposome-Encapsulated Small Interfering RNA on Ovarian Cancer Cell Growth. J. Natl. Cancer Inst. 2008, 100, 359–372. [Google Scholar] [CrossRef]
  207. Bakouny, Z.; Choueiri, T.K. IL-8 and Cancer Prognosis on Immunotherapy. Nat. Med. 2020, 26, 650–651. [Google Scholar] [CrossRef] [PubMed]
  208. Nishimura, F.; Dusak, J.E.; Eguchi, J.; Zhu, X.; Gambotto, A.; Storkus, W.J.; Okada, H. Adoptive Transfer of Type 1 CTL Mediates Effective Anti-Central Nervous System Tumor Response: Critical Roles of IFN-Inducible Protein-10. Cancer Res. 2006, 66, 4478–4487. [Google Scholar] [CrossRef]
  209. Luo, X.; Yu, Y.; Liang, A.; Xie, Y.; Liu, S.; Guo, J.; Wang, W.; Qi, R.; An, H.; Zhang, M.; et al. Intratumoral Expression of MIP-1beta Induces Antitumor Responses in a Pre-Established Tumor Model through Chemoattracting T Cells and NK Cells. Cell. Mol. Immunol. 2004, 1, 199–204. [Google Scholar]
  210. Loberg, R.D.; Ying, C.; Craig, M.; Day, L.L.; Sargent, E.; Neeley, C.; Wojno, K.; Snyder, L.A.; Yan, L.; Pienta, K.J. Targeting CCL2 with Systemic Delivery of Neutralizing Antibodies Induces Prostate Cancer Tumor Regression in Vivo. Cancer Res. 2007, 67, 9417–9424. [Google Scholar] [CrossRef] [PubMed]
  211. Tsukishiro, S.; Suzumori, N.; Nishikawa, H.; Arakawa, A.; Suzumori, K. Elevated Serum RANTES Levels in Patients with Ovarian Cancer Correlate with the Extent of the Disorder. Gynecol. Oncol. 2006, 102, 542–545. [Google Scholar] [CrossRef]
  212. Matei, D.; Kelich, S.; Cao, L.; Menning, N.; Emerson, R.E.; Rao, J.; Jeng, M.H.; Sledge, G.W. PDGF BB Induces VEGF Secretion in Ovarian Cancer. Cancer Biol. Ther. 2007, 6, 1951–1959. [Google Scholar] [CrossRef] [PubMed]
  213. Shen, G.H.; Ghazizadeh, M.; Kawanami, O.; Shimizu, H.; Jin, E.; Araki, T.; Sugisaki, Y. Prognostic Significance of Vascular Endothelial Growth Factor Expression in Human Ovarian Carcinoma. Br. J. Cancer 2000, 83, 196–203. [Google Scholar] [CrossRef] [PubMed]
  214. Burger, R.A.; Sill, M.W.; Monk, B.J.; Greer, B.E.; Sorosky, J.I. Phase II Trial of Bevacizumab in Persistent or Recurrent Epithelial Ovarian Cancer or Primary Peritoneal Cancer: A Gynecologic Oncology Group Study. J. Clin. Oncol. 2007, 25, 5165–5171. [Google Scholar] [CrossRef] [PubMed]
  215. Zhang, B.; Chen, F.; Xu, Q.; Han, L.; Xu, J.; Gao, L.; Sun, X.; Li, Y.; Li, Y.; Qian, M.; et al. Revisiting Ovarian Cancer Microenvironment: A Friend or a Foe? Protein Cell 2018, 9, 674–692. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The different origins of ovarian tumors, specifically the types of Epithelial Ovarian Carcinoma (EOC), created by Biorender.com (accessed on 26 August 2022). High-grade serous carcinoma (HGSC); Low-grade serous carcinoma (LGSC); Endometrioid carcinoma (EC); Clear cell carcinoma (CCC); Mucinous carcinoma (MC).
Figure 1. The different origins of ovarian tumors, specifically the types of Epithelial Ovarian Carcinoma (EOC), created by Biorender.com (accessed on 26 August 2022). High-grade serous carcinoma (HGSC); Low-grade serous carcinoma (LGSC); Endometrioid carcinoma (EC); Clear cell carcinoma (CCC); Mucinous carcinoma (MC).
Ijms 23 10692 g001
Figure 2. Cellular and acellular components of Malignant Ascites, created by Biorender.com (accessed on 26 August 2022) and adapted from Kim et al. [2]. MAs are composed of tumor cells (single cells or as spheroids) and non-tumor cells, including fibroblasts, mesothelial cells, endothelial cells, adipocytes, and immune cells. These types of cells communicate with each other through acellular factors, including cytokines, proteins, metabolites, and exosomes.
Figure 2. Cellular and acellular components of Malignant Ascites, created by Biorender.com (accessed on 26 August 2022) and adapted from Kim et al. [2]. MAs are composed of tumor cells (single cells or as spheroids) and non-tumor cells, including fibroblasts, mesothelial cells, endothelial cells, adipocytes, and immune cells. These types of cells communicate with each other through acellular factors, including cytokines, proteins, metabolites, and exosomes.
Ijms 23 10692 g002
Figure 3. The role of Immune Cells in the tumor microenvironment, created by Biorender.com (accessed on 26 August 2022). On the left is represented the immune cells that act as tumor killers by the production of cytokines or by phagocytosis, that can destroy tumor cells, and on the right, the cells that contribute to immune suppression. TIL- Tumor Infiltrating Lymphocytes; CD8+ T—cytotoxic T cells; Th1—CD4+ helper 1 T cell; NK—Natural Killer; M1—Macrophage Anti-tumoral; N1—Neutrophil like-type 1; DC—Dendritic cell; Treg—Regulator T cell; Th2—CD4+ helper 2 T cell; TApDC—Tumor-Associated plasmacytoid Dendritic cell; N2—Neutrophil like-type 2; M2—Macrophage Pro-tumoral; MDSc—Myeloid-Derived Suppressor Cells.
Figure 3. The role of Immune Cells in the tumor microenvironment, created by Biorender.com (accessed on 26 August 2022). On the left is represented the immune cells that act as tumor killers by the production of cytokines or by phagocytosis, that can destroy tumor cells, and on the right, the cells that contribute to immune suppression. TIL- Tumor Infiltrating Lymphocytes; CD8+ T—cytotoxic T cells; Th1—CD4+ helper 1 T cell; NK—Natural Killer; M1—Macrophage Anti-tumoral; N1—Neutrophil like-type 1; DC—Dendritic cell; Treg—Regulator T cell; Th2—CD4+ helper 2 T cell; TApDC—Tumor-Associated plasmacytoid Dendritic cell; N2—Neutrophil like-type 2; M2—Macrophage Pro-tumoral; MDSc—Myeloid-Derived Suppressor Cells.
Ijms 23 10692 g003
Figure 4. The effect of cytokines in the tumor microenvironment, created by Biorender.com (accessed on 26 August 2022), adapted by Briukhovetska et al. [185]. Immune evasion and tumor progression depend on cancer cell-intrinsic and -extrinsic cytokine signaling. It is shown that OC overexpresses certain cytokines, for example, IL-6, which act in an autocrine way to upregulate glycolysis and induce metabolic reprogramming, nuclear factor kappa-κB (NF-κB), and signal transducer and activator of transcription 3 (STAT3). These pathways in change can lead to epithelial–mesenchymal transition (EMT), increased proliferation, reduced apoptosis, increased migration, and the production of cytokines, such as IL-8 and VEGF, which induces angiogenesis. However, other cytokines, such as IL-1β, IL-13, IL-17, IL-22, IL-23, and IL-35 also induce EMT and, thus, tumor progression. Tumor-secreted IL-8 stimulates the recruitment of polymorphonuclear leukocytes (PMNs) and, in association with monocytes, they differentiate into MDSCs, inhibiting Th1 responses. MDSCs, TAMs, and M2 macrophages that are polarized by Th2-type cytokines contribute to higher levels of TGF-β that form an immunosuppressive microenvironment. In sequence, TGF-β together with IL-33 promotes the differentiation of Treg cells, which have a high affinity to IL-2 receptor (IL-2R) and are a major source of IL-10 that, under chronic conditions, suppresses antitumor responses. Additionally, TGF-β with IL-6 promotes the differentiation of Th17 cells that produce IL-17 and promote more MDSC recruitment and differentiation. pSTAT3, phosphorylated STAT3; EMT transcription factors (ZEB, SNAI1, SLUG, TWIST).
Figure 4. The effect of cytokines in the tumor microenvironment, created by Biorender.com (accessed on 26 August 2022), adapted by Briukhovetska et al. [185]. Immune evasion and tumor progression depend on cancer cell-intrinsic and -extrinsic cytokine signaling. It is shown that OC overexpresses certain cytokines, for example, IL-6, which act in an autocrine way to upregulate glycolysis and induce metabolic reprogramming, nuclear factor kappa-κB (NF-κB), and signal transducer and activator of transcription 3 (STAT3). These pathways in change can lead to epithelial–mesenchymal transition (EMT), increased proliferation, reduced apoptosis, increased migration, and the production of cytokines, such as IL-8 and VEGF, which induces angiogenesis. However, other cytokines, such as IL-1β, IL-13, IL-17, IL-22, IL-23, and IL-35 also induce EMT and, thus, tumor progression. Tumor-secreted IL-8 stimulates the recruitment of polymorphonuclear leukocytes (PMNs) and, in association with monocytes, they differentiate into MDSCs, inhibiting Th1 responses. MDSCs, TAMs, and M2 macrophages that are polarized by Th2-type cytokines contribute to higher levels of TGF-β that form an immunosuppressive microenvironment. In sequence, TGF-β together with IL-33 promotes the differentiation of Treg cells, which have a high affinity to IL-2 receptor (IL-2R) and are a major source of IL-10 that, under chronic conditions, suppresses antitumor responses. Additionally, TGF-β with IL-6 promotes the differentiation of Th17 cells that produce IL-17 and promote more MDSC recruitment and differentiation. pSTAT3, phosphorylated STAT3; EMT transcription factors (ZEB, SNAI1, SLUG, TWIST).
Ijms 23 10692 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Almeida-Nunes, D.L.; Mendes-Frias, A.; Silvestre, R.; Dinis-Oliveira, R.J.; Ricardo, S. Immune Tumor Microenvironment in Ovarian Cancer Ascites. Int. J. Mol. Sci. 2022, 23, 10692. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms231810692

AMA Style

Almeida-Nunes DL, Mendes-Frias A, Silvestre R, Dinis-Oliveira RJ, Ricardo S. Immune Tumor Microenvironment in Ovarian Cancer Ascites. International Journal of Molecular Sciences. 2022; 23(18):10692. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms231810692

Chicago/Turabian Style

Almeida-Nunes, Diana Luísa, Ana Mendes-Frias, Ricardo Silvestre, Ricardo Jorge Dinis-Oliveira, and Sara Ricardo. 2022. "Immune Tumor Microenvironment in Ovarian Cancer Ascites" International Journal of Molecular Sciences 23, no. 18: 10692. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms231810692

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