Open Access

Beyond tumor‑associated macrophages involved in spheroid formation and dissemination: Novel insights for ovarian cancer therapy (Review)

  • Authors:
    • Yuchen Liu
    • Haoyue Xiao
    • Hai Zeng
    • Ying Xiang
  • View Affiliations

  • Published online on: November 7, 2024     https://doi.org/10.3892/ijo.2024.5705
  • Article Number: 117
  • Copyright : © Liu et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].

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


Abstract

Ovarian cancer (OC) is the most common and deadly malignant tumor of the female reproductive system. When OC cells detach from the primary tumor and enter the ascitic microenvironment, they are present as individual cells or multicellular spheroids in ascites. These spheroids, composed of cancer and non‑malignant cells, are metastatic units and play a crucial role in the progression of OC. However, little is known about the mechanism of spheroid formation and dissemination. Tumor‑associated macrophages (TAMs) in the center of spheroids are key in spheroid formation and metastasis and provide a potential target for OC therapy. The present review summarizes the key biological features of spheroids, focusing on the role of TAMs in spheroid formation, survival and peritoneal metastasis, and the strategies targeting TAMs to provide new insights in treating OC.

Introduction

Ovarian cancer (OC) has the highest mortality rate (>314,000 new cases and around 207,000 new deaths worldwide in 2020) among all gynecological malignancies in the world due to lack of symptoms and effective makers at early stages (1). If the cancer is restricted to the ovaries (stage I), up to 90% of patients can be treated with currently available treatments (surgery, chemotherapy, anti-angiogenic therapy, immunotherapy). Even if the disease has spread to the pelvic organs (stage II), up to 70% of patients survive for >10 years. However, if it further disseminates into the peritoneum or the surface of abdominal organs (stage III) or outside the abdomen (stage IV), 5-year survival rate declines to ≤20% (2). For most patients with OC, tumor cytoreduction surgery is their last option at the advanced stage with intraperitoneal and extensive pelvic implantation metastasis. Even with aggressive first-line chemotherapy following optimal debulking surgery, the initial cure rate is 80%. The majority of patients with advanced-stage OC exhibit unsatisfactory response due to acquired chemoresistance, which results in recurrence and chemotherapy failure (3).

Increase in the amount of fluid in the abdominal cavity >200 ml is termed ascites. Ascites formation serves a vital role in the progression of OC, serving as a transporter of tumor cells from the primary location to metastatic sites (4). The incidence of ascites varies between the four stages of OC, ranging from 49.4 at stage I to 62.5 in stage II and 90.1 and 100.0% in stages III and IV, respectively (5). Malignant ascites (MA) contains cellular and acellular components. Cellular components include cancer, immune and mesothelial cells and fibroblasts, while acellular components include proteins, such as cytokines and growth factors, metabolites and exosomes (6). Massive ascites may cause abdominal distension, respiratory compromise, anorexia and cachexia (7). Decreased lymphatic absorption and increased fluid production via high vascular permeability are the primary factors contributing to MA formation. To date, a gold standard for clinical management of MA has not been clearly defined. Paracentesis and diuretics relieve the accumulation of ascites, but their efficacy is often partial and temporary (8).

Unlike other solid cancers, OC rarely exhibits hematogenous and lymphatic metastasis. Peritoneal (trancoelomic) metastasis is more frequent and can be detected in ~70% of OC cases (9). Tumor cells implant directly into adjacent organs after detachment from the primary site or spread to the omentum, parietal and visceral peritoneum via peritoneal fluid or ascites. In OC ascites, cancer cells float as individual cells or multicellular aggregates (also called spheroids). These malignant cells accumulate as globular structures, thus resisting anoikis and helping to spread throughout the abdominal cavity (10). Spheroids, exerting higher tumorigenic and chemoresistant properties than individual cancer cells (11), are considered metastatic units of peritoneal dissemination (12). Beside tumor cells, OC ascites also contains non-tumor cellular components, such as macrophages, lymphocytes, fibroblasts, adipocytes and mesothelial cells (13). The interaction of tumor and non-tumor cells in ascites leads to formation of heterogeneous spheroids. Such heterospheroids are more invasive and more resistant to anoikis and chemotherapeutic drugs than homospheroids composed of cancer cells alone (14).

Tumor-associated macrophages (TAMs) are the most frequent cell type (up to 50% of the total) in the ascitic microenvironment (15). Recently (16), it has been revealed that TAMs exist in the center of spheroids, regulating spheroid formation, survival and adhesion to peritoneum and then regulating peritoneal metastasis. TAMs are key for OC progression (17). Thus, it is of importance to study the role and molecular mechanisms of TAMs in spheroid formation and intraperitoneal implantation (18). Compared with previous reviews, which primarily focused on the immunosuppressive role of TAMs in cancer (15,19,20) or only briefly mentioned TAMs in OC spheroids (21-23), the present review investigates the role of TAMs in spheroid formation and dissemination in OC progression, the molecular mechanisms by which TAMs participate in peritoneal metastasis of OC, including spheroid formation, survival and dissemination, as well as strategies targeting TAMs in OC in clinical or preclinical research.

Role of spheroids in progression of OC

OC cells metastasize directly from the primary site to the abdominal cavity, where they survive and travel as individual cells or multicellular aggregates in the peritoneal fluid or ascites, then adhere to peritoneal tissue, anchor to the submesothelial matrix, and proliferate to form secondary lesions (24). These spheroids exist in, and can be isolated, from OC ascites. The role of spheroids in OC progression is summarized in Fig. 1.

Formation of spheroids in ascites

Spheroids contain cancer and non-cancer cells such as cancer-associated fibroblasts (CAFs), TAMs and rare cancer stem cells (CSCs). Spheroids have different sizes and structures (25). Spheroids may be formed by aggregation of individual stromal and cancer cells in ascites or clusters detached from the primary tumor. In mice, intraperitoneal aggregation is not the main mechanism of spheroid formation and 80% of OC spheroids are produced by clustered cells separating from the primary tumor, while intraperitoneal aggregative cells account for only a small fraction (10).

Three-dimensional (3D) cell culture is used for spheroids research in vitro. Compared with traditional monolayer cell culture, 3D spherical cells mimic the differentiation pattern in vitro and spatial contact with the intercellular and extracellular matrix (ECM) (26). The initial formation of spheroids begins with formation of relatively loose cell aggregates from integrin-ECM components, followed by expression of adhesion molecule E-cadherin, which binds to hemophilic cadherin-cadherin to form dense spheroids (27). E-cadherin has an important regulatory role in formation of tumor spheroids. Higher expression of E-cadherin is closely associated with compact spheroids (28). Mechanistically, decreased E-cadherin upregulates α5-integrin expression through epidermal growth factor receptor (EGFR)/focal adhesion kinase (FAK)/extracellular signal-regulated kinase 1 (ERK1) signaling, facilitating OC cell aggregation (29). NIH:OVCAR5 spheroids of OC cells in a 3D cell culture model of multicellular aggregates isolated from ascites show that α5β1 integrin and its ligand fibronectin are exposed on OC spheroids. Monoclonal antibodies against α5- or β1-integrin inhibit the formation of spheroids, suggesting that the interaction between α5β1-integrin and fibronectin serves a vital role in the aggregation of OC cells (30). In another study, ascitic tumor cells with high α5 integrin expression were selectively recruited by CAFs to form heterotypic spheroids (14). Moreover, EGF derived from CAFs under ascitic tumor cell stimulation is significantly enriched within heterotypic spheroids, where it increases integrin α5 expression on ascitic tumor cells, thereby strengthening interactions between ascitic tumor cells and CAFs. CAF-tumor cell spheroids are considered to be the metastatic units of high grade serous OC (14). Han et al (31) demonstrated that CAFs serve as a scaffold to gather floating tumor cells, promoting peritoneal metastasis by forming heterotypic aggregates with tumor cells. Upregulation of CD44 on the surface of OC cells is key during the progression of OC, since CD44 is a receptor for hyaluronate, a key component in ECM, which serves an important role in cell communication and adhesion between cells and the ECM. Inhibition of CD44 by its specific short hairpin RNA in OC cells decreases proliferation and spheroid formation (32).

Numerous signaling pathways are involved in spheroid formation. Chen et al (33) found that activation of signal transducer and activator of transcription 3 (STAT3) signaling pathway is associated with the formation of spheroids. In a mouse model of human ovarian carcinoma, STAT3 regulated formation of spheroids and self-renewal, while attenuation of STAT3 decreased the tumorigenicity. Wnt/β-catenin signaling is a key pathway in the regulation of the formation of spheroids through STAT3 (33). β-catenin is a marker for CSCs, which exist in the center of spheroids (34,35), and activation of β-catenin regulates the ability to initiate tumors and the formation of spheroids (36). Myeloid-derived suppressor cells enhance stemness of cancer cells, spheroid formation and cancer metastasis in an OC mouse model in vivo and microRNA (miR)-101-co-repressor gene C-terminal binding protein-2-SC core genes are involved in these effects (37). Furthermore, angiotensin II receptor contributed to the development and metastasis of OC (38). Angiotensin II significantly promotes the spheroid formation, growth and invasiveness of several OC cell lines due to the direct activation of the MAPK/ERK pathway and transactivation of EGFR), which upregulates the expression of stearoyl-CoA desaturase 1 gene, alters lipid metabolic homeostasis and inhibits endoplasmic reticulum stress within the spheroids (39).

Survival of spheroids in ascites
Resistance to anoikis

Anoikis is a specific form of apoptosis due to insufficient or poor cellular adhesion (40). Anoikis is regulated by integrins, which interact with ECM components to form adhesion complexes (41). Either through intrinsic or extrinsic apoptotic pathways, anoikis prevents epithelial cells from detaching from their original location and colonizing new sites (42). Tumor cells are less sensitive to anoikis than normal epithelial cells. Cancer cells downregulate intercellular adhesion molecules via epithelial-mesenchymal transition (EMT), inhibiting E-cadherin expression to decrease cadherin-dependent intercellular contact and allow cancer cells to resist anoikis (43). Cancer cells develop several mechanisms for abrogating anoikis, such as activating Src/AKT/ERK signaling, which is involved in anoikis resistance via blocking the mitochondrial pathway and glycolysis (44). The Notch signaling pathway, which is initiated by receptor (Notch1-4)-ligand (Δ and Jagged) interaction, has been considered a potential therapeutic target for OC (45). High expression of Notch3 is associated with poor prognosis in epithelial OC (46). Elevated Notch3 expression promotes anoikis resistance via upregulation of type IV α2 collagen (COL4A2) gene, a key component of the basement membrane that allows OC cells to maintain survival-friendly signaling by spoofing proteins responsible for detecting ECM contact, such as integrins, without making contact with ECM; FAK/AKT/ERK1/2 activation is the key mechanism (47). Hepatocyte growth factor (HGF) receptor c-Met is frequently highly expressed in OC and contributes to anoikis resistance. The effects are dependent on both phosphatidylinositol 3-kinase (PI3K)/AKT and ERK1/2 signaling pathways and Ras serves as a central role for the cross talk (48). In addition, cancer cells commonly evade apoptosis by upregulating anti-apoptotic Bcl-2 family proteins and/or downregulating pro-apoptotic proteins (49). Frizzled family receptor 7 (FZD7), which mediates both classical and non-classical Wnt signaling, plays an important role in maintaining SC properties as well as tumor development (50). A study (51) demonstrated the regulatory role of FZD7 on spheroid proliferation of CSCs in OC via activation of the Wnt/β-catenin pathway. TWIST1 is an important regulatory molecule of the Wnt3a/Wnt1/β-catenin signaling pathway, which is closely related to mesenchymal and tumor stem cell phenotypes (52). Tan et al (53) found that the FZD7/TWIST1/Bcl-2 signaling pathway played a role in the maintenance of mesenchymal phenotype and anoikis resistance and was involved in OC spheroid formation. FZD7 promoted TWIST1 expression via epigenetic modifications of H3K4me3 and H3K27ac at the TWIST1 proximal promoter; TWIST1 regulated the expression of Bcl-2, an anti-apoptotic protein (53).

In ascites, individually suspended tumor cells are more prone to anoikis than clustered cells. Multicellular spheroids consisting of tumor cells surrounded by immune and stromal cells show enhanced survival compared with individual tumor cells (54). Long non-coding RNA HOTAIR is a key indicator of poor prognosis in patients with OC. Dai et al (55) found that HOTAIR is upregulated in OC cells in suspension culture and allows cells to acquire anoikis resistance. Silencing of HOTAIR in SKOV3 cells inhibits spheroid formation, decreases aggressiveness and enhances chemosensitivity. HOTAIR promotes enhancer of zeste homolog 2 (EZH2) expression; EZH2-mediated methylation of lysine 27 on histone H3 (H3K27) contribute to the formation of spheroids (55). HOTAIR can also serve as a competitive endogenous RNA to regulate phosphoinositide-3-kinase regulatory subunit 3 (PIK3R3) and promotes proliferation, migration and invasion of OC cells. PIK3R3 is a subunit of PI3K and activation of the PI3K/AKT signaling pathway is key for cell survival (56,57). Activation of caspase-3 is a common event in both intrinsic and extrinsic anoikis. In spherical OC cells, AKT kinase is activated and promotes OC survival through inhibiting caspase-3 (58). Tropomysin-related kinase B (TrkB), a neurotrophic tyrosine kinase receptor, is overexpressed in OC tissue, particularly in greater omentum metastatic lesions and multicellular spheroids in ascites. TrkB mediates suppression of anoikis via activating the PI3K/AKT pathway (59). Furthermore, clustered cancer cells express specific adhesion molecule αv-integrin to activate survival signaling pathways. αv-integrin maintains cell survival via ERK1/2 activation, thereby enhancing the resistance of OC tumor spheroids to anoikis (60). Moreover, there are genetic differences between tumor cells in primary tissue and ascites. After tumor cells detach from primary tissue and disseminate into the abdominal cavity, tumor cells undergo independent clonal evolution. KRAS mutation in ascites leads to acquired anoikis resistance, which increases the survival of tumor cells in ascites (61).

Resistance to chemotherapeutic drugs

The spheroids in OC ascites vary in shape and size (62). It was showed that spheroids could inhibit the entry of chemotherapeutic drugs into cells to inhibit their therapeutic effects (63). The mechanism is associated a number of factors such as the 3D spatial structure inside spheroids, drug gradient penetration, cell-cell contact and low pH, dense or loose structure of spheroids and the mixed cell types (necrotic, quiescent or proliferating cells) (64). Cell adhesion molecules are key in the formation of spheroids (65). Cell division cycle 25 A (CDC25A) is a phosphatase that regulates cell cycle progression through the G1/S and G2/M checkpoints (66). CDC25A is highly expressed in patients with OC (67). CDC25A maintains the structural stability of multicellular tumor spheroids by upregulating the expression of E-cadherin protein, which improves the tolerance of OC spheroids to chemotherapeutic resistance (65). Green et al (68) showed that in HT29 multicellular spheroids cultured in vitro, E-cadherin-mediated adhesion is sensitive to 5-fluorouracil, paclitaxel, vincristine and etoposide, but not to cisplatin, after disturbance of adhesion function.

Numerous proteins involved in drug resistance are overexpressed in spheroids (69). One study compared the drug sensitivity of spherical OVAR-3 and OVAR-8 cell lines in 3D culture system with monolayer cell lines in 2D culture system: Spheroids were more resistant to cisplatin and taxol compared with monolayer of OC cell lines, and spheroids took up less taxol than monolayer cells. Large spheroids were more resistant to taxol. However, the spheroids were equally sensitive to cisplatin and there was no significant difference based on size and morphology (70). The different responses of spheroids to taxol and cisplatin may be due to the different mechanisms of these drugs: Taxol inhibits cell division by suppressing dynamics of microtubules and proper assembling of mitotic spindle; while cisplatin causes the breakage of DNA strands by formation of covalent adducts between platinum complexes and DNA (71,72).

A large number of mesothelial cells exist in spheroids (27), which may contribute to resistance to drug-induced apoptosis via releasing pro-survival factors, which activate AKT and NF-κB survival pathways. In addition, CSCs also exist in spheroids. CSCs are key for tumorigenesis, chemoresistance and recurrence. Aldehyde dehydrogenase 1 (ALDH1) is important in study of the relationship between expression and poor prognosis in ovarian tumor SCs (34). Increased ALDH activity is found in spheroids (35). CSCs are the primary cause of high recurrence rates of OC (73). When chemotherapeutic drugs are present, non-SCs in spheroids are killed by drugs but stem-like cells survive. Spherical OC cells overexpress stem cell genes under long-term treatment with chemotherapeutic drugs (74). CSC maintenance is dependent on Notch signaling pathway, especially Notch3, which also serves a key role in platinum chemoresistance (76). Inhibition of Notch3 by small interfering RNA markedly decreases the size and number of spheroids (76).

The mechanisms by which spheroids acquire resistance to anoikis and chemotherapeutic drugs are summarized in Fig. 2. After acquiring anoikis and chemotherapeutic resistance, as well as immune escape in the immunosuppressive ascitic microenvironment, spheroids may survive in the ascites, and then disseminate to the peritoneum.

Peritoneal dissemination of spheroids

The peritoneum, the largest serous membrane of the human body, covering the abdominal and pelvic cavities and visceral organs, is a preferred location for trancoelomic metastasis of numerous types of epithelial malignancy, including ovarian, colonic and gastric cancer (77). The peritoneum is composed of a layer of mesothelial cells and associated underlying ECM. These mesothelial cells serve as initial barriers for cancer cells. However, they can be induced to cancer-associated mesothelial cells (CAMs) by cancer cells. Mesothelial cells undergo to mesothelial-mesenchymal transition induced by HGF secreted by OC cells, then CAMs promote the expression of pro-tumor factors such as IL-8 and C-X-C motif chemokine ligand 5 (CXCL5) to facilitate dissemination of OC cells (78). The greater omentum is the most common metastatic site of OC, since it lacks basement membrane and mesothelial cells on the surface of milky spots (79,80).

Mesothelial cells retract during peritoneal metastasis. Unlike normal peritoneal mesothelial cells, which are flattened and spread over the entire surface of the peritoneal cavity, the mesothelial cells are rounded and separated from each other during peritoneal metastasis, exposing the submesothelial surface. OC spheroids detach from the primary tumor and disseminate to the peritoneum though ascites. These spheroids adhere to the mesothelial cells through adhesion molecules such as CD44, α5β1, αvβ1 and α2β1 integrins and mesothelial cells undergo localized retracement and detachment (81). E-cadherin loss is often associated with metastasis of OC. In OC cells, inhibited E-cadherin expression significantly upregulates the expression of α5-integrin, a subunit of fibronectin receptor α5β1-integrin, which binds with fibronectin, mediating adhesion to the peritoneal ECM (29). Once the spheroids spread over the monolayer of mesothelial cells, mesothelial cells move out from directly beneath the spreading spheroids. This is termed mesothelial clearance (82). Spheroid-induced mesothelial clearance depends on α5β1 integrin, talin I and myosin II. Following binding of mesothelial cells, spherical cancer cells utilize integrin- and talin-dependent myosin activation and traction, promoting mesothelial cells to migrate from beneath the spheroids (82).

Mesothelial clearance leads to exposure of underlying ECM and promotes further attachment of cancer cells. Cancer cells express CD44 on the cell membrane, which binds to hyaluronan in ECM to strengthen the link with peritoneal mesothelium (83), and express integrins that bind the basement membrane composed of laminin, fibronectin and types I and IV collagen (84). Spheroids readily adhere to and disaggregate from ECM substrates, particularly fibronectin and collagen I (85). Disaggregation of spheroids into individual cells is necessary for invasion of the mesothelium (86). α2β1 integrin serves a vital role in the dissemination of ovarian carcinoma spheroids: α2β1 integrin on cancer cells adheres to type I collagen, followed by secretion of serine and metalloproteinases, and contributes to the metastasis of OC into the abdominal cavity (87). In an in vitro spreading homozygous spheroid model of OC, α2β1 integrin was upregulated in spheroids and associated with disaggregation from ECM and invasion by activating MMPs such as MMP2/MMP9 (88). Blockade of α2β1 integrin using monoclonal antibodies decreases disaggregation and proteolysis of spheroids (88).

TAMs in spheroid formation, survival and metastasis

In ascites, the majority of immune cells are macrophages, which constitute >50% of MA cellular components (89). A study (17) showed that macrophages are present within all spheroids in ascites of 128 patients with OC at stage III. Compared with primary tumors, the amount of macrophages in spheroids is significantly increased and positively associated with proliferation while inversely associated with the prognosis of OC (17). In OC ascites, macrophages float in the peritoneal cavity or in the center of tumor spheroids. These macrophages may originate either from tissue-resident macrophages derived from the embryonic yolk sac or from infiltrating macrophages recruited from bone marrow-derived monocytes. They are induced to TAMs in the tumor microenvironment (TME), serving as an immunosuppressive cellular population, promoting tumor growth, immune escape, angiogenesis and metastasis. The role of TAMs in spheroid formation, survival and metastasis is summarized in Fig. 3.

Figure 3

Role of TAMs in spheroid formation, survival and metastasis. TAM-secreted VEGF binds to its receptor KDR/Flk-1 on endothelial cells, then decrease expression of junctional protein claudin-5 on endothelial cells, increase vascular permeability and contributes to ascites accumulation. Macrophages are recruited to ascites by soluble factors released by cancer cells including UBR5, M-CSF and POSTN, and chemokines released by TAMs, such as CCL18. TAM spheroids in ascites primarily detach from the primary tumor. TAM-derived EGF activates EGFR on cancer cells and increases expression of VEGF-C and VEGFR3 in cancer cells. EGF also increases the expression of αMβ2 integrin in TAMs and ICAM-1 in cancer cells to facilitate adhesion between TAMs and cancer cells. TAM-released chemokine CCL18 interacts with CCR8 to promote EMT of cancer cells. TAMs promote anoikis and drug resistance via the activation of STAT3 and Sonic hedgehog pathways and upregulation of P-gp and MRP1 in cancer cells and promote immune escape via suppressed phagocytosis of TAMs via SIRPα, inhibit T cell proliferation via B7-H4 and PD-L1 and increase Treg recruitment via CCL12. MIP-1β secreted by TAMs binds to CCR5, resulting in overexpression of P-selectin on the mesothelial cell surface. Cancer cells attached to P-selectin via CD24. TAM-derived soluble factors (such as FLT3L, leptin and HB-EGF) increase expression of MMP-9 which mediates spheroid spreading and invasion. TAM, tumor-associated macrophage; VEGF, vascular endothelial growth factor; KDR, kinase insert domain receptor; Flk, fetal liver kinase; UBR, ubiquitin protein ligase E3 component N-recognin; M-CSF, macrophage colony-stimulating factor; POSTN, periostin; CCL, chemokine (C-C motif) ligand; EGFR, epidermal growth factor receptor; CCR, C chemokine receptor; P-gp, P-glycoprotein; MRP, multidrug resistance-associated protein; SIRP, signal regulatory proteins; ASK, apoptosis signal-regulating kinase; ZEB, Zinc finger E-box binding homeobox; EMT, epithelial-mesenchymal transition; ICAM, intercellular adhesion molecule; VE-CAD, vascular endothelial-cadherin; B7-H4, B7 homolog 4; PD-L1, programmed cell death-ligand 1; MIP, macrophage Inflammatory Protein-1; Treg, regulatory T cells; FLT3L, FMS-like tyrosine kinase 3 ligand; HB, heparin-binding epidermal growth factor-like growth factor.

TAMs in angiogenesis and ascites formation

Increased permeability of vessels is a key pathophysiological process involved in ascites accumulation. TAMs are more likely to congregate at poorly vascularized sites (90) and promote angiogenesis in cancer by secreting VEGF and triggering revascularization (91). VEGF plays an essential role in angiogenesis and lymphangiogenesis by binding to receptors including kinase insert domain receptor (KDR)/fetal liver kinase (Flk)-1 and FMS-like tyrosine kinase (Flt)-1 (92). Han et al (93) found that TAM-derived chemokine CCL23 upregulates KDR/Flk-1 receptor expression in endothelial cells and promotes VEGF-mediated angiogenesis. VEGF levels are elevated significantly in OC ascites (94) and anti-VEGF treatment effectively suppressed tumor growth in a xenograft mouse model of OC and reduced ascites formation (95). Moreover, VEGF-A, VEGF-C and VEGF-D secreted by macrophages are involved in lymphangiogenesis dysfunction. Blockade of the VEGF-A/C/D pathway significantly inhibits the formation of chylous ascites in advanced OC mice (96). However, M2 macrophages downregulate the expression of very late antigen-4 (VLA4) when co-cultured with endothelial cells, decrease the levels of vascular cell adhesion molecule 1 (VCAM1) in endothelial cells and downregulate RAS-related C3 botulinum substrate 1 and reactive oxygen species, which resulted in decreased phosphorylation of proline-rich tyrosine kinase 2 and VE-cadherin. Therefore, M2 macrophages enhanced adhesion of endothelial cells and induced hypopermeability. Moreover, targeting the VLA4/VCAM1 axis enhances vascular integrity and eliminated the formation of ascites in vivo (97).

TAM recruitment into ascites

MA in OC usually contain abundant TAMs, and the prognosis is poor (98). The mass recruitment of macrophages is associated with numerous factors, for example, chemokines CCL2 and macrophage colony-stimulating factor (M-CSF), which serve a key role in the recruitment of inflammatory monocytes to tumor sites and differentiation into TAMs. Blockade of CSF1R signaling, a key factor for macrophage recruitment, reduced the infiltration of macrophages, protected against vascular permeability by normalizing disorganized peritoneal vasculature and notably decreased ascites volume (99). Ubiquitin protein ligase E3 component n-recognin 5 (UBR5), is amplified and overexpressed in many types of cancers, particularly in OC (100). High expression of UBR5 in OC is associated with poor prognosis (101). High expression of UBR5 induces high expression levels of CCL12 and M-CSF, which recruit TAMs to facilitate spheroid formation. In mice, tumor cells with UBR5 overexpression are more invasive and lead to rapid death (18). Periostin (POSTN) produced by OC cells could enhance the recruitment of macrophages, which produced transforming growth factor β (TGF-β) to promote the production of POSTN by OC cells. Blocking this positive feedback pathway may decrease migration of macrophages to ascites (102). CD276 (B7-H3), a transmembrane protein, is an immune checkpoint member of the B7 family. Tumor-expressed CD276 contributes to macrophage recruitment in spheroids (103). Besides tumor cells attracting TAMs by releasing chemokines and cytokines, the TAMs themselves produce chemotactic mediators, such as CCL5, CXCL8, IL-1 receptor antagonist, CCL18, CXCL2 and CXCL3, which also contribute to monocyte/macrophage recruitment (104). Apoptosis signal-regulating kinase 1 (ASK1) belongs to MAP3K family, activating MAP2K-JNK/p38 cascades (105). Yin et al (106) found that ASK1 deficiency in vascular endothelium cells, but not TAMs, attenuated spheroid formation and peritoneal implantation in orthotopic OC mice. Mechanistically, ASK1 promoted macrophage transmigration via degradation of endothelial junction protein VE-cadherin. Pharmacological ASK1 inhibitor decreases tumor-induced vascular leakage, macrophage infiltration and tumor growth in vivo (105).

TAMs polarization

Due to the plasticity of macrophages, undifferentiated macrophages (M0) can be polarized into two types, M1 and M2, which are distinguished by surface receptor expression, secretion pattern and function (107). The terms M1 and M2 were proposed by Mills et al (108) based on differences in arginine metabolism in macrophages from C57BL/6 and BALB/c mice, the effects of which are associated with differences between T helper (Th)1 and Th2 cell response. M1 macrophages produce a large number of pro-inflammatory cytokines under stimulation of Th1 cytokines such as IFN-γ and toll-like receptor (TLR) agonists such as lipopolysaccharide (LPS), and serve an essential role in anti-tumor response. M2 macrophages are stimulated by Th2 cytokines such as IL-4, IL-10 and TGF-β, promoting angiogenesis, and tissue repair (109). Due to the diversity of stimuli, M2 macrophages are further divided into M2a (IL-4 and IL-13), M2b (immune complex and LPS/IL-1), M2c (glucocorticoids, IL-10, TGF-β) and M2d (adenosine A2A receptor agonists and LPS) subtypes (109,110). Although the definition of M1-M2 macrophages provides a simplified paradigm for studying macrophage phenotype and function, this may oversimplify the complexity and diversity of macrophages, which exhibit mixed or unique phenotypes in many pathological conditions (111,112).

In the TME, polarized macrophages are known as TAMs. TAMs are composed of heterogeneous subpopulations, including M1 and M2 macrophages. TAMs predominantly express M2 macrophage markers and cytokines, such as CD206, CD163 and IL-10, and exhibit pro-tumor effects, and are therefore referred to as M2-like TAMs (113,114). By contrast, few TAMs in the TME express CD86 and CD80 markers and are referred to as M1-like TAMs, which typically exhibit anti-tumor effects (115,116). The ratio of M1/M2 is a prognostic indicator for OC. Patients with high M1/M2 ratio have a significantly longer overall and progression-free survival and platinum-free interval than patients with low M1/M2 (117). Plasticity is a key feature of macrophages. The phenotype of polarized M2-like TAMs can be reversed to M1-like TAMs to some extent (118). Therefore, shifting M2- to M1-like TAMs, rather than depleting TAMs, may serve as a treatment for cancer.

TAMs promote spheroid formation

In ascites of OC, TAMs either float alone or are present in the center of spheroids encircled by cancer cells, primarily displaying M2-like phenotype with high expression of CD163 and CD206 (17). CD163+ TAM is rarely found outside the spheroids (119). TAMs participate in formation of spheroids, and are associated with prognosis of OC. CD68, a transmembrane glycoprotein that is widely expressed in monocytes, is considered a marker of TAMs in the TME (120). OC cases with high percentages of CD68+ TAMs (>14.5%) in spheroids have a significantly lower 5-year overall survival than those with low proportion (<14.5%) of CD68+ TAMs (17). In 3D co-culture system, CCL18/zinc finger E-box binding protein 1 (ZEB1)/M-CSF axis facilitates spheroid formation. During the spheroid formation stage, OC cells secretes M-CSF to induce TAMs to M2 polarization, and M2-like TAMs induce EMT of cancer cells, characterized by increased expression of mesenchymal markers (including ZEB1, SNAIL and TWIST) and decreased expression of E-cadherin. Mechanistically, TAMs release chemokine CCL18, which interacts with chemokine receptor 8 (CCR8) on the surface of OC cells, subsequently increasing the expression of ZEB1, a transcriptional factor which binds the promoter of M-CSF, and enhanced M-CSF expression. Overexpression of ZEB1 in OC cells promotes cancer cell-TAM spheroid formation in vitro and in mice (16). Since EMT is a key factor involved in invasion, metastasis and chemotherapy resistance (121), the CCL18/ZEB1/M-CSF feedback loop between OC cells and TAMs not only promoted formation of spheroids in ascites, but also led to faster and earlier transcoelomic metastasis of OC (16).

TAMs can secrete various cytokines which are essential for tumor cell proliferation and survival. EGF is one of these cytokines that form homodimers or heterodimers on the cell surface and mediate cell proliferation signal transduction. TAMs are key cellular sources of EGF secretion in tumor tissues (15). EGF/EGFR signaling between TAMs and cancer cells was essential for spheroid formation. Within large spheroids, TAMs displaying M2-polarization markers (including CD163, CD206 and arginase 1) are located in the center of spheroids and surrounded by EGFR+ cancer cells. TAM-derived EGF activated EGFR on cancer cells, then increases expression of VEGF-C and VEGFR3 in cancer cells. EGFR blockade with erlotinib may inhibit spheroid formation and transcoelomic metastasis in vivo (17). Moreover, EGF secreted by TAMs increases the expression of αMβ2 integrin in TAMs and ICAM-1 in cancer cells to facilitate adhesion between TAMs and cancer cells (17). Therefore, the EGF/EGFR/VEGF-C/VEGFR3/αMβ2/ICAM-1 signaling pathway serves a key role in OC progression (17).

TAMs and anoikis resistance

Once floating in ascites, OC cells need to resist to anoikis, and multicellular spherical cells are more resistant to anoikis compared with individual cancer cells (60). TAMs serve a key role in anoikis resistance. Centrally located TAMs promote spheroid formation to provide a structural support OC cells to evade anoikis (17). STAT3 is an essential signal transduction molecule at the intersection of numerous pro-tumor signaling pathways (122), as well as mediating macrophage induction to M2-poralization (123). TAMs protected OC cells against anoikis via releasing several soluble factors, such as IL-6 and IL-10, which activated the STAT3 signaling in cancer cells, and promoted cell proliferation and peritoneal dissemination (123). When co-cultured with macrophages in vitro, especially M2 macrophages stimulated by M-CSF, ovarian cell line SKOV3 cells exhibit activation of the STAT3 signaling pathway (123). Hence, TAMs promote OC cell survival though enhanced anoikis resistance.

TAMs and drug resistance

TAMs may promote spheroid resistance to chemotherapeutic drugs in ascites. In a 3D culture model of canine mammary gland tumor cell lines with or without macrophages, compared with homogeneous spheroids composed only of tumor cells, homogeneous spheroids composed of tumor cells and macrophages displayed increased cell viability when treated with doxorubicin. Compared with monolayer tumor cells, expression levels of VEGF, TGF-β, tumor necrosis factor-α-stimulated gene/protein-6 and drug resistance-related proteins such as P-glycoprotein and multidrug resistance-associated protein 1 are significantly increased in spheroids. Furthermore, doxorubicin-induced apoptosis and G2/M cell cycle arrest are decreased in the presence of tumor cells co-cultured with macrophages (69). CSCs are involved in tumorigenicity and drug resistance. TAMs regulated CSC activities by releasing milk-fat globule-epidermal growth factor-VIII, which activates STAT3 and Sonic hedgehog pathways in CSCs and contributes to their resistance to cisplatin in vivo (124). During the interaction between M2 macrophages and ovarian CSCs, paracrine Wnt is stimulated, which may enhance the aggressive phenotype of macrophages and cancer cells (125).

The presence of TAMs enhances drug resistance in OC spheroids, and TAMs can serve as a potential therapeutic target for treatment of OC. It is possible to reduce tumor cell resistance and improve cytotoxicity of drug therapy by blocking macrophage recruitment into the TME and resetting macrophage polarization (126). Recent studies revealed that blocking M2 macrophage polarization makes OC cells more sensitive to chemotherapeutic drugs including cisplatin and poly (ADP-ribose) polymerase inhibitors in vitro and in vivo (127-129).

TAMs and immune escape

CD47-signal receptor protein-α (SIRPα) is the main innate immune checkpoint between macrophages and cancer cells. CD47 on the surface of tumor cells can release the signal 'don't eat me' by binding to the SIRPα receptor on the surface of macrophages, helping tumor cells to evade immune killing (130). Blocking the CD47/SIRPα signaling pathway can effectively promote phagocytosis of tumor cells by macrophages in vitro and in vivo (131). Blocking CD47 signaling with an oncolytic adenovirus carrying a SIRPα-IgG1Fc fusion gene (SG635-SF) significantly increases macrophage infiltration into the tumor and suppresses tumor growth in OC mice (132).

In the TME, TAMs have a markedly immunosuppressive effect on adaptive immune cells by releasing many cytokines, chemokines and enzymes, such as IL-10, TGF-β1, CCL12 and Arg-1. IL-10 causes naive CD4+ T cells to differentiate into Th2 cells, which suppress adaptive immunity, to allow malignant cells to escape immune surveillance (133). TGF-β1 may suppress T cell expansion through Smad3-dependent and -independent pathways (134). CCL22 secreted by TAMs establishes a chemokine gradient to induce regulatory T (Treg) cell migration into the local microenvironment, thereby increasing the proportion of Tregs (135). Tregs exist in ascites abundantly and accelerate tumor growth and progression via suppressing anti-tumor immunity. Macrophage-derived CCL23 induces CD8+ T cell exhaustion by upregulating molecules related to immune checkpoints, including cytotoxic T lymphocyte-associated antigen-4 (CTLA4), T cell immunoreceptor with immunoglobulin and immune receptor tyrosine-based inhibitory domain (TIGIT), T cell immunoglobulin and mucin domain 3 (TIM-3) and T cell immunoglobulin and mucin domain 3 (LAG-3) (136). TAMs express programmed cell death ligand 1 (PD-L1), PD-L2, CD80, and CD86, which restrict CD8+ T cell activation by binding to their receptors, PD1 and CTLA4 (137). B7-H3 not only serves as a costimulatory molecule that modulates T cell function directly (138), but also serves as an immune checkpoint involved in indirect T cell suppression via the CCL2-CCR2-M2 macrophage axis (139). B7-H4 is a recently identified B7 family molecule (140). A subpopulation of macrophages in OC ascites express B7-H4, a costimulatory molecule which decreases the proliferation and cytokine production of T cells (141).

TAMs promote spheroid peritoneal dissemination

TAMs accelerate tumor metastasis by promoting expression of peritoneal mesothelial cell adhesion molecules and releasing growth factors and invasive proteases (142,143). During the metastasis of OC, P-selectin is overexpressed on the surface of mesothelial cells and cancer cells attach to P-selectin through CD24, resulting in increased adhesion between cancer and mesothelial cells (142). In a co-culture model with cancer and mesothelial cells and M2 macrophages, expression of P-selectin was regulated by M2 macrophages which secrete macrophage inflammatory pro1tein-1β (MIP-1β) that activates CCR5/PI3K signaling in mesothelial cells, resulting in upregulation of P-selectin on the mesothelial cell surface. MIP-1β treatment increases P-selectin expression in peritoneal mesothelial cells of mice and enhances OC cell adhesion in vitro and in vivo (142). Analysis of samples from patients with high-grade serous OC confirmed increased MIP-1β and P-selectin expression, suggesting that TAMs in ascites secrete MIP-1β, which increases expression of P-selectin on the surface of mesothelial cells (142).

OC spheroids depart from the primary tumor and reattach throughout the peritoneal cavity. Once cancer cells are implanted at new sites, formation of metastatic lesions is dependent on disintegration of spheroids and subsequent spread across the ECM. Soluble factors such as FMS-like tyrosine kinase 3 ligand, leptin, or heparin-binding EGF derived from TAMs are responsible for spheroids spreading across the underlying ECM. The common signaling pathway of these soluble factors is Janus kinase 2 (JAK2)/STAT3 activation followed by MMP-9 mediated spreading (144). In addition, macrophages increase the invasion ability of cancer cells though TNF-α and NF-κB pathways. Macrophages can release VEGF to promote the dissemination of OC cells onto the peritoneum of mice. In mice, deletion of macrophages alone results in decreased expression of VEGF, inhibition of ascites formation and peritoneal metastasis (143). Moreover, TAMs also secrete EGF to facilitate OC metastasis via activation of EGFR/ERK signaling and suppression of long non-coding (lnc)RNA inhibiting metastasis (LIMT) expression. Following co-cultured with M2 macrophages, OC cells showed greater migration capacity, and these effects could be reversed by inhibiting EGF and overexpressing lncRNA LIMT (145).

TAMs as a target for treating OC

MA and spheroids are key for OC progression and recurrence, leading to relapse following classical therapy. TAMs play a critical role in spheroid formation and dissemination, as well as ascites formation. In addition, TAMs contribute to chemotherapeutic resistance and the suppression of the immune microenvironment characterized by enriched Tregs and exhausted CD8+ T cells, thereby promoting survival of cancer cells. Therefore, TAMs are a promising target in OC treatment. Strategies to target TAMs include blocking the recruitment of macrophages to the TME, shifting TAM polarization from M2 to M1 type, increasing phagocytosis of TAMs, blocking formation and intraperitoneal metastasis of TAM-tumor cell spheroids and improving chemotherapeutic sensitivity. Effects of TAM-based treatment for OC are summarized in Table I.

Table I

TAMs as a therapeutic target in OC.

Table I

TAMs as a therapeutic target in OC.

MechanismModelDrugFindingsStageFirst author, year(Refs.)
Inhibition of TAM recruitmentMouseGW2580 (M-CSFR inhibitor)GW2580, a M-CSF receptor kinase inhibitor, binds with M-CSFR on macrophages and monocytes, decreases the infiltration of M2 macrophages and significantly decreases the amount of ascitesPreclinicalMoughon, et al, 2015(99)
Cell (bone marrow-derived monocytes), mouse, humanTrabectedinTrabectedin inhibits recruitment of circulating monocytes into tumor tissue and results in macrophage depletion by inhibiting production of CCL2, thus inhibiting OC progressionClinicalGermano et al, 2013(146)
HumanCarlumab (CCL2 monoclonal antibody)Carlumab is well-tolerated with evidence of transient free CCL2 suppression and preliminary antitumor activity. A patient with OC achieved CA125 reduction >50% and RECIST SD for 10.5 monthsIb clinicalSandhu et al, 2013(147)
HumanPexidartinib (M-CSFR tyrosine kinase inhibitor)Combined with paclitaxel, 1/6 patients showed complete response (response duration 189 days) and 1/6 showed partial response (response duration 94 days)I clinicalWesolowski et al, 2019(148)
Shifting TAM polarization from M2 to M1 typeCell (murine epithelial ovarian cancer cell line ID8), mousePlerixafor (CXCR4 antagonist)Plerixafor downregulates the expression of CXCL12 and CXCR4 and it promoted macrophage polarization from M2 to M1. Compared with αPD-1 therapy, plerixafor + αPD-1 significantly inhibits tumor growth and prolongs survival of tumor-bearing micePreclinicalZeng et al, 2019(149)
Cell (murine RAW 264.7), mouse, humanPaclitaxelPaclitaxel inhibits the polarization of M2 macrophages induced by the IL4/STAT6 pathway and reprograms them into M1 macrophages via the TLR4/NF-κB pathway, exerting an antitumor effectClinicalWanderley et al, 2018(150)
Call (murine macrophage cell line Raw 264.7), mouse, HGSOC samplesNeferineNeferine exerts antiangiogenic effects in HGSOC primarily by inhibiting polarization of M2 macrophages. Neferine decreases Arg-1 expression in macrophages, but increased the expression of iNOS, a marker of M1 macrophagesPreclinicalZhang et al, 2018(151)
Cell (human acute monocytic leukemia THP-1), mouseInfusion of IRF5 mRNA and IKKβ nanoparticlesInfusion of IRF5 mRNA and IKKβ nanoparticles reverses the immunosuppressive and tumor-supporting state of TAMs, significantly decreases the density of M2 macrophages and reprograms them into M1 macrophages and promotes the expression of IL-12, IFN-γ and TNF-α.PreclinicalZhang et al, 2019(152)
Cell (murine Raw 264.7), mouseChloroquineChloroquine serves as an immunomodulator and mediates its anti-tumor efficacy by increasing lysosomal pH to reset TAMs from M2 to M1 phenotype, and decreases immunosuppressive infiltration of myeloid-derived suppressor cells and Tregs, thus enhancing antitumor T-cell immunityPreclinicalChen et al, 2018(153)
HumanLPSTAMs from ascites of patients with OC display. M2 phenotype. Upon TLR stimulation by LPS, TAMs acquire a classically activated functional phenotype (M1), release immunostimulatory cytokines (IL-12 and soluble IL-18) and efficiently trigger the cytolytic activity of NK cellsClinicalBellora et al, 2014(154)
Increased phagocytosis of TAMs by immune checkpoint inhibitionCell (human peripheral blood mononuclear cells), mouse, monkeyHuNb1-IgG4 (anti-CD47 nanobody)HuNb1-IgG4 enhances macrophage-mediated OC cell phagocytosis. Because of its low affinity with red blood cells, it does not cause platelet aggregation or hemagglutination.PreclinicalMa et al, 2020(155)
HumanHu5F9-G4 (CD47 antibody)Of 13 patients with OC, two showed partial responses lasting 5.2-9.2 monthsClinicalSikic et al, 2019(131)
Cell (human ovarian carcinoma SKOV3 and HO8910), mouseSG635-SF (oncolytic adenovirus carrying SIRP-IgG1 Fc fusion gene)SG635-SF blocks CD47 signaling in SK-OV3 and HO8910 OC cells expressing high levels of CD47. Macrophage infiltration into the tumor significantly increases and tumor cell killing is observed in xenograft tissuePreclinicalHuang et al, 2020(132)
Inhibition of spheroid formationMouseErlotinib (EGFR inhibitor)Pharmacological blockade of EGFR or antibody neutralization of ICAM-1 in TAMs inhibits spheroid formation and OC progressionPreclinicalYin et al, 2016(17)
Inhibition of spheroid spreading on peritoneumMouse Clodronate-containing liposomes Clodronate-containing liposomes deplete peritoneal macrophages, which decreases tumor progression, as assessed by ascites formation and peritoneal metastasis. Inflammation facilitates ovarian tumor metastasis, primarily mediated by macrophages, which may involve stromal vascular endothelial growth factor productionPreclinicalRobinson-Smith et al, 2007(143)
Improvement of chemotherapeutic sensitivityCell (human monocytes), mouseAntisense oligonucleotide targeting circITGB6circITGB6 stabilizes FGF9 RNA, which induces polarization of TAM toward the M2 phenotype, Antisense oligonucleotide targeting circITGB6 blocks M2 macrophage polarization and makes OC cells more sensitive to chemotherapeutic drug cisplatinPreclinicalLi et al, 2022(128)
Cell (human ovarian cancer UWB1.289), mouseSTING agonistsSTING agonists induce reprogramming of immunosuppressive myeloid cells, which inhibited TAM polarization toward M2 type and increases OC cell sensitivity to PARP inhibitorsPreclinicalDing et al, 2023(129)
Cell (murine macrophage cell line RAW 264.7), mouseTriptolide and miR-497Exosome-liposome hybrid nanoparticle codelivery of triptolide and miR-497 promotes dephosphorylation of the overactivated PI3K/AKT/mTOR signaling pathway and increases ROS generation and polarization of macrophages from M2 to M1, making cells more sensitive to cisplatin in vitro and in vivoPreclinicalLi et al, 2022(127)

[i] M-CSFR, macrophage colony-stimulating factor receptor; CCL2, Chemokine (C-C motif) ligand-2; CA125, carbohydrate antigen 125; RECIST, response evaluation criteria in solid tumors SD, stable disease; CXCR4, C-X-C chemokine receptor type 4; CXCL, C-X-C motif chemokine, αPD-1, anti-programmed death-1 antibody; TLR4, toll-like receptor 4, HGSOC, high-grade serous ovarian cancer; Arg-1, arginase-1; iNOS, inducible nitric oxide synthase; IRF5, interferon regulatory factor 5; IKKβ, IkappaB kinase-beta; TNF-α, tumor necrosis factor alpha; Tregs, regulatory T cells; TAMs, tumor-associated macrophages; LPS, lipopolysaccharide; NK cells, natural killer cells; SIRP, signal regulatory protein; EGFR, epidermal growth factor receptor; ICAM-1, intercellular adhesion molecule-1; circITGB6, circular RNA encoding integrin beta 6; FGF9, fibroblast growth factor 9; STING, stimulator of interferon genes; PARP, Poly ADP-ribose polymerase; miR-497, microRNA-497; PI3K, phosphoinositide 3-kinase; ROS, reactive oxygen species.

Conclusion

Cancer cells in OC ascites exist in two forms, individual cells and multicellular spheroids. Spheroids are divided into homospheroids and heterotypical spheroids, which contain tumor and non-tumor cells. The spheroids are involved in progression of OC, since they are more resistant to anoikis and chemotherapeutic drugs and are considered to be metastatic units. TAMs serve an essential role in OC progression. Recently, some groups demonstrated functions of TAMs in the formation of spheroids and dissemination (16,21,156). Although some researchers have revealed the discrete molecular mechanisms of spheroid formation, survival and metastasis (16,142), there are no data showing crosstalk between these pathways. Studies investigating the role of TAMs in OC spheroids typically use co-culture of TAMs and tumor cells in 3D culture model in the presence or absence of Matrigel or other scaffolds (16,157). Most studies have utilized OC cell lines and M2 macrophages to mimic OC cells and TAMs, respectively (16,142,144).These findings needed to be further clarified in patients. The majority of studies only co-cultured TAMs and tumor cells, and lack of other cellular components and non-cellular components present in the TME (16,144). These components are likely to influence TAM effects on the spheroids, making the results less convincing.

More molecules and signal pathways need to be investigated in spheroid formation, survival and metastasis, and the network among these molecules should be identified. Standardized methods are required for 3D culture model, such as description of size of spheroids, and presence or absence of scaffolds. Results obtained in primary cancer cells and TAMs from patients with OC are more persuasive than cell lines. Patient-derived organoids may mimic the TME. Cellular and non-cellular components in ascites can be separated from patients with OC and used to mimic the TME.

Availability of data and materials

Not applicable.

Authors' contributions

YL and YX designed the review. YL and HX wrote the manuscript. HZ and YX revised the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Abbreviations:

MA

malignant ascites

TAM

tumor-associated macrophage

ECM

extracellular matrix

EGFR

epidermal growth factor receptor

CAF

cancer-associated fibroblast

CSC

cancer stem cell

OC

ovarian cancer

M-CSF

macrophage-colony stimulating factor

UBR5

ubiquitin protein ligase E3 component n-recognin 5

EZH2

enhancer of zeste homolog 2

CAM

cancer-associated mesothelial cell

TME

tumor microenvironment

VEGF

vascular endothelial growth factor

VCAM1

vascular cell adhesion molecule 1

POSTN

periostin

ASK1

apoptosis signal-regulating kinase 1

TLR

toll-like receptor

Acknowledgments

Not applicable.

Funding

The present study was supported by National Natural Science Foundation of China (grant no. 81602303) and Start Fund of First Affiliated Hospital of Yangtze University (grant no. 2022DIF01).

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Bast RC, Han CY, Lu Z and Lu KH: Next steps in the early detection of ovarian cancer. Commun Med (Lond). 1:362021. View Article : Google Scholar : PubMed/NCBI

3 

Yang L, Xie HJ, Li YY, Wang X, Liu XX and Mai J: Molecular mechanisms of platinum-based chemotherapy resistance in ovarian cancer (review). Oncol Rep. 47:822022. View Article : Google Scholar

4 

Almeida-Nunes DL, Mendes-Frias A, Silvestre R, Dinis-Oliveira RJ and Ricardo S: Immune tumor microenvironment in ovarian cancer ascites. Int J Mol Sci. 23:106922022. View Article : Google Scholar : PubMed/NCBI

5 

Huang H, Li YJ, Lan CY, Huang QD, Feng YL, Huang YW and Liu JH: Clinical significance of ascites in epithelial ovarian cancer. Neoplasma. 60:546–552. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Kim S, Kim B and Song YS: Ascites modulates cancer cell behavior, contributing to tumor heterogeneity in ovarian cancer. Cancer Sci. 107:1173–1178. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Quan Q, Zhou S, Liu Y, Yin W, Liao Q, Ren S, Zhang F, Meng Y and Mu X: Relationship between ascites volume and clinical outcomes in epithelial ovarian cancer. J Obstet Gynaecol Res. 47:1527–1535. 2021. View Article : Google Scholar : PubMed/NCBI

8 

Cavazzoni E, Bugiantella W, Graziosi L, Franceschini MS and Donini A: Malignant ascites: Pathophysiology and treatment. Int J Clin Oncol. 18:1–9. 2013. View Article : Google Scholar

9 

Yeung TL, Leung CS, Yip KP, Au Yeung CL, Wong ST and Mok SC: Cellular and molecular processes in ovarian cancer metastasis. A review in the theme: Cell and molecular processes in cancer metastasis. Am J Physiol Cell Physiol. 309:C444–C456. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Al Habyan S, Kalos C, Szymborski J and McCaffrey L: Multicellular detachment generates metastatic spheroids during intra-abdominal dissemination in epithelial ovarian cancer. Oncogene. 37:5127–5135. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Ahmed N and Stenvers KL: Getting to know ovarian cancer ascites: Opportunities for targeted therapy-based translational research. Front Oncol. 3:2562013. View Article : Google Scholar : PubMed/NCBI

12 

Dhaliwal D and Shepherd TG: Molecular and cellular mechanisms controlling integrin-mediated cell adhesion and tumor progression in ovarian cancer metastasis: A review. Clin Exp Metastasis. 39:291–301. 2022. View Article : Google Scholar :

13 

Worzfeld T, Pogge von Strandmann E, Huber M, Adhikary T, Wagner U, Reinartz S and Müller R: The unique molecular and cellular microenvironment of ovarian cancer. Front Oncol. 7:242017. View Article : Google Scholar : PubMed/NCBI

14 

Gao Q, Yang Z, Xu S, Li X, Yang X, Jin P, Liu Y, Zhou X, Zhang T, Gong C, et al: Heterotypic CAF-tumor spheroids promote early peritoneal metastatis of ovarian cancer. J Exp Med. 216:688–703. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Pan Y, Yu Y, Wang X and Zhang T: Tumor-associated macrophages in tumor immunity. Front Immunol. 11:5830842020. View Article : Google Scholar : PubMed/NCBI

16 

Long L, Hu Y, Long T, Lu X, Tuo Y, Li Y and Ke Z: Tumor-associated macrophages induced spheroid formation by CCL18-ZEB1-M-CSF feedback loop to promote transcoelomic metastasis of ovarian cancer. J Immunother Cancer. 9:e0039732021. View Article : Google Scholar

17 

Yin M, Li X, Tan S, Zhou HJ, Ji W, Bellone S, Xu X, Zhang H, Santin AD, Lou G and Min W: Tumor-associated macrophages drive spheroid formation during early transcoelomic metastasis of ovarian cancer. J Clin Invest. 126:4157–4173. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Song M, Yeku OO, Rafiq S, Purdon T, Dong X, Zhu L, Zhang T, Wang H, Yu Z, Mai J, et al: Tumor derived UBR5 promotes ovarian cancer growth and metastasis through inducing immunosuppressive macrophages. Nat Commun. 11:62982020. View Article : Google Scholar : PubMed/NCBI

19 

El-Arabey AA, Alkhalil SS, Al-Shouli ST, Awadalla ME, Alhamdi HW, Almanaa TN, Mohamed SSEM and Abdalla M: Revisiting macrophages in ovarian cancer microenvironment: Development, function and interaction. Med Oncol. 40:1422023. View Article : Google Scholar : PubMed/NCBI

20 

Larionova I, Tuguzbaeva G, Ponomaryova A, Stakheyeva M, Cherdyntseva N, Pavlov V, Choinzonov E and Kzhyshkowska J: Tumor-associated macrophages in human breast, colorectal, lung, ovarian and prostate cancers. Front Oncol. 10:5665112020. View Article : Google Scholar : PubMed/NCBI

21 

Miyamoto T, Murphy B and Zhang N: Intraperitoneal metastasis of ovarian cancer: New insights on resident macrophages in the peritoneal cavity. Front Immunol. 14:11046942023. View Article : Google Scholar : PubMed/NCBI

22 

Yin M, Shen J, Yu S, Fei J, Zhu X, Zhao J, Zhai L, Sadhukhan A and Zhou J: Tumor-associated macrophages (TAMs): A critical activator in ovarian cancer metastasis. Onco Targets Ther. 12:8687–8699. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Jazwinska DE, Kulawiec DG and Zervantonakis IK: Cancer-mesothelial and cancer-macrophage interactions in the ovarian cancer microenvironment. Am J Physiol Cell Physiol. 325:C721–C730. 2023. View Article : Google Scholar : PubMed/NCBI

24 

Klymenko Y, Johnson J, Bos B, Lombard R, Campbell L, Loughran E and Stack MS: Heterogeneous cadherin expression and multicellular aggregate dynamics in ovarian cancer dissemination. Neoplasia. 19:549–563. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Ford CE, Werner B, Hacker NF and Warton K: The untapped potential of ascites in ovarian cancer research and treatment. Br J Cancer. 123:9–16. 2020. View Article : Google Scholar : PubMed/NCBI

26 

Habanjar O, Diab-Assaf M, Caldefie-Chezet F and Delort L: 3D cell culture systems: Tumor application, advantages, and disadvantages. Int J Mol Sci. 22:122002021. View Article : Google Scholar : PubMed/NCBI

27 

Matte I, Legault CM, Garde-Granger P, Laplante C, Bessette P, Rancourt C and Piché A: Mesothelial cells interact with tumor cells for the formation of ovarian cancer multicellular spheroids in peritoneal effusions. Clin Exp Metastasis. 33:839–852. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Xu S, Yang Y, Dong L, Qiu W, Yang L, Wang X and Liu L: Construction and characteristics of an E-cadherin-related three-dimensional suspension growth model of ovarian cancer. Sci Rep. 4:56462014. View Article : Google Scholar : PubMed/NCBI

29 

Sawada K, Mitra AK, Radjabi AR, Bhaskar V, Kistner EO, Tretiakova M, Jagadeeswaran S, Montag A, Becker A, Kenny HA, et al: Loss of E-cadherin promotes ovarian cancer metastasis via alpha 5-integrin, which is a therapeutic target. Cancer Res. 68:2329–2339. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Casey RC, Burleson KM, Skubitz KM, Pambuccian SE, Oegema TR Jr, Ruff LE and Skubitz AP: Beta 1-integrins regulate the formation and adhesion of ovarian carcinoma multicellular spheroids. Am J Pathol. 159:2071–2080. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Han Q, Huang B, Huang Z, Cai J, Gong L, Zhang Y, Jiang J, Dong W and Wang Z: Tumor cell-fibroblast heterotypic aggregates in malignant ascites of patients with ovarian cancer. Int J Mol Med. 44:2245–2255. 2019.PubMed/NCBI

32 

Hassn Mesrati M, Syafruddin SE, Mohtar MA and Syahir A: CD44: A multifunctional mediator of cancer progression. Biomolecules. 11:18502021. View Article : Google Scholar : PubMed/NCBI

33 

Chen MW, Yang ST, Chien MH, Hua KT, Wu CJ, Hsiao SM, Lin H, Hsiao M, Su JL and Wei LH: The STAT3-miRNA-92-Wnt signaling pathway regulates spheroid formation and malignant progression in ovarian cancer. Cancer Res. 77:1955–1967. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Li SS, Ma J and Wong AST: Chemoresistance in ovarian cancer: Exploiting cancer stem cell metabolism. J Gynecol Oncol. 29:e322018. View Article : Google Scholar : PubMed/NCBI

35 

Casagrande N, Borghese C, Agostini F, Durante C, Mazzucato M, Colombatti A and Aldinucci D: In ovarian cancer multicellular spheroids, platelet releasate promotes growth, expansion of ALDH+ and CD133+ cancer stem cells, and protection against the cytotoxic effects of cisplatin, carboplatin and paclitaxel. Int J Mol Sci. 22:30192021. View Article : Google Scholar : PubMed/NCBI

36 

Condello S, Morgan CA, Nagdas S, Cao L, Turek J, Hurley TD and Matei D: β-Catenin-regulated ALDH1A1 is a target in ovarian cancer spheroids. Oncogene. 34:2297–2308. 2015. View Article : Google Scholar

37 

Cui TX, Kryczek I, Zhao L, Zhao E, Kuick R, Roh MH, Vatan L, Szeliga W, Mao Y, Thomas DG, et al: Myeloid-derived suppressor cells enhance stemness of cancer cells by inducing microRNA101 and suppressing the corepressor CtBP2. Immunity. 39:611–621. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Suganuma T, Ino K, Shibata K, Kajiyama H, Nagasaka T, Mizutani S and Kikkawa F: Functional expression of the angiotensin II type 1 receptor in human ovarian carcinoma cells and its blockade therapy resulting in suppression of tumor invasion, angiogenesis, and peritoneal dissemination. Clin Cancer Res. 11:2686–2694. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Zhang Q, Yu S, Lam MMT, Poon TCW, Sun L, Jiao Y, Wong AST and Lee LTO: Angiotensin II promotes ovarian cancer spheroid formation and metastasis by upregulation of lipid desaturation and suppression of endoplasmic reticulum stress. J Exp Clin Cancer Res. 38:1162019. View Article : Google Scholar : PubMed/NCBI

40 

Adeshakin FO, Adeshakin AO, Afolabi LO, Yan D, Zhang G and Wan X: Mechanisms for modulating anoikis resistance in cancer and the relevance of metabolic reprogramming. Front Oncol. 11:6265772021. View Article : Google Scholar : PubMed/NCBI

41 

Frisch SM and Screaton RA: Anoikis mechanisms. Curr Opin Cell Biol. 13:555–562. 2001. View Article : Google Scholar : PubMed/NCBI

42 

Grossmann J: Molecular mechanisms of 'detachment-induced apoptosis-Anoikis'. Apoptosis. 7:247–260. 2002. View Article : Google Scholar : PubMed/NCBI

43 

Taddei ML, Giannoni E, Fiaschi T and Chiarugi P: Anoikis: An emerging hallmark in health and diseases. J Pathol. 226:380–393. 2012. View Article : Google Scholar

44 

Cai Q, Yan L and Xu Y: Anoikis resistance is a critical feature of highly aggressive ovarian cancer cells. Oncogene. 34:3315–3324. 2015. View Article : Google Scholar :

45 

Cancer Genome Atlas Research Network: Integrated genomic analyses of ovarian carcinoma. Nature. 474:609–615. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Park JT, Shih IeM and Wang TL: Identification of Pbx1, a potential oncogene, as a Notch3 target gene in ovarian cancer. Cancer Res. 68:8852–8860. 2008. View Article : Google Scholar : PubMed/NCBI

47 

Brown CW, Brodsky AS and Freiman RN: Notch3 overexpression promotes anoikis resistance in epithelial ovarian cancer via upregulation of COL4A2. Mol Cancer Res. 13:78–85. 2015. View Article : Google Scholar :

48 

Tang MKS, Zhou HY, Yam JW and Wong AS: c-Met overexpression contributes to the acquired apoptotic resistance of nonadherent ovarian cancer cells through a cross talk mediated by phosphatidylinositol 3-kinase and extracellular signal-regulated kinase 1/2. Neoplasia. 12:128–138. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Lopez A, Reyna DE, Gitego N, Kopp F, Zhou H, Miranda-Roman MA, Nordstrøm LU, Narayanagari SR, Chi P, Vilar E, et al: Co-targeting of BAX and BCL-XL proteins broadly overcomes resistance to apoptosis in cancer. Nat Commun. 13:11992022. View Article : Google Scholar : PubMed/NCBI

50 

Mei H, Nakatsu MN, Baclagon ER and Deng SX: Frizzled 7 maintains the undifferentiated state of human limbal stem/progenitor cells. Stem Cells. 32:938–945. 2014. View Article : Google Scholar

51 

Condello S, Sima L, Ivan C, Cardenas H, Schiltz G, Mishra RK and Matei D: Tissue tranglutaminase regulates interactions between ovarian cancer stem cells and the tumor niche. Cancer Res. 78:2990–3001. 2018. View Article : Google Scholar : PubMed/NCBI

52 

Qin Q, Xu Y, He T, Qin C and Xu J: Normal and disease-related biological functions of Twist1 and underlying molecular mechanisms. Cell Res. 22:90–106. 2012. View Article : Google Scholar :

53 

Tan M, Asad M, Heong V, Wong MK, Tan TZ, Ye J, Kuay KT, Thiery JP, Scott C and Huang RY: The FZD7-TWIST1 axis is responsible for anoikis resistance and tumorigenesis in ovarian carcinoma. Mol Oncol. 13:757–780. 2019. View Article : Google Scholar :

54 

Motohara T, Masuda K, Morotti M, Zheng Y, El-Sahhar S, Chong KY, Wietek N, Alsaadi A, Carrami EM, Hu Z, et al: An evolving story of the metastatic voyage of ovarian cancer cells: Cellular and molecular orchestration of the adipose-rich metastatic microenvironment. Oncogene. 38:2885–2898. 2019. View Article : Google Scholar :

55 

Dai ZY, Jin SM, Luo HQ, Leng HL and Fang JD: LncRNA HOTAIR regulates anoikis-resistance capacity and spheroid formation of ovarian cancer cells by recruiting EZH2 and influencing H3K27 methylation. Neoplasma. 68:509–518. 2021. View Article : Google Scholar : PubMed/NCBI

56 

Zhang L, Huang J, Yang N, Greshock J, Liang S, Hasegawa K, Giannakakis A, Poulos N, O'Brien-Jenkins A, Katsaros D, et al: Integrative genomic analysis of phosphatidylinositol 3′-kinase family identifies PIK3R3 as a potential therapeutic target in epithelial ovarian cancer. Clin Cancer Res. 13:5314–5321. 2007. View Article : Google Scholar : PubMed/NCBI

57 

Dong L and Hui L: HOTAIR promotes proliferation, migration, and invasion of ovarian cancer SKOV3 cells through regulating PIK3R3. Med Sci Monit. 22:325–331. 2016. View Article : Google Scholar : PubMed/NCBI

58 

Dolinschek R, Hingerl J, Benge A, Zafiu C, Schüren E, Ehmoser EK, Lössner D and Reuning U: Constitutive activation of integrin αvβ3 contributes to anoikis resistance of ovarian cancer cells. Mol Oncol. 15:503–522. 2021. View Article : Google Scholar

59 

Yu X, Liu L, Cai B, He Y and Wan X: Suppression of anoikis by the neurotrophic receptor TrkB in human ovarian cancer. Cancer Sci. 99:543–552. 2008. View Article : Google Scholar : PubMed/NCBI

60 

Carduner L, Picot CR, Leroy-Dudal J, Blay L, Kellouche S and Carreiras F: Cell cycle arrest or survival signaling through αv integrins, activation of PKC and ERK1/2 lead to anoikis resistance of ovarian cancer spheroids. Exp Cell Res. 320:329–342. 2014. View Article : Google Scholar

61 

Kim S, Kim S, Kim J, Kim B, Kim SI, Kim MA, Kwon S and Song YS: Evaluating tumor evolution via genomic profiling of individual tumor spheroids in a malignant ascites. Sci Rep. 8:127242018. View Article : Google Scholar : PubMed/NCBI

62 

Allen HJ, Porter C, Gamarra M, Piver MS and Johnson EA: Isolation and morphologic characterization of human ovarian carcinoma cell clusters present in effusions. Exp Cell Biol. 55:194–208. 1987.PubMed/NCBI

63 

Azharuddin M, Roberg K, Dhara AK, Jain MV, Darcy P, Hinkula J, Slater NKH and Patra HK: Dissecting multi drug resistance in head and neck cancer cells using multicellular tumor spheroids. Sci Rep. 9:200662019. View Article : Google Scholar : PubMed/NCBI

64 

Świerczewska M, Sterzyńska K, Ruciński M, Andrzejewska M, Nowicki M and Januchowski R: The response and resistance to drugs in ovarian cancer cell lines in 2D monolayers and 3D spheroids. Biomed Pharmacother. 165:1151522023. View Article : Google Scholar : PubMed/NCBI

65 

Sun Y, Li S, Yang L, Zhang D, Zhao Z, Gao J and Liu L: CDC25A facilitates chemo-resistance in ovarian cancer multicellular spheroids by promoting E-cadherin expression and arresting cell cycles. J Cancer. 10:2874–2884. 2019. View Article : Google Scholar : PubMed/NCBI

66 

Shen T and Huang S: The role of Cdc25A in the regulation of cell proliferation and apoptosis. Anticancer Agents Med Chem. 12:631–639. 2012. View Article : Google Scholar : PubMed/NCBI

67 

Broggini M, Buraggi G, Brenna A, Riva L, Codegoni AM, Torri V, Lissoni AA, Mangioni C and D'Incalci M: Cell cycle-related phosphatases CDC25A and B expression correlates with survival in ovarian cancer patients. Anticancer Res. 20:4835–4840. 2000.

68 

Green SK, Francia G, Isidoro C and Kerbel RS: Antiadhesive antibodies targeting E-cadherin sensitize multicellular tumor spheroids to chemotherapy in vitro. Mol Cancer Ther. 3:149–159. 2004. View Article : Google Scholar : PubMed/NCBI

69 

Lim GH, An JH, Park SM, Youn GH, Oh YI, Seo KW and Youn HY: Macrophage induces anti-cancer drug resistance in canine mammary gland tumor spheroid. Sci Rep. 13:103942023. View Article : Google Scholar : PubMed/NCBI

70 

Makhija S, Taylor DD, Gibb RK and Gerçel-Taylor C: Taxol-induced bcl-2 phosphorylation in ovarian cancer cell monolayer and spheroids. Int J Oncol. 14:515–521. 1999.PubMed/NCBI

71 

Yvon AM, Wadsworth P and Jordan MA: Taxol suppresses dynamics of individual microtubules in living human tumor cells. Mol Biol Cell. 10:947–959. 1999. View Article : Google Scholar : PubMed/NCBI

72 

Makovec T: Cisplatin and beyond: Molecular mechanisms of action and drug resistance development in cancer chemotherapy. Radiol Oncol. 53:148–158. 2019. View Article : Google Scholar : PubMed/NCBI

73 

Kwon MJ and Shin YK: Regulation of ovarian cancer stem cells or tumor-initiating cells. Int J Mol Sci. 14:6624–6648. 2013. View Article : Google Scholar : PubMed/NCBI

74 

Liao J, Qian F, Tchabo N, Mhawech-Fauceglia P, Beck A, Qian Z, Wang X, Huss WJ, Lele SB, Morrison CD and Odunsi K: Ovarian cancer spheroid cells with stem cell-like properties contribute to tumor generation, metastasis and chemotherapy resistance through hypoxia-resistant metabolism. PLoS One. 9:e849412014. View Article : Google Scholar : PubMed/NCBI

75 

McAuliffe SM, Morgan SL, Wyant GA, Tran LT, Muto KW, Chen YS, Chin KT, Partridge JC, Poole BB, Cheng KH, et al: Targeting Notch, a key pathway for ovarian cancer stem cells, sensitizes tumors to platinum therapy. Proc Natl Acad Sci USA. 109:E2939–E2948. 2012. View Article : Google Scholar : PubMed/NCBI

76 

Kang H, Jeong JY, Song JY, Kim TH, Kim G, Huh JH, Kwon AY, Jung SG and An HJ: Notch3-specific inhibition using siRNA knockdown or GSI sensitizes paclitaxel-resistant ovarian cancer cells. Mol Carcinog. 55:1196–1209. 2016. View Article : Google Scholar

77 

van Baal JOAM, Van de Vijver KK, Nieuwland R, van Noorden CJF, van Driel WJ, Sturk A, Kenter GG, Rikkert LG and Lok CAR: The histophysiology and pathophysiology of the peritoneum. Tissue Cell. 49:95–105. 2017. View Article : Google Scholar

78 

Nakamura M, Ono YJ, Kanemura M, Tanaka T, Hayashi M, Terai Y and Ohmichi M: Hepatocyte growth factor secreted by ovarian cancer cells stimulates peritoneal implantation via the mesothelial-mesenchymal transition of the peritoneum. Gynecol Oncol. 139:345–354. 2015. View Article : Google Scholar : PubMed/NCBI

79 

Yousefi M, Dehghani S, Nosrati R, Ghanei M, Salmaninejad A, Rajaie S, Hasanzadeh M and Pasdar A: Current insights into the metastasis of epithelial ovarian cancer-hopes and hurdles. Cell Oncol (Dordr). 43:515–538. 2020. View Article : Google Scholar : PubMed/NCBI

80 

Cui L, Johkura K, Liang Y, Teng R, Ogiwara N, Okouchi Y, Asanuma K and Sasaki K: Biodefense function of omental milky spots through cell adhesion molecules and leukocyte proliferation. Cell Tissue Res. 310:321–330. 2002. View Article : Google Scholar : PubMed/NCBI

81 

Niedbala MJ, Crickard K and Bernacki RJ: Interactions of human ovarian tumor cells with human mesothelial cells grown on extracellular matrix. An in vitro model system for studying tumor cell adhesion and invasion. Exp Cell Res. 160:499–513. 1985. View Article : Google Scholar : PubMed/NCBI

82 

Iwanicki MP, Davidowitz RA, Ng MR, Besser A, Muranen T, Merritt M, Danuser G, Ince TA and Brugge JS: Ovarian cancer spheroids use myosin-generated force to clear the mesothelium. Cancer Discov. 1:144–157. 2011. View Article : Google Scholar

83 

Cannistra SA, Kansas GS, Niloff J, DeFranzo B, Kim Y and Ottensmeier C: Binding of ovarian cancer cells to peritoneal mesothelium in vitro is partly mediated by CD44H. Cancer Res. 53:3830–3838. 1993.PubMed/NCBI

84 

Witz CA, Montoya-Rodriguez IA, Cho S, Centonze VE, Bonewald LF and Schenken RS: Composition of the extracellular matrix of the peritoneum. J Soc Gynecol Investig. 8:299–304. 2001. View Article : Google Scholar : PubMed/NCBI

85 

Burleson KM, Casey RC, Skubitz KM, Pambuccian SE, Oegema TR Jr and Skubitz AP: Ovarian carcinoma ascites spheroids adhere to extracellular matrix components and mesothelial cell monolayers. Gynecol Oncol. 93:170–181. 2004. View Article : Google Scholar : PubMed/NCBI

86 

Burleson KM, Boente MP, Pambuccian SE and Skubitz AP: Disaggregation and invasion of ovarian carcinoma ascites spheroids. J Transl Med. 4:62006. View Article : Google Scholar : PubMed/NCBI

87 

Moser TL, Pizzo SV, Bafetti LM, Fishman DA and Stack MS: Evidence for preferential adhesion of ovarian epithelial carcinoma cells to type I collagen mediated by the alpha2beta1 integrin. Int J Cancer. 67:695–701. 1996. View Article : Google Scholar : PubMed/NCBI

88 

Shield K, Riley C, Quinn MA, Rice GE, Ackland ML and Ahmed N: Alpha2beta1 integrin affects metastatic potential of ovarian carcinoma spheroids by supporting disaggregation and proteolysis. J Carcinog. 6:112007. View Article : Google Scholar : PubMed/NCBI

89 

Gupta V, Yull F and Khabele D: Bipolar tumor-associated macrophages in ovarian cancer as targets for therapy. Cancers (Basel). 10:3662018. View Article : Google Scholar : PubMed/NCBI

90 

Leek RD, Lewis CE, Whitehouse R, Greenall M, Clarke J and Harris AL: Association of macrophage infiltration with angiogenesis and prognosis in invasive breast carcinoma. Cancer Res. 56:4625–4629. 1996.PubMed/NCBI

91 

Lewis JS, Landers RJ, Underwood JC, Harris AL and Lewis CE: Expression of vascular endothelial growth factor by macrophages is up-regulated in poorly vascularized areas of breast carcinomas. J Pathol. 192:150–158. 2000. View Article : Google Scholar : PubMed/NCBI

92 

Shibuya M: Vascular endothelial growth factor and its receptor system: Physiological functions in angiogenesis and pathological roles in various diseases. J Biochem. 153:13–19. 2013. View Article : Google Scholar

93 

Han KY, Kim CW, Lee TH, Son Y and Kim J: CCL23 up-regulates expression of KDR/Flk-1 and potentiates VEGF-induced proliferation and migration of human endothelial cells. Biochem Biophys Res Commun. 382:124–128. 2009. View Article : Google Scholar : PubMed/NCBI

94 

Hefler LA, Zeillinger R, Grimm C, Sood AK, Cheng WF, Gadducci A, Tempfer CB and Reinthaller A: Preoperative serum vascular endothelial growth factor as a prognostic parameter in ovarian cancer. Gynecol Oncol. 103:512–517. 2006. View Article : Google Scholar : PubMed/NCBI

95 

Byrne AT, Ross L, Holash J, Nakanishi M, Hu L, Hofmann JI, Yancopoulos GD and Jaffe RB: Vascular endothelial growth factor-trap decreases tumor burden, inhibits ascites, and causes dramatic vascular remodeling in an ovarian cancer model. Clin Cancer Res. 9:5721–5728. 2003.PubMed/NCBI

96 

Jeon BH, Jang C, Han J, Kataru RP, Piao L, Jung K, Cha HJ, Schwendener RA, Jang KY, Kim KS, et al: Profound but dysfunctional lymphangiogenesis via vascular endothelial growth factor ligands from CD11b+ macrophages in advanced ovarian cancer. Cancer Res. 68:1100–1109. 2008. View Article : Google Scholar : PubMed/NCBI

97 

Zhang S, Xie B, Wang L, Yang H, Zhang H, Chen Y, Wang F, Liu C and He H: Macrophage-mediated vascular permeability via VLA4/VCAM1 pathway dictates ascites development in ovarian cancer. J Clin Invest. 131:e1403152021. View Article : Google Scholar :

98 

Reinartz S, Schumann T, Finkernagel F, Wortmann A, Jansen JM, Meissner W, Krause M, Schwörer AM, Wagner U, Müller-Brüsselbach S and Müller R: Mixed-polarization phenotype of ascites-associated macrophages in human ovarian carcinoma: Correlation of CD163 expression, cytokine levels and early relapse. Int J Cancer. 134:32–42. 2014. View Article : Google Scholar :

99 

Moughon DL, He H, Schokrpur S, Jiang ZK, Yaqoob M, David J, Lin C, Iruela-Arispe ML, Dorigo O and Wu L: Macrophage blockade using CSF1R inhibitors reverses the vascular leakage underlying malignant ascites in late-stage epithelial ovarian cancer. Cancer Res. 75:4742–4752. 2015. View Article : Google Scholar : PubMed/NCBI

100 

Clancy JL, Henderson MJ, Russell AJ, Anderson DW, Bova RJ, Campbell IG, Choong DY, Macdonald GA, Mann GJ, Nolan T, et al: EDD, the human orthologue of the hyperplastic discs tumour suppressor gene, is amplified and overexpressed in cancer. Oncogene. 22:5070–5081. 2003. View Article : Google Scholar : PubMed/NCBI

101 

Shearer RF, Iconomou M, Watts CK and Saunders DN: Functional roles of the E3 ubiquitin ligase UBR5 in cancer. Mol Cancer Res. 13:1523–1532. 2015. View Article : Google Scholar : PubMed/NCBI

102 

Tang M, Liu B, Bu X and Zhao P: Cross-talk between ovarian cancer cells and macrophages through periostin promotes macrophage recruitment. Cancer Sci. 109:1309–1318. 2018. View Article : Google Scholar : PubMed/NCBI

103 

Durlanik S, Fundel-Clemens K, Viollet C, Huber HJ, Lenter M, Kitt K and Pflanz S: CD276 is an important player in macrophage recruitment into the tumor and an upstream regulator for PAI-1. Sci Rep. 11:148492021. View Article : Google Scholar : PubMed/NCBI

104 

Worzfeld T, Finkernagel F, Reinartz S, Konzer A, Adhikary T, Nist A, Stiewe T, Wagner U, Looso M, Graumann J and Müller R: Proteotranscriptomics reveal signaling networks in the ovarian cancer microenvironment. Mol Cell Proteomics. 17:270–289. 2018. View Article : Google Scholar :

105 

Ichijo H, Nishida E, Irie K, ten Dijke P, Saitoh M, Moriguchi T, Takagi M, Matsumoto K, Miyazono K and Gotoh Y: Induction of apoptosis by ASK1, a mammalian MAPKKK that activates SAPK/JNK and p38 signaling pathways. Science. 275:90–94. 1997. View Article : Google Scholar : PubMed/NCBI

106 

Yin M, Zhou HJ, Zhang J, Lin C, Li H, Li X, Li Y, Zhang H, Breckenridge DG, Ji W and Min W: ASK1-dependent endothelial cell activation is critical in ovarian cancer growth and metastasis. JCI Insight. 2:e918282017. View Article : Google Scholar : PubMed/NCBI

107 

Sica A and Mantovani A: Macrophage plasticity and polarization: In vivo veritas. J Clin Invest. 122:787–795. 2012. View Article : Google Scholar : PubMed/NCBI

108 

Mills CD, Kincaid K, Alt JM, Heilman MJ and Hill AM: M-1/M-2 macrophages and the Th1/Th2 paradigm. J Immunol. 164:6166–6173. 2000. View Article : Google Scholar : PubMed/NCBI

109 

Mantovani A, Sozzani S, Locati M, Allavena P and Sica A: Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 23:549–555. 2002. View Article : Google Scholar : PubMed/NCBI

110 

Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A and Locati M: The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 25:677–686. 2004. View Article : Google Scholar : PubMed/NCBI

111 

Wen Y and Crowley SD: The varying roles of macrophages in kidney injury and repair. Curr Opin Nephrol Hypertens. 29:286–292. 2020. View Article : Google Scholar : PubMed/NCBI

112 

Garofalo RS, Orena SJ, Rafidi K, Torchia AJ, Stock JL, Hildebrandt AL, Coskran T, Black SC, Brees DJ, Wicks JR, et al: Severe diabetes, age-dependent loss of adipose tissue, and mild growth deficiency in mice lacking Akt2/PKB beta. J Clin Invest. 112:197–208. 2003. View Article : Google Scholar : PubMed/NCBI

113 

Gao J, Liang Y and Wang L: Shaping polarization of tumor-associated macrophages in cancer immunotherapy. Front Immunol. 13:8887132022. View Article : Google Scholar : PubMed/NCBI

114 

Wan C, Sun Y, Tian Y, Lu L, Dai X, Meng J, Huang J, He Q, Wu B, Zhang Z, et al: Irradiated tumor cell-derived microparticles mediate tumor eradication via cell killing and immune reprogramming. Sci Adv. 6:eaay97892020. View Article : Google Scholar : PubMed/NCBI

115 

Shu Y and Cheng P: Targeting tumor-associated macrophages for cancer immunotherapy. Biochim Biophys Acta Rev Cancer. 1874:1884342020. View Article : Google Scholar : PubMed/NCBI

116 

He J, Yin P and Xu K: Effect and molecular mechanisms of traditional Chinese medicine on tumor targeting tumor-associated macrophages. Drug Des Devel Ther. 14:907–919. 2020. View Article : Google Scholar : PubMed/NCBI

117 

Macciò A, Gramignano G, Cherchi MC, Tanca L, Melis L and Madeddu C: Role of M1-polarized tumor-associated macrophages in the prognosis of advanced ovarian cancer patients. Sci Rep. 10:60962020. View Article : Google Scholar : PubMed/NCBI

118 

Guiducci C, Vicari AP, Sangaletti S, Trinchieri G and Colombo MP: Redirecting in vivo elicited tumor infiltrating macrophages and dendritic cells towards tumor rejection. Cancer Res. 65:3437–3446. 2005. View Article : Google Scholar : PubMed/NCBI

119 

Capellero S, Erriquez J, Battistini C, Porporato R, Scotto G, Borella F, Di Renzo MF, Valabrega G and Olivero M: Ovarian cancer cells in ascites form aggregates that display a hybrid epithelial-mesenchymal phenotype and allows survival and proliferation of metastasizing cells. Int J Mol Sci. 23:8332022. View Article : Google Scholar : PubMed/NCBI

120 

Sun S, Pan X, Zhao L, Zhou J, Wang H and Sun Y: The expression and relationship of CD68-tumor-associated macrophages and microvascular density with the prognosis of patients with laryngeal squamous cell carcinoma. Clin Exp Otorhinolaryngol. 9:270–277. 2016. View Article : Google Scholar : PubMed/NCBI

121 

Sadrkhanloo M, Entezari M, Orouei S, Ghollasi M, Fathi N, Rezaei S, Hejazi ES, Kakavand A, Saebfar H, Hashemi M, et al: STAT3-EMT axis in tumors: Modulation of cancer metastasis, stemness and therapy response. Pharmacol Res. 182:1063112022. View Article : Google Scholar : PubMed/NCBI

122 

Liang R, Chen X, Chen L, Wan F, Chen K, Sun Y and Zhu X: STAT3 signaling in ovarian cancer: A potential therapeutic target. J Cancer. 11:837–848. 2020. View Article : Google Scholar : PubMed/NCBI

123 

Takaishi K, Komohara Y, Tashiro H, Ohtake H, Nakagawa T, Katabuchi H and Takeya M: Involvement of M2-polarized macrophages in the ascites from advanced epithelial ovarian carcinoma in tumor progression via Stat3 activation. Cancer Sci. 101:2128–2136. 2010. View Article : Google Scholar : PubMed/NCBI

124 

Jinushi M, Chiba S, Yoshiyama H, Masutomi K, Kinoshita I, Dosaka-Akita H, Yagita H, Takaoka A and Tahara H: Tumor-associated macrophages regulate tumorigenicity and anticancer drug responses of cancer stem/initiating cells. Proc Natl Acad Sci USA. 108:12425–12430. 2011. View Article : Google Scholar : PubMed/NCBI

125 

Raghavan S, Mehta P, Xie Y, Lei YL and Mehta G: Ovarian cancer stem cells and macrophages reciprocally interact through the WNT pathway to promote pro-tumoral and malignant phenotypes in 3D engineered microenvironments. J Immunother Cancer. 7:1902019. View Article : Google Scholar : PubMed/NCBI

126 

Ruffell B and Coussens LM: Macrophages and therapeutic resistance in cancer. Cancer Cell. 27:462–472. 2015. View Article : Google Scholar : PubMed/NCBI

127 

Li L, He D, Guo Q, Zhang Z, Ru D, Wang L, Gong K, Liu F, Duan Y and Li H: Exosome-liposome hybrid nanoparticle codelivery of TP and miR497 conspicuously overcomes chemoresistant ovarian cancer. J Nanobiotechnology. 20:502022. View Article : Google Scholar : PubMed/NCBI

128 

Li H, Luo F, Jiang X, Zhang W, Xiang T, Pan Q, Cai L, Zhao J, Weng D, Li Y, et al: CircITGB6 promotes ovarian cancer cisplatin resistance by resetting tumor-associated macrophage polarization toward the M2 phenotype. J Immunother Cancer. 10:e0040292022. View Article : Google Scholar : PubMed/NCBI

129 

Ding L, Wang Q, Martincuks A, Kearns MJ, Jiang T, Lin Z, Cheng X, Qian C, Xie S, Kim HJ, et al: STING agonism overcomes STAT3-mediated immunosuppression and adaptive resistance to PARP inhibition in ovarian cancer. J Immunother Cancer. 11:e0056272023. View Article : Google Scholar : PubMed/NCBI

130 

Willingham SB, Volkmer JP, Gentles AJ, Sahoo D, Dalerba P, Mitra SS, Wang J, Contreras-Trujillo H, Martin R, Cohen JD, et al: The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc Natl Acad Sci USA. 109:6662–6667. 2012. View Article : Google Scholar : PubMed/NCBI

131 

Sikic BI, Lakhani N, Patnaik A, Shah SA, Chandana SR, Rasco D, Colevas AD, O'Rourke T, Narayanan S, Papadopoulos K, et al: First-in-human, first-in-class phase I trial of the anti-CD47 antibody Hu5F9-G4 in patients with advanced cancers. J Clin Oncol. 37:946–953. 2019. View Article : Google Scholar : PubMed/NCBI

132 

Huang Y, Lv SQ, Liu PY, Ye ZL, Yang H, Li LF, Zhu HL, Wang Y, Cui LZ, Jiang DQ, et al: A SIRPα-Fc fusion protein enhances the antitumor effect of oncolytic adenovirus against ovarian cancer. Mol Oncol. 14:657–668. 2020. View Article : Google Scholar : PubMed/NCBI

133 

Batchu RB, Gruzdyn OV, Kolli BK, Dachepalli R, Umar PS, Rai SK, Singh N, Tavva PS, Weaver DW and Gruber SA: IL-10 signaling in the tumor microenvironment of ovarian cancer. Adv Exp Med Biol. 1290:51–65. 2021. View Article : Google Scholar : PubMed/NCBI

134 

McKarns SC, Schwartz RH and Kaminski NE: Smad3 is essential for TGF-beta 1 to suppress IL-2 production and TCR-induced proliferation, but not IL-2-induced proliferation. J Immunol. 172:4275–4284. 2004. View Article : Google Scholar : PubMed/NCBI

135 

Wertel I, Surówka J, Polak G, Barczyński B, Bednarek W, Jakubowicz-Gil J, Bojarska-Junak A and Kotarski J: Macrophage-derived chemokine CCL22 and regulatory T cells in ovarian cancer patients. Tumour Biol. 36:4811–4817. 2015. View Article : Google Scholar : PubMed/NCBI

136 

Kamat K, Krishnan V and Dorigo O: Macrophage-derived CCL23 upregulates expression of T-cell exhaustion markers in ovarian cancer. Br J Cancer. 127:1026–1033. 2022. View Article : Google Scholar : PubMed/NCBI

137 

Noy R and Pollard JW: Tumor-associated macrophages: From mechanisms to therapy. Immunity. 41:49–61. 2014. View Article : Google Scholar : PubMed/NCBI

138 

Chapoval AI, Ni J, Lau JS, Wilcox RA, Flies DB, Liu D, Dong H, Sica GL, Zhu G, Tamada K and Chen L: B7-H3: A costimulatory molecule for T cell activation and IFN-gamma production. Nat Immunol. 2:269–274. 2001. View Article : Google Scholar : PubMed/NCBI

139 

Miyamoto T, Murakami R, Hamanishi J, Tanigaki K, Hosoe Y, Mise N, Takamatsu S, Mise Y, Ukita M, Taki M, et al: B7-H3 suppresses antitumor immunity via the CCL2-CCR2-M2 macrophage axis and contributes to ovarian cancer progression. Cancer Immunol Res. 10:56–69. 2022. View Article : Google Scholar

140 

Liu Z, Jin K, Zeng H, Shao F, Chang Y, Wang Y, Xu L, Wang Z, Cui X, Zhu Y and Xu J: B7-H4 correlates with clinical outcome and immunotherapeutic benefit in muscle-invasive bladder cancer. Eur J Cancer. 171:133–142. 2022. View Article : Google Scholar : PubMed/NCBI

141 

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. 203:871–881. 2006. View Article : Google Scholar : PubMed/NCBI

142 

Carroll MJ, Fogg KC, Patel HA, Krause HB, Mancha AS, Patankar MS, Weisman PS, Barroilhet L and Kreeger PK: Alternatively-activated macrophages upregulate mesothelial expression of P-selectin to enhance adhesion of ovarian cancer cells. Cancer Res. 78:3560–3573. 2018. View Article : Google Scholar : PubMed/NCBI

143 

Robinson-Smith TM, Isaacsohn I, Mercer CA, Zhou M, Van Rooijen N, Husseinzadeh N, McFarland-Mancini MM and Drew AF: Macrophages mediate inflammation-enhanced metastasis of ovarian tumors in mice. Cancer Res. 67:5708–5716. 2007. View Article : Google Scholar : PubMed/NCBI

144 

Fogg KC, Olson WR, Miller JN, Khan A, Renner C, Hale I, Weisman PS and Kreeger PK: Alternatively activated macrophage-derived secretome stimulates ovarian cancer spheroid spreading through a JAK2/STAT3 pathway. Cancer Lett. 458:92–101. 2019. View Article : Google Scholar : PubMed/NCBI

145 

Zeng XY, Xie H, Yuan J, Jiang XY, Yong JH, Zeng D, Dou YY and Xiao SS: M2-like tumor-associated macrophages-secreted EGF promotes epithelial ovarian cancer metastasis via activating EGFR-ERK signaling and suppressing lncRNA LIMT expression. Cancer Biol Ther. 20:956–966. 2019. View Article : Google Scholar : PubMed/NCBI

146 

Germano G, Frapolli R, Belgiovine C, Anselmo A, Pesce S, Liguori M, Erba E, Uboldi S, Zucchetti M, Pasqualini F, et al: Role of macrophage targeting in the antitumor activity of trabectedin. Cancer Cell. 23:249–262. 2013. View Article : Google Scholar : PubMed/NCBI

147 

Sandhu SK, Papadopoulos K, Fong PC, Patnaik A, Messiou C, Olmos D, Wang G, Tromp BJ, Puchalski TA, Balkwill F, et al: A first-in-human, first-in-class, phase I study of carlumab (CNTO 888), a human monoclonal antibody against CC-chemokine ligand 2 in patients with solid tumors. Cancer Chemother Pharmacol. 71:1041–1050. 2013. View Article : Google Scholar : PubMed/NCBI

148 

Wesolowski R, Sharma N, Reebel L, Rodal MB, Peck A, West BL, Marimuthu A, Severson P, Karlin DA, Dowlati A, et al: Phase Ib study of the combination of pexidartinib (PLX3397), a CSF-1R inhibitor, and paclitaxel in patients with advanced solid tumors. Ther Adv Med Oncol. 11:17588359198542382019. View Article : Google Scholar : PubMed/NCBI

149 

Zeng Y, Li B, Liang Y, Reeves PM, Qu X, Ran C, Liu Q, Callahan MV, Sluder AE, Gelfand JA, et al: Dual blockade of CXCL12-CXCR4 and PD-1-PD-L1 pathways prolongs survival of ovarian tumor-bearing mice by prevention of immunosuppression in the tumor microenvironment. FASEB J. 33:6596–6608. 2019. View Article : Google Scholar : PubMed/NCBI

150 

Wanderley CW, Colón DF, Luiz JPM, Oliveira FF, Viacava PR, Leite CA, Pereira JA, Silva CM, Silva CR, Silva RL, et al: Paclitaxel reduces tumor growth by reprogramming tumor-associated macrophages to an M1 Profile in a TLR4-dependent manner. Cancer Res. 78:5891–5900. 2018. View Article : Google Scholar : PubMed/NCBI

151 

Zhang Q, Li Y, Miao C, Wang Y, Xu Y, Dong R, Zhang Z, Griffin BB, Yuan C, Yan S, et al: Anti-angiogenesis effect of Neferine via regulating autophagy and polarization of tumor-associated macrophages in high-grade serous ovarian carcinoma. Cancer Lett. 432:144–155. 2018. View Article : Google Scholar : PubMed/NCBI

152 

Zhang F, Parayath NN, Ene CI, Stephan SB, Koehne AL, Coon ME, Holland EC and Stephan MT: Genetic programming of macrophages to perform anti-tumor functions using targeted mRNA nanocarriers. Nat Commun. 10:39742019. View Article : Google Scholar : PubMed/NCBI

153 

Chen D, Xie J, Fiskesund R, Dong W, Liang X, Lv J, Jin X, Liu J, Mo S, Zhang T, et al: Chloroquine modulates antitumor immune response by resetting tumor-associated macrophages toward M1 phenotype. Nat Commun. 9:8732018. View Article : Google Scholar : PubMed/NCBI

154 

Bellora F, Castriconi R, Dondero A, Pessino A, Nencioni A, Liggieri G, Moretta L, Mantovani A, Moretta A and Bottino C: TLR activation of tumor-associated macrophages from ovarian cancer patients triggers cytolytic activity of NK cells. Eur J Immunol. 44:1814–1822. 2014. View Article : Google Scholar : PubMed/NCBI

155 

Ma L, Zhu M, Gai J, Li G, Chang Q, Qiao P, Cao L, Chen W, Zhang S and Wan Y: Preclinical development of a novel CD47 nanobody with less toxicity and enhanced anti-cancer therapeutic potential. J Nanobiotechnology. 18:122020. View Article : Google Scholar : PubMed/NCBI

156 

Rakina M, Kazakova A, Villert A, Kolomiets L and Larionova I: Spheroid formation and peritoneal metastasis in ovarian cancer: The role of stromal and immune components. Int J Mol Sci. 23:62152022. View Article : Google Scholar : PubMed/NCBI

157 

Long L, Yin M and Min W: 3D co-culture system of tumor-associated macrophages and ovarian cancer cells. Bio Protoc. 8:e28152018.PubMed/NCBI

Related Articles

Journal Cover

December-2024
Volume 65 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu Y, Xiao H, Zeng H and Xiang Y: Beyond tumor‑associated macrophages involved in spheroid formation and dissemination: Novel insights for ovarian cancer therapy (Review). Int J Oncol 65: 117, 2024.
APA
Liu, Y., Xiao, H., Zeng, H., & Xiang, Y. (2024). Beyond tumor‑associated macrophages involved in spheroid formation and dissemination: Novel insights for ovarian cancer therapy (Review). International Journal of Oncology, 65, 117. https://doi.org/10.3892/ijo.2024.5705
MLA
Liu, Y., Xiao, H., Zeng, H., Xiang, Y."Beyond tumor‑associated macrophages involved in spheroid formation and dissemination: Novel insights for ovarian cancer therapy (Review)". International Journal of Oncology 65.6 (2024): 117.
Chicago
Liu, Y., Xiao, H., Zeng, H., Xiang, Y."Beyond tumor‑associated macrophages involved in spheroid formation and dissemination: Novel insights for ovarian cancer therapy (Review)". International Journal of Oncology 65, no. 6 (2024): 117. https://doi.org/10.3892/ijo.2024.5705