Abstract
Ovarian cancer is the most lethal gynecological malignancy. It is usually diagnosed at a late stage, with a 5-yr survival rate of <30%. The majority of ovarian cancer cases are diagnosed after tumors have widely spread within the peritoneal cavity, limiting the effectiveness of debulking surgery and chemotherapy. Owing to a substantially lower survival rate at late stages of disease than at earlier stages, the major cause of ovarian cancer deaths is believed to be therapy-resistant metastasis. Although metastasis plays a crucial role in promoting ovarian tumor progression and decreasing patient survival rates, the underlying mechanisms of ovarian cancer spread have yet to be thoroughly explored. For many years, researchers have believed that ovarian cancer metastasizes via a passive mechanism by which ovarian cancer cells are shed from the primary tumor and carried by the physiological movement of peritoneal fluid to the peritoneum and omentum. However, the recent discovery of hematogenous metastasis of ovarian cancer to the omentum via circulating tumor cells instigated rethinking of the mode of ovarian cancer metastasis and the importance of the “seed-and-soil” hypothesis for ovarian cancer metastasis. In this review we discuss the possible mechanisms by which ovarian cancer cells metastasize from the primary tumor to the omentum, the cross-talk signaling events between ovarian cancer cells and various stromal cells that play crucial roles in ovarian cancer metastasis, and the possible clinical implications of these findings in the management of this deadly, highly metastatic disease.
Keywords: ovarian cancer, metastasis, stromal-tumoral interaction, passive dissemination, hematogenous metastasis
ovarian cancer is the fifth-leading cause of cancer deaths in women in the United States. Researchers have estimated that ∼21,290 new cases of epithelial ovarian cancer would be diagnosed in the United States in 2015 and that 14,180 deaths would occur (143). Despite ongoing efforts to develop effective surgery and treatment regimens for ovarian cancer, the overall survival rate remains a dismal 30%, which in large part results from diagnosis at a late stage. The late diagnosis of ovarian cancer is attributed to the fact that typical ovarian cancer symptoms, including abdominal discomfort, bloating, gas, nausea, and urinary urgency, are not explicit and are often mistaken for gastrointestinal problems (63, 144). When ovarian cancer is detected at an early stage, when the tumor tissue is confined to one or both ovaries, the cure rate with conventional therapies, such as cytoreductive surgery and chemotherapy, is as high as 90%. However, after the disease has spread to other pelvic organs, such as the uterus and bladder (stage 2), the abdomen (stage 3), or beyond the peritoneal cavity (stage 4), the cure rate decreases substantially owing to the limited efficacy of optimal debulking and tumor management (10, 31). Because >70% of ovarian cancer cases are diagnosed at a late stage, when cancer cells are actively metastasizing, understanding ovarian cancer pathogenesis and the mechanism of its metastasis is crucial for the management of this deadly, highly metastatic disease.
Ovarian cancer often metastasizes throughout the peritoneal cavity, to the omentum, and even to the parenchyma of the liver or lung. However, its spread is widely believed to occur via the peritoneal circulation, associated with ascites formation, instead of the classic patterns of metastasis via the hematogenous route (91). This is because implantation of peritoneovenous shunts to palliate discomfort of ascites and the resulting infusion of billions of cancer cells into the patient's circulation result in confinement of the disease within the abdomen (153).
However, does ovarian cancer metastasis occur exclusively via intraperitoneal seeding, or does it follow the “seed-and-soil” hypothesis, which plays an important role in the confinement of the disease within the abdomen? According to the seed-and-soil hypothesis proposed by the English surgeon Stephen Paget in 1889, the pattern of metastasis is not a random outcome. Instead, certain tumor cells (seeds) have specific affinity for certain organs (soil) (119). Paget stated that a plant's seeds are carried in all directions but can only live and grow if they fall on congenial soil. Thus he concluded that metastases form only when the tumor cells and organs are compatible. Indeed, after frequent revisiting of this theory in recent years, several new findings on the intrinsic properties of tumor cells surrounding stromal cells and the signaling cross talk of cancer cells with the host, which contribute to the organ-specific pattern of metastasis, have been reported. A clear understanding of these cellular and molecular mechanisms will definitely shed light on possible therapeutic strategies to increase survival rates in ovarian cancer patients. In this review we discuss the possible mechanisms by which ovarian cancer cells metastasize from the primary tumor to the omentum, the cross-talk signaling events between ovarian cancer cells and various stromal cells that play crucial roles in ovarian cancer metastasis, and the possible clinical implications of these findings in the management of this deadly, highly metastatic disease.
Passive Dissemination of Ovarian Tumor Nodules
Detachment of ovarian cancer cells from the primary tumor and physiological movement of peritoneal fluid to the peritoneum and omentum, as well as direct extension of tumor lesions to adjacent organs, are widely accepted as the most common routes of metastasis of epithelial ovarian cancer (91, 107). The cells follow the circulatory path of peritoneal fluid along with the respiratory force from the pelvis up the paracolic gutters along the intestinal mesentery to the right hemidiaphragm, facilitating implantation at different peritoneal organs (28, 41, 101). However, this transcoelomic route of dissemination only occurs when excess fluid is present in the peritoneum; otherwise, the motion of cancer cells is restricted to the primary site (28, 152). Therefore, accumulation of ascites in the abdomen of an ovarian cancer patient, which contributes significantly to morbidity owing to gastrointestinal symptoms and abdominal discomfort, is believed to play a key role in this passive mechanism of ovarian cancer cell spread.
The etiology of ascites is not well understood, but preclinical and clinical observations demonstrated that the vascular permeability-enhancing factor vascular endothelial growth factor (VEGF) is responsible for ascites accumulation (87, 163), and obstruction of lymphatic vessels by cancer cells may also cause the accumulation of ascites (152). In most cases of ovarian cancer, ascitic fluid is composed of malignant cells, a large number of white blood cells, and a high level of lactate dehydrogenase (9, 51, 133). Detached cancer cells usually float in the ascites as multicellular spheroids (91), and implantation of the spheroids in peritoneal organs involves interaction between the cancer cells and the mesothelium, which covers all organs within the peritoneal cavity, including the diaphragm, bowel serosa, omentum, and entire peritoneum (45, 85). The mesothelium is a single layer of mesothelial cells covering a basement membrane predominantly composed of collagen types I and IV, fibronectin, and laminin (83, 84, 162). Early adhesion and metastasis of ovarian cancer cells are mediated by the matrix metalloproteinase (MMP) family of proteins, the expression of which is upregulated during ovarian cancer progression (49). In particular, MMP-2 enhances peritoneal adhesion of ovarian cancer cells via cleavage of fibronectin and vitronectin into small fragments, as cancer cells adhere more strongly to these fragments using fibronectin (α5β1-integrin) and vitronectin (αVβ3-integrin) receptors than to the whole extracellular matrix proteins (84). Another type IV collagenase, MMP-9, which is secreted by the tumor stroma, is also involved in the release of VEGF from tumor cells and stimulates ascites formation in ovarian cancer patients (13). Moreover, a recent study demonstrated that cancer-associated mesothelial cells facilitate initial metastatic colonization of ovarian cancer cells, secreting fibronectin and providing access to the submesothelial extracellular matrix (82). In addition, upregulation of E-cadherin expression, a feature of metastatic epithelial ovarian cancer cells (149, 150), may increase adhesion of circulating ovarian cancer cells to peritoneal organs (76).
Once attached to the peritoneal surface, metastatic cancer cells proliferate and invade the mesothelium, which can be promoted by various ascitic constituents (152). For instance, lysophosphatidic acid, which is highly expressed in the ascites and plasma of ovarian cancer patients, promotes ovarian cancer motility and invasiveness (50) via induction of secretion of extracellular matrix-degrading proteinases, including MMPs (53) and urokinase plasminogen activator (126). In addition, lysophosphatidic acid activates mitogen-activated protein kinase 1, which facilitates the redistribution of focal adhesion kinase to focal adhesion complexes, as well as cell motility and migration (16). On the other hand, CXCL12 in ascitic fluid binds to its receptor, CXCR4, on ovarian cancer cells, increasing the expression of β1-integrin and the migratory potential of epithelial ovarian cancer cells (7). Furthermore, malignant ascites stimulates expression of CD44 by cancer cells. CD44 is the principal cell surface receptor for hyaluronic acid. Engagement of CD44 by hyaluronic acid facilitates epithelial ovarian cancer cell migration and binding to the peritoneal surface (29, 147).
After ovarian cancer cell implantation, the resultant inflammation and injury stimulate the peritoneal cells and their associated immune and stromal cells to release cytokines such as interleukin (IL)-1, -6, and -8, which subsequently enhance tumor angiogenesis and further ascites formation via increased secretion of VEGF by cancer cells (57, 145). This creates a favorable microenvironment to support the growth of the implants. Indeed, high levels of VEGF expression in serum and ascites of ovarian cancer patients have been associated with ovarian tumor progression and poor prognosis (111). It has also been demonstrated that ovarian cancer cells preferentially metastasize to milky spots on the subdiaphragmatic surface and omentum and use the associated lymphatic vessels for spreading (12, 51). These milky spots are composed of aggregations of mesenchymal cells, mainly macrophages and lymphocytes. The spots are easily infiltrated by tumor cells and participate in intraperitoneal immune reactions (11, 140).
Lastly, microarray studies (1, 73), comparative genomic hybridization (78), and high-resolution single-nucleotide polymorphism analysis (72) demonstrated similar genetic alterations in primary ovarian tumors and their respective metastases, further supporting the passive mechanism of ovarian cancer metastasis described above. Therefore, transcoelomic metastasis is widely accepted as the most common route of metastasis of epithelial ovarian cancer.
Hematogenous Metastasis of Ovarian Cancer
Although direct surface spread via intraperitoneal dissemination has long been thought to be the dominant mechanism of ovarian cancer metastasis, traces of evidence have prompted scientists to investigate an alternative route of metastasis. First, instead of a completely random distribution of metastases within the peritoneal cavity, investigators have consistently identified a predilection of metastases for implantation in the omentum (51, 124). This prompted scientists to consider the existence of an active mechanism of metastasis of epithelial ovarian cancer to this large, apronlike fold of visceral tissue that hangs down from the stomach. In addition, at the initial diagnosis, some ovarian cancer patients have retroperitoneal or distant metastases and even submesothelial disease (3, 30, 94), which is not a common consequence of peritoneal dissemination. Finally, circulating tumor cells have been found in blood samples obtained from ovarian cancer patients (120, 122), suggesting a blood-borne route of ovarian cancer metastasis.
Recently, using a parabiosis animal model, Pradeep and colleagues demonstrated that ovarian circulating tumor cells preferentially implant and grow in the omentum and subsequently spread to other peritoneal surfaces (124). Specifically, they excised skin of female mice from the shoulder to the hip joint and then surgically anastomosed pairs of mice. SKOV3ip1 ovarian cancer cells injected into the peritoneal cavity or ovaries of host mice were able to hematogenously metastasize to the omentum in the guest mice. Immunofluorescent staining of sections of the anastomosed skin for blood (CD31) and lymphatic (LYVE1) vessel markers confirmed that the paired mice in the parabiosis model shared blood but not lymphatic vessels.
To delineate the mechanisms by which ovarian cancer cells preferentially metastasize to the omentum, Pradeep and colleagues also compared gene expression profiles for SKOV3ip1 cells and SKOV3-OM3, a cell line derived from omental tumors in the guest mice, in three repeated parabiosis cycles (124). In a network-based analysis of the gene expression data, they identified the epidermal growth factor receptor/ErbB2 network as having the greatest number of differentially expressed genes. Furthermore, they found markedly higher expression of ERBB3 and NRG1, which encode for the ERBB3 ligand neuregulin 1 (NRG1), in SKOV3-OM3 than SKOV3ip1 cells. In line with the fact that NRG1 is known to increase cell motility potential, the expression of several mesenchymal phenotype-related genes, such as vimentin, MMP3, COL1A, HMGA2, and HGF, was higher in SKOV3-OM3 than SKOV3ip1 cells, whereas the expression of negative epithelial-mesenchymal transition regulators, such as E-cadherin, SFRP1, EFNA1, and SMAD7, was lower. Lastly, silencing of tumoral ErbB3 and omental NRG1 expression via systematic delivery of small interfering RNA using a nanoliposomal platform inhibited omental metastasis in mouse models of ovarian cancer, suggesting that the ErbB3-NRG1 axis is responsible for the hematogenous omental metastasis of circulating ovarian cancer cells.
Molecular Mechanism of Ovarian Cancer Metastasis
The physical mechanisms by which ovarian cancer cells metastasize via passive dissemination and the hematogenous route appear to differ (Fig. 1). Whereas passive dissemination consists of relatively straightforward migration, adhesion, and invasion of disseminated cancer cells into the preferential soil (omentum), metastasis via the hematogenous route requires a series of sequential interrelated steps of invasion into surrounding tissue. In metastasis of ovarian cancer cells via the hematogenous route, intravasation, which is the invasion of cancer cells through the basal membrane into blood or lymphatic vessels, is the first step. The cancer cells then transit in the blood or lymph and undergo extravasation, which is the exit of cells from the blood or lymph vessels. Subsequently, ovarian cancer cells establish secondary tumors at the metastatic site (108, 115). Despite the differences between these two mechanisms of ovarian cancer metastasis, both are primarily cell motility-driven, involving cycles of actin polymerization, cell adhesion, and actomyosin contraction (115).
Fig. 1.
Two mechanisms of ovarian cancer metastasis and the protein factors involved. Left: as ovarian cancer progresses, cancer metastasis could occur via passive dissemination of cancer cells and hematogenous metastasis. For the passive dissemination model, ovarian cancer cells detached from the primary tumor are carried by peritoneal fluid and ascites. At the metastatic site, cancer cells adhere to the surface of the organ, establishing tumor nodules. Right: for hematogenous metastasis, ovarian cancer cells at the primary tumor site invade through the basal membrane of blood vessels and enter the circulation via intravasation. Subsequently, cancer cells leave the circulation at the metastatic site via extravasation, establishing metastatic tumor nodules. VEGF, vascular endothelial growth factor; MMP, matrix metalloproteinase; UPA, urokinase plasminogen activator; LPA, lysophosphatidic acid; FAK, focal adhesion kinase; MLCK, myosin light-chain kinase; NRG1, neuregulin 1.
Reorganization of the actin cytoskeleton is the primary mechanism of cell motility and is driven by polymerization of actin monomers into polarized filaments called F-actin (123, 127). This polymerization is regulated by members of the Rho family of small GTPases, such as Rho, Rac, and Cdc42 (166). These GTPases act as molecular switches by cycling between a guanosine diphosphate-bound inactive state and a guanosine triphosphate-bound active state in response to stimuli, controlling the formation of filopodia, lamellipodia, and stress fibers for cell movement (128). Filopodia are F-actin-rich protrusions at the front of the cell, lamellipodia are sheets of F-actin that extend over the substrate, and stress fibers are thick actin cables in the cell body. In highly invasive cancer cells, proteolytically active plasma membrane protrusions called invadopodia form to induce focal degradation of extracellular matrix components. The potential of invadopodia formation by cancer cells is often correlated with their ability to enter the vasculature (165). These different actin filament structures are responsible for the diversity of actin organization in migrating cancer cells (35, 115).
To connect actin filaments to the plasma membrane and cell-matrix adhesions, the cytoskeletal protein α-actinin forms complexes with focal adhesion proteins such as vinculin and zyxin after phosphorylation by focal adhesion kinase and cross-linking with actomyosin stress fibers and generates traction force to be transmitted to the substrate, driving cell motion (56, 66). The components of focal adhesion complexes are regulated by Ca2+ influx, which can occur in the form of waves, sparks, or flickers (125). Such increases in Ca2+ concentration can result from Ca2+ entry through membrane-bound Ca2+-permeable channels and/or Ca2+ release from intracellular endoplasmic reticulum Ca2+ stores through ryanodine and/or inositol triphosphate receptor channels. These signals stimulate downstream effectors such as Ca2+-dependent myosin light-chain kinase, which mediates myosin light-chain phosphorylation and myosin II contraction to induce coordinated dynamic formation and disassembly of cell adhesions, known as focal adhesion turnover, which facilitates cell migration (20, 69).
Using traction force microscopy, McGrail and colleagues recently showed that the tropism of ovarian cancer metastasis to the soft omentum results from not only chemical signals but also mechanical cues (99). They reported an increase in both the magnitude of traction forces and the degree of polarization in human ovarian cancer cells cultured on soft substrates. Moreover, ovarian cancer cells underwent epithelial-mesenchymal transition via the Rho/ROCK signaling pathway, suggesting that this transition is a critical step in both passive dissemination and the hematogenous route of ovarian cancer metastasis.
Stromal Contributions to Ovarian Cancer Metastasis
Researchers have demonstrated that both intraperitoneal dissemination and hematogenous spread of circulating tumor cells are possible mechanisms of ovarian cancer metastasis. However, what are the factors and signaling pathways that cue the metastatic events? With increasing evidence demonstrating the importance of stromal involvement in cancer progression and the ability of stroma-derived factors to confer aggressive phenotypes to ovarian cancer (151, 155), we describe recent findings on the cross-talk signaling events between ovarian cancer cells and various stromal cells that play crucial roles in ovarian cancer metastasis (Fig. 2).
Fig. 2.
Cellular cross talk and signaling events in ovarian cancer metastasis. Ovarian cancer progression and metastasis from the primary site to the omental metastatic tumor site are facilitated by the interaction between cancer cells and various stromal components. Left: at the primary tumor site (ovary), ovarian cancer cell motility is enhanced by cancer-associated fibroblast (CAF)-derived secretory proteins. Versican (VCAN) expression is upregulated in CAFs via activation of transforming growth factor (TGF)-β signaling. VCAN then activates NF-κB signaling in ovarian cancer cells and promotes cancer cell motility and invasion potential via upregulation of CD44, hyaluronan-mediated motility receptor (HMMR), and MMP9. At the same time, CAF-derived microfibrillar-associated protein 5 (MFAP5) binds to the αVβ3-integrin receptors on the ovarian cancer cell surface, activating the Ca2+-dependent FAK/cAMP response element-binding protein/troponin C type 1 signaling pathways. Activation of such signaling stimulates reorganization of the F-actin cytoskeleton and enhances generation of cell traction force, thereby increasing the migration potential of ovarian cancer cells. In addition, chemokine (C-X-C motif) ligand 12 (CXCL12) is a well-reported tumor-promoting factor from CAFs. CXCL12 binds to its receptor, chemokine (C-X-C motif) receptor type 4 (CXCR4), on cancer cells to increase tumor cell proliferation and migration and CXCR4 on endothelial cells to induce tumor angiogenesis. On the other hand, cancer cell-derived fibroblast growth factor 1 (FGF1) and fibroblast growth factor 18 (FGF18), in addition to promoting tumor progression via an autocrine mechanism by binding to its receptor, fibroblast growth factor receptor 4 (FGFR4), on cancer cells, also promote tumor angiogenesis through a paracrine mechanism by binding to FGFR4 on endothelial cells. Together with cancer cell-derived VEGF, these proangiogenic factors promote the establishment of tumor microvessels, which could subsequently facilitate hematogenous metastasis of ovarian cancer cells. Right: in the omentum, a common metastatic site for ovarian cancer, secretory factors from adipocytes and macrophages contribute to a favorable microenvironment for tumor development. Interactions between adipocytes and cancer cells in the omentum promote tumor progression. In addition to providing an energy source for cancer cells, adipocytes also secrete multiple cytokines, including TNF-α, IL-6, and IL-8, which act on ovarian cancer cells and accelerate tumor progression. Macrophages are highly abundant within the omentum, especially at the milky spots. Macrophages actively produce and secrete TNF-α and VEGF into the tumor microenvironment, which promotes tumor progression and tumor angiogenesis, respectively.
Cancer-Associated Fibroblasts
Stroma is a large component of many advanced ovarian tumors, with a median relative proportion of tumor tissue of 50% (range 7–83%) (89). Although the tumor microenvironment is composed of a complex variety of mesenchymal cells, including fibroblasts, endothelial cells, pericytes, and diverse immune cells, cancer-associated fibroblasts (CAFs) often represent the majority of stromal cells in various types of human carcinoma (117). Common upregulated CAF markers include α-smooth muscle actin (74), stromal-derived factor 1α (CXCL12) (136), fibroblast activation protein-1α (117), vimentin (96), and fibroblast-specific protein-1 (148). CAFs possess very high contractile ability, promote angiogenesis, and stimulate epithelial cell growth via production of extracellular matrix and secretion of growth factors and cytokines (104, 139). Therefore, CAF-derived tumor-promoting factors may serve as both prognostic markers and therapeutic targets.
Multiple signaling pathways involved in the tumor-promoting roles of CAFs, such as the well-reported CXCL12-CXCR4 axis, which plays prominent roles in inducing angiogenesis at the tumor site and increasing tumor cell proliferation and migration in different cancer systems, have been reported (32, 44, 98, 113, 114). However, most of these studies were performed using in vitro co-culture systems; also, previous large-scale transcriptome profiling studies, including the Cancer Genome Atlas, which identified the prognostic gene signature for high-grade serous ovarian cancer (HGSOC), used mostly bulk ovarian tumor samples (14, 25, 90). As a result, the prognostic significance of CAF-specific gene expression in ovarian cancer cells remains largely unknown.
In 2014, Leung and colleagues presented large-scale transcriptome profiling data on microdissected tumor samples obtained from HGSOC patients and identified a specific gene signature for CAFs that was associated with survival (93). In addition, they functionally characterized microfibrillar-associated protein 5, a prognostic marker of poor survival of HGSOC, and demonstrated that the protein stimulates ovarian cancer motility and metastatic potential via the Ca2+-dependent focal adhesion kinase/cAMP response element-binding protein/troponin C type 1 signaling pathway. In vivo animal studies demonstrated that targeting stromal microfibrillar-associated protein 5 using small interfering RNA encapsulated in nanoparticles markedly reduced ovarian tumor growth and metastasis (93), suggesting that targeting the CAF-derived factor microfibrillar-associated protein 5 is a new treatment modality for HGSOC.
Using the same transcriptome profiling data on microdissected ovarian tumor samples, Yeung and colleagues identified versican as a key upregulated gene in CAFs that promotes the motility and invasion of ovarian cancer cells by activating the nuclear factor-κB signaling pathway and upregulating CD44, MMP-9, and hyaluronan-mediated motility receptor expression in cancer cells (168). Interestingly, they found that versican expression in CAFs was modulated by the activation of transforming growth factor (TGF)-β signaling in CAFs induced by TGF-β ligands secreted by ovarian cancer cells. Although TGF-β ligands have limited direct effects on ovarian cancer cells (6, 164), the cross talk between cancer cells and CAFs via versican plays a critical role in TGF-β-stimulated ovarian cancer progression. These findings provide important implications for the development and refinement of TGF-β-targeted therapy for ovarian cancer.
Whereas CAFs are normally regarded as transformed normal fibroblasts (103) or differentiated bone marrow-derived progenitor cells recruited to the tumor site (117), other efforts to identify factors that maintain the “CAF state” also possess important therapeutic value in controlling ovarian cancer (2). Targeting CAFs for cancer treatment is believed to have two benefits: 1) the ongoing function of CAFs is critical to the growth of nearby neoplastic cells, and 2) stromal cells are more genetically stable than carcinoma cells, which can accumulate adaptive mutations during the course of therapy to acquire drug resistance (79, 81, 102).
Endothelial Cells
Blood vessels are fundamentally lined at the luminal side by endothelial cells. Interconnected endothelial cells form tubes that direct and maintain blood flow and are an important gateway for hematogenous metastasis of ovarian cancer (70). Tumor-induced angiogenesis is a process by which growing solid tumors produce diffusible angiogenic factors that induce host capillary endothelial cells to proliferate, migrate, and form new vessels (18, 43, 54, 55). The most well-studied angiogenesis inducer is VEGF-A, which is secreted by tumor and stromal cells, including macrophages, endothelial cells, and fibroblasts, to facilitate vascular sprouting and increase vascular permeability (62). VEGF-A binds to the VEGF receptor tyrosine kinases VEGFR1 and VEGFR2 on endothelial cells, leading to the formation of VEGFR homodimers or heterodimers and the induction of downstream signaling events (116). Moreover, VEGFR signaling can be modulated by co-receptors such as neuropilins, which may increase the half-life of the receptor complex.
Other well-known inducers of angiogenesis include the fibroblast growth factors (FGFs) and their transmembrane tyrosine kinase receptors (FGFRs) (19, 58). Birrer and colleagues showed that amplification of chromosomal region 5q31-5q35.3 and FGF1 in ovarian cancer tissues is a negative prognostic indicator for advanced-stage HGSOC, as this amplification promotes tumor angiogenesis and increases ovarian cancer cell motility and survival in an autocrine manner (17). In addition, Zaid and colleagues showed that overexpression of FGFR4, one of the key receptors for FGF1, in ovarian cancer cells was highly associated with decreased patient survival (169). In addition, silencing of FGFR4 in ovarian cancer cells markedly abrogated the FGF1-activated mitogen-activated protein kinase, nuclear factor-κB, and WNT signaling pathways. Silencing of FGFR4 with FGFR4-specific small interfering RNAs and blockage of FGFR4 activation by FGFR4 trap protein effectively attenuated ovarian tumor growth in vivo (169). In comparison, Wei and colleagues identified FGF18 as an independent predictive marker for poor survival in patients with advanced-stage HGSOC (160). Functional studies demonstrated that FGF18 stimulates ovarian cancer cell migration, invasion, and tumorigenicity via nuclear factor-κB activation and enhances tumor angiogenesis and M2 macrophage infiltration. Their observations of increased intratumoral microvessel density and M2 macrophage infiltration in clinical ovarian tumor samples confirm the in vitro data and suggest that FGF18 is a therapeutic target for ovarian cancer (160). Taken together, these findings demonstrate that FGFs and their receptors are important regulators of ovarian tumor progression and angiogenesis. Enhanced tumor angiogenesis induced by the FGF-FGFR signaling axis may facilitate hematogenous metastasis of ovarian cancer.
Expression of VEGF, VEGFRs, basic FGF, and neuropilin-1 is upregulated in ovarian cancer cells (5, 34), and microvessel density is a prognostic marker for multiple tumor types (26, 33, 86, 131), including ovarian cancer (146). Therefore, investigators have developed multiple therapeutic agents targeting angiogenesis inducers, such as bevacizumab, a VEGF-A-targeting monoclonal antibody, and various FGF inhibitors (27). Most clinical trials of antiangiogenic agents have included bevacizumab. In particular, two studies (GOG218 and ICON7) demonstrated the efficacy of bevacizumab, in addition to carboplatin and paclitaxel, followed by maintenance treatment, in patients with International Federation of Gynecology and Obstetrics stage 3/4 ovarian cancer with residual tumors after primary surgery (22, 121).
Macrophages
The omentum, which is the most common site of ovarian cancer metastasis via the transcoelomic and hematogenous routes, is composed mainly of adipose tissue, immune aggregates, and structural elements surrounding capillary beds (51, 140). Although researchers have speculated that ovarian cancer cells preferentially localize to the omentum owing to 1) lack of a basement membrane and few mesothelial cells on milky spot surfaces, which facilitate cancer cell invasion, and 2) participation of the omentum in fluid drainage from the peritoneal cavity, which may passively increase the chances of cancer cell attachment to the omentum (140), rapid localization of ovarian cancer cells to the omentum within minutes after intraperitoneal injection of ovarian cancer cells (68, 88) suggests that an active component is involved in cancer cell homing and invasion (40, 68, 88, 157). In comparing lodging of a panel of ovarian cancer cells on milky spot-containing (omentum and splenoportal fat) and milky spot-deficient (uterine fat, gonadal fat, and mesentery) peritoneal adipose tissue, Clark and colleagues clearly demonstrated that the milky spots dramatically enhance early cancer cell lodging on the omentum (40).
Milky spots are cellular aggregations of mesenchymal cells composed mainly of macrophages and lymphocytes (140). To identify the key components involved in early cancer cell lodging, Clark and colleagues injected ovarian cancer cells into various immunodeficient mice, including athymic nude, Rag1, Igh6, and BN XID mice (40). The results demonstrate that ovarian cancer cell colonization is not affected by deficiency or absence of T, B, and/or natural killer cells in these mouse strains (40). Because macrophages are frequently observed in milky spots (11, 141, 142, 161) and stimulation of omental macrophages promotes tumor growth on the omentum (105), macrophages may contribute to the rapid and specific colonization of ovarian cancer cells on omental milky spots.
Robust local and systemic host inflammatory responses have been reported to accompany ovarian tumor progression, particularly during tumor dissemination (71, 132, 158). To determine the cell types responsible for the ovarian tumor-promoting effect of enhanced inflammatory responses, Robinson-Smith and colleagues depleted specific innate immune cell populations from the peritoneal cavity in animal models (132). Their results demonstrated that neither neutrophil nor natural killer cell depletion significantly affected either ovarian tumor growth or peritoneal metastasis. In contrast, macrophage depletion had a profound suppressive effect on both primary tumor development and tumor progression (132). They reported that the reduced tumor dissemination in macrophage-depleted mice was not solely a result of decreased primary tumor growth, as they also observed this effect in an experimental metastasis model (132). Possible tumor-promoting mechanisms of macrophages include an increase in the vascularity of tumors, destruction of the extracellular matrix, and release of tumor-stimulating factors (132). Indeed, the vasculature in milky spots of the omentum is CD105-positive and contains vascular sprouts, indicating active angiogenesis (61). Also, reduced VEGF expression has been observed in macrophage-depleted mice (132). Because VEGF production induces ascites production, tumor angiogenesis, and metastasis (64, 100, 106, 170), macrophages and VEGF are logical therapeutic targets for ovarian cancer. Regarding extracellular matrix remodeling, investigators showed that macrophage-derived MMP-9 contributed to ovarian tumor angiogenesis and human ovarian cancer cell growth (52, 75). Furthermore, Hagemann and colleagues demonstrated that macrophages induce invasiveness of epithelial cancer cells via nuclear factor-κB and c-Jun NH2-terminal kinase signaling (67). A relationship between ovarian tumor progression and other ovarian tumor macrophage-derived proteins, including survival factors such as IL-6, IL-8, and VEGF-C, which promote lymphatic metastasis, has also been reported (137, 158).
Adipocytes
Ovarian cancer metastasis is not a random event, as 80% of patients with serous ovarian carcinoma present with omental metastases (109). Because the omentum is a fat pad primarily composed of adipocytes, these cells may contribute to the predilection of the metastatic cascade for the omentum. In fact, researchers introduced the term cancer-associated adipocytes (CAAs) in 2010 for adipocytes in close proximity to cancer cells (47). Studies demonstrated that CAAs promote the growth of cancer cells (46, 97, 156). Nowicka and colleagues showed that the origin of CAAs (42), adipose-derived mesenchymal stem cells in the omentum, promote proliferation, migration, and chemoresistance of ovarian cancer cells (112). In addition, several adipokines secreted by adipose tissue, including tumor necrosis factor-α, IL-6, IL-8, and monocyte chemoattractant protein-1, have been implicated in tumor progression (65, 77, 134). Furthermore, several reports suggest that cancer cells induce metabolic changes in adipocytes and stimulate delipidation of mature differentiated adipocytes to release their lipids and promote tumor progression (4, 46, 109).
In a study using adipocyte-conditioned medium and primary human omental adipocytes, Nieman and colleagues showed that adipocytes promote homing, migration, and invasion of ovarian cancer cells, possibly by soluble factors (109). A cytokine array and antibody-mediated inhibition indicated that adipocyte-secreted IL-6 and IL-8 promoted the early steps of ovarian cancer metastasis to the omentum, including homing and adhesion. Furthermore, in co-culturing adipocytes loaded with fluorescently labeled lipids and SKOV3ip1 ovarian cancer cells, Nieman and colleagues demonstrated direct transfer of fatty acids from adipocytes to ovarian cancer cells. In addition, co-cultivation of ovarian cancer cells with adipocytes activated hormone-sensitive lipase-mediated lipolysis in adipocytes and β-oxidation in cancer cells, suggesting that adipocytes support ovarian cancer cell growth by providing energy-dense lipids to and stimulating mitochondrion metabolism in cancer cells. Consistent with the role of lipids as an energy source for ovarian cancer cell growth, Clark and colleagues observed an inverse relationship between the metastatic burden and the adipocyte content in the omentum in in vivo time-course studies (40). Finally, upregulation of fatty acid-binding protein 4 expression in omental metastases and a drastically reduced tumor burden in a fatty acid-binding protein 4-deficient mouse model indicate that this protein plays a key role in ovarian cancer metastasis (109).
However, because obesity is not significantly associated with ovarian cancer incidence and mortality (23, 129), the tumor-promoting effect of adipocytes is believed to be a local effect mediated by direct contact of cancer-associated adipocytes with ovarian cancer cells and/or paracrine signaling, leading to remodeling of the tumor microenvironment (110). Recent findings demonstrate that treatment with metformin inhibits adipocyte-mediated ovarian cancer cell proliferation, migration, and adipogenesis via regulation of transcription factors (CCAAT-enhancer binding protein-α, CCAAT-enhancer binding protein-β, and sterol regulatory element-binding protein-1) and activation of 5′-AMP-activated protein kinase (154). These findings demonstrate that metformin is a potential therapeutic agent for ovarian cancer via targeting of the interaction between adipocytes and ovarian cancer cells.
Clinical Implications of Stromal Involvement in Ovarian Cancer Metastasis
Involvement of four major stromal cell types, CAFs, endothelial cells, cancer-associated macrophages, and CAAs, has been demonstrated in ovarian cancer progression and metastasis in multiple studies. Despite increasing understanding of stromal involvement in ovarian cancer metastasis, therapeutic regimens that target the ovarian cancer stromal components are still relatively underdeveloped. Because therapeutic approaches that target both cancer cells and the tumor-supporting microenvironment may be more effective than those that target cancer cells alone, investigators are actively pursuing approaches that can suppress tumor metastasis and progression by targeting tumor stroma. To that end, researchers have begun to evaluate the potential of inhibiting tumor progression via stromal ablation. However, multiple studies demonstrate that ablation of CAFs in tumors actually promotes tumor metastasis and progression and reduces survival durations in mice (118, 130). One of the possible reasons for these findings is that global depletion of CAFs may drastically change the architecture of tumor tissue as well as the cell-cell interactions in the tumor microenvironment; these changes may actually promote tumor progression and reduce survival durations. Therefore, instead of global stromal ablation, researchers have proposed that CAF reprogramming by targeting specific CAF-derived tumor-promoting factors may be a more effective therapeutic approach for cancer (167). Identification and validation of metastasis-promoting genes are crucial for achieving the goal of suppressing tumor metastasis by targeting stroma-derived factors. However, identification of these genes and evaluation of their functional roles in a systemic approach remain challenging because of complex interactions among multiple cell types in tumor tissues. Because bulk tumor samples often contain various numbers of stromal components, normalization and identification of stromal-specific tumor-promoting factors may be problematic. To address this, researchers have performed transcriptome profiling and next-generation sequencing using microdissected tumors. For example, large-scale expression profiling of ovarian CAFs (93, 168) and endothelial cells (21) has identified stromal-specific factors that are highly correlated with clinical outcomes and disease progression in ovarian cancer patients. In addition to generation of highly cell-type-specific expression profiles using laser microdissection, computational scientists have used these expression profiles to identify cross-talk networks of cancer and stromal cells and potential signaling pathways involved in disease progression (37). A combination of these approaches would allow for identification of stroma-derived metastasis-promoting factors for the development of targeted therapies for ovarian cancer.
Despite the challenges faced by researchers and clinicians during the development of stroma-targeting therapies, great research effort has been put into uncovering the mechanisms involved in stroma-mediated tumor growth, metastasis, and drug resistance. With increasing understanding of stroma-tumor interactions, new therapeutic targets from the tumor stroma are being identified. For example, recently, Chi and colleagues reported that tumor-associated macrophages and fibroblasts respond to cisplatin and 5-fluorouracil treatment by activating the CCAAT/enhancer binding protein-δ, which subsequently transcriptionally upregulates pentraxin 3 (36). Using a pentraxin 3 peptide inhibitor, RI37, researchers were able to suppress cancer progression and metastasis (36). Also targeting tumor-associated macrophages, Cieslewicz and colleagues described the use of proapoptotic peptides that target M2 macrophages in mouse (39). The peptide sequence that preferentially binds to murine M2 cells was fused with a proapoptotic peptide and then delivered into colon cancer-bearing mice via tail vein injection. Animals treated with M2 macrophage-targeting fusion peptide had significantly delayed mortality and a selective reduction in M2-like tumor-associated macrophage population (39). The endothelial cell has long been the target for antiangiogenic therapies. In addition to bevacizumab, the VEGF-targeting monoclonal antibody mentioned above, the therapeutic efficacy on ovarian cancer of multiple antiangiogenic small-molecule tyrosine kinase inhibitors has been evaluated in phase 2 and 3 clinical trials (60). While sunitinib, which targets VEGFR, platelet-derived growth factor receptor (PDGFR), RET, FLT3, c-kit, and colony-stimulating factor 1 (38), demonstrated modest activity in ovarian cancer (15, 24, 92), pazopanib, which targets VEGFR1/2/3, PDGFR, and c-kit (138), demonstrated significant improvement in patient progression-free survival (PFS) compared with placebo (hazard ratio = 0.77, P = 0.0021) in the AGO-OVAR 16 trial, in which 940 International Federation of Gynecology and Obstetrics stage 2–4 ovarian cancer patients were enrolled (48). Cediranib, a tyrosine kinase inhibitor that binds VEGFR, PDGFR, and c-kit (159), has been tested in a recent phase 2 study in combination with olaparib, a poly(ADP-ribose) polymerase (PARP) inhibitor. Ninety ovarian cancer patients were randomized to receive either PARP inhibitor alone or PARP inhibitor in combination with cediranid. Clinical trial results showed that median PFS is significantly longer for the combination arm than the PARP inhibitor single-agent arm (17.7 mo vs. 9.0 mo, P = 0.005) (95). These research findings suggest that, in addition, to single-agent therapies that target the stromal microenvironment, stroma-targeting agents could be used in combination with small-molecule drugs (e.g., PARP inhibitors) or cytotoxic chemoagents (e.g., cisplatin and paclitaxel) to achieve a higher therapeutic efficacy.
With increasing understanding of the molecular mechanisms by which stromal cells support ovarian tumor progression and metastasis, as well as chemoresistance, new therapeutic targets are being discovered and validated. While the implementation of new therapeutic agents that target newly discovered proteins or signaling pathways could be costly and time-consuming, drug repositioning by placing noncancer therapies into new therapeutic niches offers an alternative approach for the discovery of targeted therapeutic agents (135). With the aid of modern bioinformatics tools and analysis platforms, US Food and Drug Administration-approved drugs with favorable toxicity profile can be efficiently prioritized and integrated into existing drug-screening pipelines (59, 80). Although further clinical and epidemiological studies are required, several repositioned drugs, including peroxisome proliferator-activated receptor ligands and ritonavir, have demonstrated a tumor-suppressive effect in ovarian cancer (8). While current drug-repositioning efforts have mostly focused on targeting cancer cells, we believe that repositioned drugs can also be used to target stromal cells and their cross talks with cancer cells. Taking tumor-stroma interaction into account when identifying repositioned drugs would facilitate the identification of effective therapeutic agents.
Conclusions
Ovarian cancer metastasis is a highly regulated process that involves interaction between cancer cells that either disseminate from the primary tumor into the peritoneal cavity or migrate via the bloodstream and the omentum, which is the preferred metastatic site. In this review we described the two major routes of ovarian cancer metastasis and the molecular mechanisms involved in conferring motility to metastatic ovarian cancer cells. We also highlighted the involvement and roles of different stromal cell types, including CAFs, endothelial cells, cancer-associated macrophages, and CAAs, in metastasis. With increasing knowledge of the overall ovarian cancer pathogenesis and mechanisms of ovarian cancer metastasis, new approaches to the management of metastatic disease may be developed. More importantly, with researchers further delineating the roles of different stromal components in the tumor microenvironment in ovarian cancer progression and metastasis, an increasingly comprehensive landscape of the disease is being generated. With authors reporting on newly identified and validated prognostic markers and therapeutic targets, novel effective treatment regimens are being developed.
GRANTS
This work was supported in part by National Cancer Institute Grants R01 CA-133057 and R01 CA-142832, Cancer Prevention Research Institute of Texas Grant RP100094, and a grant from the Mary K. Chapman Foundation.
DISCLOSURES
No conflicts of interest, financial or otherwise, are declared by the authors.
AUTHOR CONTRIBUTIONS
T.-L.Y. and C.S.L. prepared the figures; T.-L.Y., C.S.L., K.-P.Y., C.L.A.Y., S.T.W., and S.C.M. drafted the manuscript; T.-L.Y., C.S.L., K.-P.Y., S.T.W., and S.C.M. edited and revised the manuscript; T.-L.Y., C.S.L., K.-P.Y., C.L.A.Y., S.T.W., and S.C.M. approved the final version of the manuscript.
REFERENCES
- 1.Adib TR, Henderson S, Perrett C, Hewitt D, Bourmpoulia D, Ledermann J, Boshoff C. Predicting biomarkers for ovarian cancer using gene-expression microarrays. Br J Cancer 90: 686–692, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Akahane T, Hirasawa A, Tsuda H, Kataoka F, Nishimura S, Tanaka H, Tominaga E, Nomura H, Chiyoda T, Iguchi Y, Yamagami W, Susumu N, Aoki D. The origin of stroma surrounding epithelial ovarian cancer cells. Int J Gynecol Pathol 32: 26–30, 2013. [DOI] [PubMed] [Google Scholar]
- 3.Akin O, Sala E, Moskowitz CS, Ishill N, Soslow RA, Chi DS, Hricak H. Perihepatic metastases from ovarian cancer: sensitivity and specificity of CT for the detection of metastases with and those without liver parenchymal invasion. Radiology 248: 511–517, 2008. [DOI] [PubMed] [Google Scholar]
- 4.Andarawewa KL, Motrescu ER, Chenard MP, Gansmuller A, Stoll I, Tomasetto C, Rio MC. Stromelysin-3 is a potent negative regulator of adipogenesis participating to cancer cell-adipocyte interaction/crosstalk at the tumor invasive front. Cancer Res 65: 10862–10871, 2005. [DOI] [PubMed] [Google Scholar]
- 5.Baba T, Kariya M, Higuchi T, Mandai M, Matsumura N, Kondoh E, Miyanishi M, Fukuhara K, Takakura K, Fujii S. Neuropilin-1 promotes unlimited growth of ovarian cancer by evading contact inhibition. Gynecol Oncol 105: 703–711, 2007. [DOI] [PubMed] [Google Scholar]
- 6.Baldwin RL, Tran H, Karlan BY. Loss of c-myc repression coincides with ovarian cancer resistance to transforming growth factor-β growth arrest independent of transforming growth factor-β/Smad signaling. Cancer Res 63: 1413–1419, 2003. [PubMed] [Google Scholar]
- 7.Balkwill F. Cancer and the chemokine network. Nat Rev Cancer 4: 540–550, 2004. [DOI] [PubMed] [Google Scholar]
- 8.Banno K, Iida M, Yanokura M, Irie H, Masuda K, Kobayashi Y, Tominaga E, Aoki D. Drug repositioning for gynecologic tumors: a new therapeutic strategy for cancer. Sci World J 2015: 341362, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Bansal S, Kaur K, Bansal AK. Diagnosing ascitic etiology on a biochemical basis. Hepatogastroenterology 45: 1673–1677, 1998. [PubMed] [Google Scholar]
- 10.Bast RC Jr, Hennessy B, Mills GB. The biology of ovarian cancer: new opportunities for translation. Nat Rev Cancer 9: 415–428, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Beelen RH, Fluitsma DM, Hoefsmit EC. The cellular composition of omentum milky spots and the ultrastructure of milky spot macrophages and reticulum cells. J Reticuloendothel Soc 28: 585–599, 1980. [PubMed] [Google Scholar]
- 12.Bellingan GJ, Caldwell H, Howie SE, Dransfield I, Haslett C. In vivo fate of the inflammatory macrophage during the resolution of inflammation: inflammatory macrophages do not die locally, but emigrate to the draining lymph nodes. J Immunol 157: 2577–2585, 1996. [PubMed] [Google Scholar]
- 13.Belotti D, Paganoni P, Manenti L, Garofalo A, Marchini S, Taraboletti G, Giavazzi R. Matrix metalloproteinases (MMP9 and MMP2) induce the release of vascular endothelial growth factor (VEGF) by ovarian carcinoma cells: implications for ascites formation. Cancer Res 63: 5224–5229, 2003. [PubMed] [Google Scholar]
- 14.Berchuck A, Iversen ES, Lancaster JM, Pittman J, Luo J, Lee P, Murphy S, Dressman HK, Febbo PG, West M, Nevins JR, Marks JR. Patterns of gene expression that characterize long-term survival in advanced stage serous ovarian cancers. Clin Cancer Res 11: 3686–3696, 2005. [DOI] [PubMed] [Google Scholar]
- 15.Biagi JJ, Oza AM, Chalchal HI, Grimshaw R, Ellard SL, Lee U, Hirte H, Sederias J, Ivy SP, Eisenhauer EA. A phase II study of sunitinib in patients with recurrent epithelial ovarian and primary peritoneal carcinoma: an NCIC Clinical Trials Group Study. Ann Oncol 22: 335–340, 2011. [DOI] [PubMed] [Google Scholar]
- 16.Bian D, Su S, Mahanivong C, Cheng RK, Han Q, Pan ZK, Sun P, Huang S. Lysophosphatidic acid stimulates ovarian cancer cell migration via a ras-MEK kinase 1 pathway. Cancer Res 64: 4209–4217, 2004. [DOI] [PubMed] [Google Scholar]
- 17.Birrer MJ, Johnson ME, Hao K, Wong KK, Park DC, Bell A, Welch WR, Berkowitz RS, Mok SC. Whole genome oligonucleotide-based array comparative genomic hybridization analysis identified fibroblast growth factor 1 as a prognostic marker for advanced-stage serous ovarian adenocarcinomas. J Clin Oncol 25: 2281–2287, 2007. [DOI] [PubMed] [Google Scholar]
- 18.Blood CH, Zetter BR. Tumor interactions with the vasculature: angiogenesis and tumor metastasis. Biochim Biophys Acta 1032: 89–118, 1990. [DOI] [PubMed] [Google Scholar]
- 19.Brooks AN, Kilgour E, Smith PD. Molecular pathways: fibroblast growth factor signaling: a new therapeutic opportunity in cancer. Clin Cancer Res 18: 1855–1862, 2012. [DOI] [PubMed] [Google Scholar]
- 20.Brundage RA, Fogarty KE, Tuft RA, Fay FS. Calcium gradients underlying polarization and chemotaxis of eosinophils. Science 254: 703–706, 1991. [DOI] [PubMed] [Google Scholar]
- 21.Buckanovich RJ, Facciabene A, Kim S, Benencia F, Sasaroli D, Balint K, Katsaros D, O'Brien-Jenkins A, Gimotty PA, Coukos G. Endothelin B receptor mediates the endothelial barrier to T cell homing to tumors and disables immune therapy. Nat Med 14: 28–36, 2008. [DOI] [PubMed] [Google Scholar]
- 22.Burger RA, Brady MF, Bookman MA, Fleming GF, Monk BJ, Huang H, Mannel RS, Homesley HD, Fowler J, Greer BE, Boente M, Birrer MJ, Liang SX, Gynecologic Oncology Group. Incorporation of bevacizumab in the primary treatment of ovarian cancer. N Engl J Med 365: 2473–2483, 2011. [DOI] [PubMed] [Google Scholar]
- 23.Calle EE, Rodriguez C, Walker-Thurmond K, Thun MJ. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of US adults. N Engl J Med 348: 1625–1638, 2003. [DOI] [PubMed] [Google Scholar]
- 24.Campos SM, Penson RT, Matulonis U, Horowitz NS, Whalen C, Pereira L, Tyburski K, Roche M, Szymonifka J, Berlin S. A phase II trial of sunitinib malate in recurrent and refractory ovarian, fallopian tube and peritoneal carcinoma. Gynecol Oncol 128: 215–220, 2013. [DOI] [PubMed] [Google Scholar]
- 25.Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 474: 609–615, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Canoglu A, Gogus C, Beduk Y, Orhan D, Tulunay O, Baltaci S. Microvessel density as a prognostic marker in bladder carcinoma: correlation with tumor grade, stage and prognosis. Int Urol Nephrol 36: 401–405, 2004. [DOI] [PubMed] [Google Scholar]
- 27.Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature 473: 298–307, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Carmignani CP, Sugarbaker TA, Bromley CM, Sugarbaker PH. Intraperitoneal cancer dissemination: mechanisms of the patterns of spread. Cancer Metastasis Rev 22: 465–472, 2003. [DOI] [PubMed] [Google Scholar]
- 29.Casey RC, Skubitz AP. CD44 and β1-integrins mediate ovarian carcinoma cell migration toward extracellular matrix proteins. Clin Exp Metastasis 18: 67–75, 2000. [DOI] [PubMed] [Google Scholar]
- 30.Castadot P, Magne N, Berghmans T, Drowart A, Baeyens L, Smets D, Van Houtte P. [Ovarian metastasis and lung adenocarcinoma: a case report]. Cancer Radiother 9: 183–186, 2005. [DOI] [PubMed] [Google Scholar]
- 31.Chambers AF, Groom AC, MacDonald IC. Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer 2: 563–572, 2002. [DOI] [PubMed] [Google Scholar]
- 32.Chang SC, Lin PC, Yang SH, Wang HS, Li AF, Lin JK. SDF-1α G801A polymorphism predicts lymph node metastasis in stage T3 colorectal cancer. Ann Surg Oncol 16: 2323–2330, 2009. [DOI] [PubMed] [Google Scholar]
- 33.Chavez-Macgregor M, Aviles-Salas A, Green D, Fuentes-Alburo A, Gomez-Ruiz C, Aguayo A. Angiogenesis in the bone marrow of patients with breast cancer. Clin Cancer Res 11: 5396–5400, 2005. [DOI] [PubMed] [Google Scholar]
- 34.Chen H, Ye D, Xie X, Chen B, Lu W. VEGF, VEGFRs expressions and activated STATs in ovarian epithelial carcinoma. Gynecol Oncol 94: 630–635, 2004. [DOI] [PubMed] [Google Scholar]
- 35.Chhabra ES, Higgs HN. The many faces of actin: matching assembly factors with cellular structures. Nat Cell Biol 9: 1110–1121, 2007. [DOI] [PubMed] [Google Scholar]
- 36.Chi JY, Hsiao YW, Li CF, Lo YC, Lin ZY, Hong JY, Liu YM, Han X, Wang SM, Chen BK, Tsai KK, Wang JM. Targeting chemotherapy-induced PTX3 in tumor stroma to prevent the progression of drug-resistant cancers. Oncotarget. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Choi H, Sheng J, Gao D, Li F, Durrans A, Ryu S, Lee SB, Narula N, Rafii S, Elemento O, Altorki NK, Wong ST, Mittal V. Transcriptome analysis of individual stromal cell populations identifies stroma-tumor crosstalk in mouse lung cancer model. Cell Rep 10: 1187–1201, 2015. [DOI] [PubMed] [Google Scholar]
- 38.Chow LQ, Eckhardt SG. Sunitinib: from rational design to clinical efficacy. J Clin Oncol 25: 884–896, 2007. [DOI] [PubMed] [Google Scholar]
- 39.Cieslewicz M, Tang J, Yu JL, Cao H, Zavaljevski M, Motoyama K, Lieber A, Raines EW, Pun SH. Targeted delivery of proapoptotic peptides to tumor-associated macrophages improves survival. Proc Natl Acad Sci USA 110: 15919–15924, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Clark R, Krishnan V, Schoof M, Rodriguez I, Theriault B, Chekmareva M, Rinker-Schaeffer C. Milky spots promote ovarian cancer metastatic colonization of peritoneal adipose in experimental models. Am J Pathol 183: 576–591, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Coakley FV, Hricak H. Imaging of peritoneal and mesenteric disease: key concepts for the clinical radiologist. Clin Radiol 54: 563–574, 1999. [DOI] [PubMed] [Google Scholar]
- 42.Cristancho AG, Lazar MA. Forming functional fat: a growing understanding of adipocyte differentiation. Nat Rev Mol Cell Biol 12: 722–734, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.D'Amore PA. Capillary growth: a two-cell system. Semin Cancer Biol 3: 49–56, 1992. [PubMed] [Google Scholar]
- 44.Daly AJ, McIlreavey L, Irwin CR. Regulation of HGF and SDF-1 expression by oral fibroblasts—implications for invasion of oral cancer. Oral Oncol 44: 646–651, 2008. [DOI] [PubMed] [Google Scholar]
- 45.Daya D, McCaughey WT. Pathology of the peritoneum: a review of selected topics. Semin Diagn Pathol 8: 277–289, 1991. [PubMed] [Google Scholar]
- 46.Dirat B, Bochet L, Dabek M, Daviaud D, Dauvillier S, Majed B, Wang YY, Meulle A, Salles B, Le Gonidec S, Garrido I, Escourrou G, Valet P, Muller C. Cancer-associated adipocytes exhibit an activated phenotype and contribute to breast cancer invasion. Cancer Res 71: 2455–2465, 2011. [DOI] [PubMed] [Google Scholar]
- 47.Dirat B, Bochet L, Escourrou G, Valet P, Muller C. Unraveling the obesity and breast cancer links: a role for cancer-associated adipocytes? Endocr Dev 19: 45–52, 2010. [DOI] [PubMed] [Google Scholar]
- 48.du Bois A, Floquet A, Kim JW, Rau J, del Campo JM, Friedlander M, Pignata S, Fujiwara K, Vergote I, Colombo N, Mirza MR, Monk BJ, Kimmig R, Ray-Coquard I, Zang R, Diaz-Padilla I, Baumann KH, Mouret-Reynier MA, Kim JH, Kurzeder C, Lesoin A, Vasey P, Marth C, Canzler U, Scambia G, Shimada M, Calvert P, Pujade-Lauraine E, Kim BG, Herzog TJ, Mitrica I, Schade-Brittinger C, Wang Q, Crescenzo R, Harter P. Incorporation of pazopanib in maintenance therapy of ovarian cancer. J Clin Oncol 32: 3374–3382, 2014. [DOI] [PubMed] [Google Scholar]
- 49.Egeblad M, Werb Z. New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer 2: 161–174, 2002. [DOI] [PubMed] [Google Scholar]
- 50.Fang X, Schummer M, Mao M, Yu S, Tabassam FH, Swaby R, Hasegawa Y, Tanyi JL, LaPushin R, Eder A, Jaffe R, Erickson J, Mills GB. Lysophosphatidic acid is a bioactive mediator in ovarian cancer. Biochim Biophys Acta 1582: 257–264, 2002. [DOI] [PubMed] [Google Scholar]
- 51.Feki A, Berardi P, Bellingan G, Major A, Krause KH, Petignat P, Zehra R, Pervaiz S, Irminger-Finger I. Dissemination of intraperitoneal ovarian cancer: discussion of mechanisms and demonstration of lymphatic spreading in ovarian cancer model. Crit Rev Oncol Hematol 72: 1–9, 2009. [DOI] [PubMed] [Google Scholar]
- 52.Fishman DA, Bafetti LM, Banionis S, Kearns AS, Chilukuri K, Stack MS. Production of extracellular matrix-degrading proteinases by primary cultures of human epithelial ovarian carcinoma cells. Cancer 80: 1457–1463, 1997. [DOI] [PubMed] [Google Scholar]
- 53.Fishman DA, Liu Y, Ellerbroek SM, Stack MS. Lysophosphatidic acid promotes matrix metalloproteinase (MMP) activation and MMP-dependent invasion in ovarian cancer cells. Cancer Res 61: 3194–3199, 2001. [PubMed] [Google Scholar]
- 54.Folkman J. The role of angiogenesis in tumor growth. Semin Cancer Biol 3: 65–71, 1992. [PubMed] [Google Scholar]
- 55.Folkman J, Ingber D. Inhibition of angiogenesis. Semin Cancer Biol 3: 89–96, 1992. [PubMed] [Google Scholar]
- 56.Fournier MF, Sauser R, Ambrosi D, Meister JJ, Verkhovsky AB. Force transmission in migrating cells. J Cell Biol 188: 287–297, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Freedman RS, Deavers M, Liu J, Wang E. Peritoneal inflammation: a microenvironment for epithelial ovarian cancer (EOC). J Transl Med 2: 23, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Friesel RE, Maciag T. Molecular mechanisms of angiogenesis: fibroblast growth factor signal transduction. FASEB J 9: 919–925, 1995. [DOI] [PubMed] [Google Scholar]
- 59.Fu C, Jin G, Gao J, Zhu R, Ballesteros-Villagrana E, Wong ST. DrugMap Central: an on-line query and visualization tool to facilitate drug repositioning studies. Bioinformatics 29: 1834–1836, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Gadducci A, Lanfredini N, Sergiampietri C. Antiangiogenic agents in gynecological cancer: state of art and perspectives of clinical research. Crit Rev Oncol Hematol. In press. [DOI] [PubMed] [Google Scholar]
- 61.Gerber SA, Rybalko VY, Bigelow CE, Lugade AA, Foster TH, Frelinger JG, Lord EM. Preferential attachment of peritoneal tumor metastases to omental immune aggregates and possible role of a unique vascular microenvironment in metastatic survival and growth. Am J Pathol 169: 1739–1752, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Goel HL, Mercurio AM. VEGF targets the tumour cell. Nat Rev Cancer 13: 871–882, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Goff BA, Mandel L, Muntz HG, Melancon CH. Ovarian carcinoma diagnosis. Cancer 89: 2068–2075, 2000. [DOI] [PubMed] [Google Scholar]
- 64.Gossmann A, Helbich TH, Mesiano S, Shames DM, Wendland MF, Roberts TP, Ferrara N, Jaffe RB, Brasch RC. Magnetic resonance imaging in an experimental model of human ovarian cancer demonstrating altered microvascular permeability after inhibition of vascular endothelial growth factor. Am J Obstet Gynecol 183: 956–963, 2000. [DOI] [PubMed] [Google Scholar]
- 65.Guerrero J, Tobar N, Caceres M, Espinoza L, Escobar P, Dotor J, Smith PC, Martinez J. Soluble factors derived from tumor mammary cell lines induce a stromal mammary adipose reversion in human and mice adipose cells. Possible role of TGF-β1 and TNF-α. Breast Cancer Res Treat 119: 497–508, 2010. [DOI] [PubMed] [Google Scholar]
- 66.Gunning PW, Schevzov G, Kee AJ, Hardeman EC. Tropomyosin isoforms: divining rods for actin cytoskeleton function. Trends Cell Biol 15: 333–341, 2005. [DOI] [PubMed] [Google Scholar]
- 67.Hagemann T, Wilson J, Kulbe H, Li NF, Leinster DA, Charles K, Klemm F, Pukrop T, Binder C, Balkwill FR. Macrophages induce invasiveness of epithelial cancer cells via NF-κB and JNK. J Immunol 175: 1197–1205, 2005. [DOI] [PubMed] [Google Scholar]
- 68.Hagiwara A, Takahashi T, Sawai K, Taniguchi H, Shimotsuma M, Okano S, Sakakura C, Tsujimoto H, Osaki K, Sasaki S, et al. Milky spots as the implantation site for malignant cells in peritoneal dissemination in mice. Cancer Res 53: 687–692, 1993. [PubMed] [Google Scholar]
- 69.Hahn K, DeBiasio R, Taylor DL. Patterns of elevated free calcium and calmodulin activation in living cells. Nature 359: 736–738, 1992. [DOI] [PubMed] [Google Scholar]
- 70.Hanahan D, Folkman J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 86: 353–364, 1996. [DOI] [PubMed] [Google Scholar]
- 71.Haskill S, Koren H, Becker S, Fowler W, Walton L. Mononuclear-cell infiltration in ovarian cancer. III. Suppressor-cell and ADCC activity of macrophages from ascitic and solid ovarian tumours. Br J Cancer 45: 747–753, 1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Haverty PM, Hon LS, Kaminker JS, Chant J, Zhang Z. High-resolution analysis of copy number alterations and associated expression changes in ovarian tumors. BMC Med Genomics 2: 21, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Hibbs K, Skubitz KM, Pambuccian SE, Casey RC, Burleson KM, Oegema TR Jr, Thiele JJ, Grindle SM, Bliss RL, Skubitz AP. Differential gene expression in ovarian carcinoma: identification of potential biomarkers. Am J Pathol 165: 397–414, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Huang M, Li Y, Zhang H, Nan F. Breast cancer stromal fibroblasts promote the generation of CD44+CD24− cells through SDF-1/CXCR4 interaction. J Exp Clin Cancer Res 29: 80, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Huang S, Van Arsdall M, Tedjarati S, McCarty M, Wu W, Langley R, Fidler IJ. Contributions of stromal metalloproteinase-9 to angiogenesis and growth of human ovarian carcinoma in mice. J Natl Cancer Inst 94: 1134–1142, 2002. [DOI] [PubMed] [Google Scholar]
- 76.Imai T, Horiuchi A, Shiozawa T, Osada R, Kikuchi N, Ohira S, Oka K, Konishi I. Elevated expression of E-cadherin and α-, β-, and γ-catenins in metastatic lesions compared with primary epithelial ovarian carcinomas. Hum Pathol 35: 1469–1476, 2004. [DOI] [PubMed] [Google Scholar]
- 77.Inouye KE, Shi H, Howard JK, Daly CH, Lord GM, Rollins BJ, Flier JS. Absence of CC chemokine ligand 2 does not limit obesity-associated infiltration of macrophages into adipose tissue. Diabetes 56: 2242–2250, 2007. [DOI] [PubMed] [Google Scholar]
- 78.Israeli O, Gotlieb WH, Friedman E, Korach J, Friedman E, Goldman B, Zeltzer A, Ben-Baruch G, Rienstein S, Aviram-Goldring A. Genomic analyses of primary and metastatic serous epithelial ovarian cancer. Cancer Genet Cytogenet 154: 16–21, 2004. [DOI] [PubMed] [Google Scholar]
- 79.Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science 307: 58–62, 2005. [DOI] [PubMed] [Google Scholar]
- 80.Jin G, Wong ST. Toward better drug repositioning: prioritizing and integrating existing methods into efficient pipelines. Drug Discov Today 19: 637–644, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Joyce JA. Therapeutic targeting of the tumor microenvironment. Cancer Cell 7: 513–520, 2005. [DOI] [PubMed] [Google Scholar]
- 82.Kenny HA, Chiang CY, White EA, Schryver EM, Habis M, Romero IL, Ladanyi A, Penicka CV, George J, Matlin K, Montag A, Wroblewski K, Yamada SD, Mazar AP, Bowtell D, Lengyel E. Mesothelial cells promote early ovarian cancer metastasis through fibronectin secretion. J Clin Invest 124: 4614–4628, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Kenny HA, Dogan S, Zillhardt MK, Mitra A, Yamada SD, Krausz T, Lengyel E. Organotypic models of metastasis: a three-dimensional culture mimicking the human peritoneum and omentum for the study of the early steps of ovarian cancer metastasis. Cancer Treat Res 149: 335–351, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Kenny HA, Kaur S, Coussens LM, Lengyel E. The initial steps of ovarian cancer cell metastasis are mediated by MMP-2 cleavage of vitronectin and fibronectin. J Clin Invest 118: 1367–1379, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Kenny HA, Krausz T, Yamada SD, Lengyel E. Use of a novel 3D culture model to elucidate the role of mesothelial cells, fibroblasts and extra-cellular matrices on adhesion and invasion of ovarian cancer cells to the omentum. Int J Cancer 121: 1463–1472, 2007. [DOI] [PubMed] [Google Scholar]
- 86.Korkolopoulou P, Thymara I, Kavantzas N, Vassilakopoulos TP, Angelopoulou MK, Kokoris SI, Dimitriadou EM, Siakantaris MP, Anargyrou K, Panayiotidis P, Tsenga A, Androulaki A, Doussis-Anagnostopoulou IA, Patsouris E, Pangalis GA. Angiogenesis in Hodgkin's lymphoma: a morphometric approach in 286 patients with prognostic implications. Leukemia 19: 894–900, 2005. [DOI] [PubMed] [Google Scholar]
- 87.Kraft A, Weindel K, Ochs A, Marth C, Zmija J, Schumacher P, Unger C, Marme D, Gastl G. Vascular endothelial growth factor in the sera and effusions of patients with malignant and nonmalignant disease. Cancer 85: 178–187, 1999. [PubMed] [Google Scholar]
- 88.Krist LF, Kerremans M, Broekhuis-Fluitsma DM, Eestermans IL, Meyer S, Beelen RH. Milky spots in the greater omentum are predominant sites of local tumour cell proliferation and accumulation in the peritoneal cavity. Cancer Immunol Immunother 47: 205–212, 1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Labiche A, Heutte N, Herlin P, Chasle J, Gauduchon P, Elie N. Stromal compartment as a survival prognostic factor in advanced ovarian carcinoma. Int J Gynecol Cancer 20: 28–33, 2010. [DOI] [PubMed] [Google Scholar]
- 90.Lancaster JM, Dressman HK, Whitaker RS, Havrilesky L, Gray J, Marks JR, Nevins JR, Berchuck A. Gene expression patterns that characterize advanced stage serous ovarian cancers. J Soc Gynecol Investig 11: 51–59, 2004. [DOI] [PubMed] [Google Scholar]
- 91.Lengyel E. Ovarian cancer development and metastasis. Am J Pathol 177: 1053–1064, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Leone Roberti Maggiore U, Valenzano Menada M, Venturini PL, Ferrero S. The potential of sunitinib as a therapy in ovarian cancer. Expert Opin Investig Drugs 22: 1671–1686, 2013. [DOI] [PubMed] [Google Scholar]
- 93.Leung CS, Yeung TL, Yip KP, Pradeep S, Balasubramanian L, Liu J, Wong KK, Mangala LS, Armaiz-Pena GN, Lopez-Berestein G, Sood AK, Birrer MJ, Mok SC. Calcium-dependent FAK/CREB/TNNC1 signalling mediates the effect of stromal MFAP5 on ovarian cancer metastatic potential. Nat Commun 5: 5092, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Lim MC, Kang S, Lee KS, Han SS, Park SJ, Seo SS, Park SY. The clinical significance of hepatic parenchymal metastasis in patients with primary epithelial ovarian cancer. Gynecol Oncol 112: 28–34, 2009. [DOI] [PubMed] [Google Scholar]
- 95.Liu JF, Barry WT, Birrer M, Lee JM, Buckanovich RJ, Fleming GF, Rimel B, Buss MK, Nattam S, Hurteau J, Luo W, Quy P, Whalen C, Obermayer L, Lee H, Winer EP, Kohn EC, Ivy SP, Matulonis UA. Combination cediranib and olaparib versus olaparib alone for women with recurrent platinum-sensitive ovarian cancer: a randomised phase 2 study. Lancet Oncol 15: 1207–1214, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Liu Y, Hu T, Shen J, Li SF, Lin JW, Zheng XH, Gao QH, Zhou HM. Separation, cultivation and biological characteristics of oral carcinoma-associated fibroblasts. Oral Dis 12: 375–380, 2006. [DOI] [PubMed] [Google Scholar]
- 97.Manabe Y, Toda S, Miyazaki K, Sugihara H. Mature adipocytes, but not preadipocytes, promote the growth of breast carcinoma cells in collagen gel matrix culture through cancer-stromal cell interactions. J Pathol 201: 221–228, 2003. [DOI] [PubMed] [Google Scholar]
- 98.Matsuo Y, Ochi N, Sawai H, Yasuda A, Takahashi H, Funahashi H, Takeyama H, Tong Z, Guha S. CXCL8/IL-8 and CXCL12/SDF-1α co-operatively promote invasiveness and angiogenesis in pancreatic cancer. Int J Cancer 124: 853–861, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.McGrail DJ, Kieu QM, Dawson MR. The malignancy of metastatic ovarian cancer cells is increased on soft matrices through a mechanosensitive Rho-ROCK pathway. J Cell Sci 127: 2621–2626, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Mesiano S, Ferrara N, Jaffe RB. Role of vascular endothelial growth factor in ovarian cancer: inhibition of ascites formation by immunoneutralization. Am J Pathol 153: 1249–1256, 1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Meyers MA, Oliphant M, Berne AS, Feldberg MA. The peritoneal ligaments and mesenteries: pathways of intraabdominal spread of disease. Radiology 163: 593–604, 1987. [DOI] [PubMed] [Google Scholar]
- 102.Micke P, Ostman A. Tumour-stroma interaction: cancer-associated fibroblasts as novel targets in anti-cancer therapy? Lung Cancer 45 Suppl 2: S163–S175, 2004. [DOI] [PubMed] [Google Scholar]
- 103.Mueller L, Goumas FA, Affeldt M, Sandtner S, Gehling UM, Brilloff S, Walter J, Karnatz N, Lamszus K, Rogiers X, Broering DC. Stromal fibroblasts in colorectal liver metastases originate from resident fibroblasts and generate an inflammatory microenvironment. Am J Pathol 171: 1608–1618, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Mueller MM, Fusenig NE. Friends or foes—bipolar effects of the tumour stroma in cancer. Nat Rev Cancer 4: 839–849, 2004. [DOI] [PubMed] [Google Scholar]
- 105.Murphy P, Alexander P, Senior PV, Fleming J, Kirkham N, Taylor I. Mechanisms of organ selective tumour growth by bloodborne cancer cells. Br J Cancer 57: 19–31, 1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Nagy JA, Masse EM, Herzberg KT, Meyers MS, Yeo KT, Yeo TK, Sioussat TM, Dvorak HF. Pathogenesis of ascites tumor growth: vascular permeability factor, vascular hyperpermeability, and ascites fluid accumulation. Cancer Res 55: 360–368, 1995. [PubMed] [Google Scholar]
- 107.Naora H, Montell DJ. Ovarian cancer metastasis: integrating insights from disparate model organisms. Nat Rev Cancer 5: 355–366, 2005. [DOI] [PubMed] [Google Scholar]
- 108.Nguyen DX, Bos PD, Massague J. Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer 9: 274–284, 2009. [DOI] [PubMed] [Google Scholar]
- 109.Nieman KM, Kenny HA, Penicka CV, Ladanyi A, Buell-Gutbrod R, Zillhardt MR, Romero IL, Carey MS, Mills GB, Hotamisligil GS, Yamada SD, Peter ME, Gwin K, Lengyel E. Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth. Nat Med 17: 1498–1503, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Nieman KM, Romero IL, Van Houten B, Lengyel E. Adipose tissue and adipocytes support tumorigenesis and metastasis. Biochim Biophys Acta 1831: 1533–1541, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Nishida N, Yano H, Komai K, Nishida T, Kamura T, Kojiro M. Vascular endothelial growth factor C and vascular endothelial growth factor receptor 2 are related closely to the prognosis of patients with ovarian carcinoma. Cancer 101: 1364–1374, 2004. [DOI] [PubMed] [Google Scholar]
- 112.Nowicka A, Marini FC, Solley TN, Elizondo PB, Zhang Y, Sharp HJ, Broaddus R, Kolonin M, Mok SC, Thompson MS, Woodward WA, Lu K, Salimian B, Nagrath D, Klopp AH. Human omental-derived adipose stem cells increase ovarian cancer proliferation, migration, and chemoresistance. PLos One 8: e81859, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Ohira S, Itatsu K, Sasaki M, Harada K, Sato Y, Zen Y, Ishikawa A, Oda K, Nagasaka T, Nimura Y, Nakanuma Y. Local balance of transforming growth factor-β1 secreted from cholangiocarcinoma cells and stromal-derived factor-1 secreted from stromal fibroblasts is a factor involved in invasion of cholangiocarcinoma. Pathol Int 56: 381–389, 2006. [DOI] [PubMed] [Google Scholar]
- 114.Ohira S, Sasaki M, Harada K, Sato Y, Zen Y, Isse K, Kozaka K, Ishikawa A, Oda K, Nimura Y, Nakanuma Y. Possible regulation of migration of intrahepatic cholangiocarcinoma cells by interaction of CXCR4 expressed in carcinoma cells with tumor necrosis factor-α and stromal-derived factor-1 released in stroma. Am J Pathol 168: 1155–1168, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Olson MF, Sahai E. The actin cytoskeleton in cancer cell motility. Clin Exp Metastasis 26: 273–287, 2009. [DOI] [PubMed] [Google Scholar]
- 116.Olsson AK, Dimberg A, Kreuger J, Claesson-Welsh L. VEGF receptor signalling—in control of vascular function. Nat Rev Mol Cell Biol 7: 359–371, 2006. [DOI] [PubMed] [Google Scholar]
- 117.Orimo A, Weinberg RA. Stromal fibroblasts in cancer: a novel tumor-promoting cell type. Cell Cycle 5: 1597–1601, 2006. [DOI] [PubMed] [Google Scholar]
- 118.Ozdemir BC, Pentcheva-Hoang T, Carstens JL, Zheng X, Wu CC, Simpson TR, Laklai H, Sugimoto H, Kahlert C, Novitskiy SV, De Jesus-Acosta A, Sharma P, Heidari P, Mahmood U, Chin L, Moses HL, Weaver VM, Maitra A, Allison JP, LeBleu VS, Kalluri R. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell 25: 719–734, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Paget S. The distribution of secondary growths in cancer of the breast. Cancer Metastasis Rev 8: 98–101, 1989. [PubMed] [Google Scholar]
- 120.Pecot CV, Bischoff FZ, Mayer JA, Wong KL, Pham T, Bottsford-Miller J, Stone RL, Lin YG, Jaladurgam P, Roh JW, Goodman BW, Merritt WM, Pircher TJ, Mikolajczyk SD, Nick AM, Celestino J, Eng C, Ellis LM, Deavers MT, Sood AK. A novel platform for detection of CK+ and CK− CTCs. Cancer Discov 1: 580–586, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Perren TJ, Swart AM, Pfisterer J, Ledermann JA, Pujade-Lauraine E, Kristensen G, Carey MS, Beale P, Cervantes A, Kurzeder C, du Bois A, Sehouli J, Kimmig R, Stahle A, Collinson F, Essapen S, Gourley C, Lortholary A, Selle F, Mirza MR, Leminen A, Plante M, Stark D, Qian W, Parmar MK, Oza AM, ICON7 Investigators. A phase 3 trial of bevacizumab in ovarian cancer. N Engl J Med 365: 2484–2496, 2011. [DOI] [PubMed] [Google Scholar]
- 122.Phillips KG, Velasco CR, Li J, Kolatkar A, Luttgen M, Bethel K, Duggan B, Kuhn P, McCarty OJ. Optical quantification of cellular mass, volume, and density of circulating tumor cells identified in an ovarian cancer patient. Front Oncol 2: 72, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Pollard TD, Borisy GG. Cellular motility driven by assembly and disassembly of actin filaments. Cell 112: 453–465, 2003. [DOI] [PubMed] [Google Scholar]
- 124.Pradeep S, Kim SW, Wu SY, Nishimura M, Chaluvally-Raghavan P, Miyake T, Pecot CV, Kim SJ, Choi HJ, Bischoff FZ, Mayer JA, Huang L, Nick AM, Hall CS, Rodriguez-Aguayo C, Zand B, Dalton HJ, Arumugam T, Lee HJ, Han HD, Cho MS, Rupaimoole R, Mangala LS, Sehgal V, Oh SC, Liu J, Lee JS, Coleman RL, Ram P, Lopez-Berestein G, Fidler IJ, Sood AK. Hematogenous metastasis of ovarian cancer: rethinking mode of spread. Cancer Cell 26: 77–91, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Prevarskaya N, Skryma R, Shuba Y. Calcium in tumour metastasis: new roles for known actors. Nat Rev Cancer 11: 609–618, 2011. [DOI] [PubMed] [Google Scholar]
- 126.Pustilnik TB, Estrella V, Wiener JR, Mao M, Eder A, Watt MA, Bast RC Jr, Mills GB. Lysophosphatidic acid induces urokinase secretion by ovarian cancer cells. Clin Cancer Res 5: 3704–3710, 1999. [PubMed] [Google Scholar]
- 127.Rafelski SM, Theriot JA. Crawling toward a unified model of cell mobility: spatial and temporal regulation of actin dynamics. Annu Rev Biochem 73: 209–239, 2004. [DOI] [PubMed] [Google Scholar]
- 128.Raftopoulou M, Hall A. Cell migration: Rho GTPases lead the way. Dev Biol 265: 23–32, 2004. [DOI] [PubMed] [Google Scholar]
- 129.Renehan AG, Tyson M, Egger M, Heller RF, Zwahlen M. Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies. Lancet 371: 569–578, 2008. [DOI] [PubMed] [Google Scholar]
- 130.Rhim AD, Oberstein PE, Thomas DH, Mirek ET, Palermo CF, Sastra SA, Dekleva EN, Saunders T, Becerra CP, Tattersall IW, Westphalen CB, Kitajewski J, Fernandez-Barrena MG, Fernandez-Zapico ME, Iacobuzio-Donahue C, Olive KP, Stanger BZ. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell 25: 735–747, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Ribas C, Colleoni GW, Silva MR, Carregoza MJ, Bordin JO. Prognostic significance of vascular endothelial growth factor immunoexpression in the context of adverse standard prognostic factors in multiple myeloma. Eur J Haematol 73: 311–317, 2004. [DOI] [PubMed] [Google Scholar]
- 132.Robinson-Smith TM, Isaacsohn I, Mercer CA, Zhou M, Van Rooijen N, Husseinzadeh N, McFarland-Mancini MM, Drew AF. Macrophages mediate inflammation-enhanced metastasis of ovarian tumors in mice. Cancer Res 67: 5708–5716, 2007. [DOI] [PubMed] [Google Scholar]
- 133.Runyon BA, Hoefs JC, Morgan TR. Ascitic fluid analysis in malignancy-related ascites. Hepatology 8: 1104–1109, 1988. [DOI] [PubMed] [Google Scholar]
- 134.Sartipy P, Loskutoff DJ. Monocyte chemoattractant protein 1 in obesity and insulin resistance. Proc Natl Acad Sci USA 100: 7265–7270, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Saxena A, Becker D, Preeshagul I, Lee K, Katz E, Levy B. Therapeutic effects of repurposed therapies in non-small cell lung cancer: what is old is new again. Oncologist 20: 934–945, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Schauer IG, Sood AK, Mok S, Liu J. Cancer-associated fibroblasts and their putative role in potentiating the initiation and development of epithelial ovarian cancer. Neoplasia 13: 393–405, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Schoppmann SF, Fenzl A, Nagy K, Unger S, Bayer G, Geleff S, Gnant M, Horvat R, Jakesz R, Birner P. VEGF-C expressing tumor-associated macrophages in lymph node positive breast cancer: impact on lymphangiogenesis and survival. Surgery 139: 839–846, 2006. [DOI] [PubMed] [Google Scholar]
- 138.Schutz FA, Choueiri TK, Sternberg CN. Pazopanib: clinical development of a potent anti-angiogenic drug. Crit Rev Oncol Hematol 77: 163–171, 2011. [DOI] [PubMed] [Google Scholar]
- 139.Serini G, Gabbiani G. Mechanisms of myofibroblast activity and phenotypic modulation. Exp Cell Res 250: 273–283, 1999. [DOI] [PubMed] [Google Scholar]
- 140.Shimotsuma M, Shields JW, Simpson-Morgan MW, Sakuyama A, Shirasu M, Hagiwara A, Takahashi T. Morpho-physiological function and role of omental milky spots as omentum-associated lymphoid tissue (OALT) in the peritoneal cavity. Lymphology 26: 90–101, 1993. [PubMed] [Google Scholar]
- 141.Shimotsuma M, Simpson-Morgan MW, Takahashi T, Hagiwara A. Activation of omental milky spots and milky spot macrophages by intraperitoneal administration of a streptococcal preparation, OK-432. Cancer Res 52: 5400–5402, 1992. [PubMed] [Google Scholar]
- 142.Shimotsuma M, Takahashi T, Kawata M, Dux K. Cellular subsets of the milky spots in the human greater omentum. Cell Tissue Res 264: 599–601, 1991. [DOI] [PubMed] [Google Scholar]
- 143.Siegel RL, Miller KD, Jemal A. Cancer statistics 2015. CA Cancer J Clin 65: 5–29, 2015. [DOI] [PubMed] [Google Scholar]
- 144.Smith LH, Morris CR, Yasmeen S, Parikh-Patel A, Cress RD, Romano PS. Ovarian cancer: can we make the clinical diagnosis earlier? Cancer 104: 1398–1407, 2005. [DOI] [PubMed] [Google Scholar]
- 145.Stadlmann S, Amberger A, Pollheimer J, Gastl G, Offner FA, Margreiter R, Zeimet AG. Ovarian carcinoma cells and IL-1β-activated human peritoneal mesothelial cells are possible sources of vascular endothelial growth factor in inflammatory and malignant peritoneal effusions. Gynecol Oncol 97: 784–789, 2005. [DOI] [PubMed] [Google Scholar]
- 146.Stone PJ, Goodheart MJ, Rose SL, Smith BJ, DeYoung BR, Buller RE. The influence of microvessel density on ovarian carcinogenesis. Gynecol Oncol 90: 566–571, 2003. [DOI] [PubMed] [Google Scholar]
- 147.Strobel T, Swanson L, Cannistra SA. In vivo inhibition of CD44 limits intra-abdominal spread of a human ovarian cancer xenograft in nude mice: a novel role for CD44 in the process of peritoneal implantation. Cancer Res 57: 1228–1232, 1997. [PubMed] [Google Scholar]
- 148.Sugimoto H, Mundel TM, Kieran MW, Kalluri R. Identification of fibroblast heterogeneity in the tumor microenvironment. Cancer Biol Ther 5: 1640–1646, 2006. [DOI] [PubMed] [Google Scholar]
- 149.Sundfeldt K. Cell-cell adhesion in the normal ovary and ovarian tumors of epithelial origin; an exception to the rule. Mol Cell Endocrinol 202: 89–96, 2003. [DOI] [PubMed] [Google Scholar]
- 150.Sundfeldt K, Piontkewitz Y, Ivarsson K, Nilsson O, Hellberg P, Brannstrom M, Janson PO, Enerback S, Hedin L. E-cadherin expression in human epithelial ovarian cancer and normal ovary. Int J Cancer 74: 275–280, 1997. [DOI] [PubMed] [Google Scholar]
- 151.Swartz MA, Iida N, Roberts EW, Sangaletti S, Wong MH, Yull FE, Coussens LM, DeClerck YA. Tumor microenvironment complexity: emerging roles in cancer therapy. Cancer Res 72: 2473–2480, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Tan DS, Agarwal R, Kaye SB. Mechanisms of transcoelomic metastasis in ovarian cancer. Lancet Oncol 7: 925–934, 2006. [DOI] [PubMed] [Google Scholar]
- 153.Tarin D, Price JE, Kettlewell MG, Souter RG, Vass AC, Crossley B. Mechanisms of human tumor metastasis studied in patients with peritoneovenous shunts. Cancer Res 44: 3584–3592, 1984. [PubMed] [Google Scholar]
- 154.Tebbe C, Chhina J, Dar SA, Sarigiannis K, Giri S, Munkarah AR, Rattan R. Metformin limits the adipocyte tumor-promoting effect on ovarian cancer. Oncotarget 5: 4746–4764, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Tlsty TD, Coussens LM. Tumor stroma and regulation of cancer development. Annu Rev Pathol 1: 119–150, 2006. [DOI] [PubMed] [Google Scholar]
- 156.Tokuda Y, Satoh Y, Fujiyama C, Toda S, Sugihara H, Masaki Z. Prostate cancer cell growth is modulated by adipocyte-cancer cell interaction. BJU Int 91: 716–720, 2003. [DOI] [PubMed] [Google Scholar]
- 157.Tsujimoto H, Takhashi T, Hagiwara A, Shimotsuma M, Sakakura C, Osaki K, Sasaki S, Shirasu M, Sakakibara T, Ohyama T, et al. Site-specific implantation in the milky spots of malignant cells in peritoneal dissemination: immunohistochemical observation in mice inoculated intraperitoneally with bromodeoxyuridine-labelled cells. Br J Cancer 71: 468–472, 1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Wang X, Deavers M, Patenia R, Bassett RL Jr, Mueller P, Ma Q, Wang E, Freedman RS. Monocyte/macrophage and T-cell infiltrates in peritoneum of patients with ovarian cancer or benign pelvic disease. J Transl Med 4: 30, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Wedge SR, Kendrew J, Hennequin LF, Valentine PJ, Barry ST, Brave SR, Smith NR, James NH, Dukes M, Curwen JO, Chester R, Jackson JA, Boffey SJ, Kilburn LL, Barnett S, Richmond GH, Wadsworth PF, Walker M, Bigley AL, Taylor ST, Cooper L, Beck S, Jurgensmeier JM, Ogilvie DJ. AZD2171: a highly potent, orally bioavailable, vascular endothelial growth factor receptor-2 tyrosine kinase inhibitor for the treatment of cancer. Cancer Res 65: 4389–4400, 2005. [DOI] [PubMed] [Google Scholar]
- 160.Wei W, Mok SC, Oliva E, Kim SH, Mohapatra G, Birrer MJ. FGF18 as a prognostic and therapeutic biomarker in ovarian cancer. J Clin Invest 123: 4435–4448, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Wijffels JF, Hendrickx RJ, Steenbergen JJ, Eestermans IL, Beelen RH. Milky spots in the mouse omentum may play an important role in the origin of peritoneal macrophages. Res Immunol 143: 401–409, 1992. [DOI] [PubMed] [Google Scholar]
- 162.Witz CA, Montoya-Rodriguez IA, Cho S, Centonze VE, Bonewald LF, Schenken RS. Composition of the extracellular matrix of the peritoneum. J Soc Gynecol Investig 8: 299–304, 2001. [DOI] [PubMed] [Google Scholar]
- 163.Xu L, Yoneda J, Herrera C, Wood J, Killion JJ, Fidler IJ. Inhibition of malignant ascites and growth of human ovarian carcinoma by oral administration of a potent inhibitor of the vascular endothelial growth factor receptor tyrosine kinases. Int J Oncol 16: 445–454, 2000. [DOI] [PubMed] [Google Scholar]
- 164.Yamada SD, Baldwin RL, Karlan BY. Ovarian carcinoma cell cultures are resistant to TGF-β1-mediated growth inhibition despite expression of functional receptors. Gynecol Oncol 75: 72–77, 1999. [DOI] [PubMed] [Google Scholar]
- 165.Yamaguchi H, Pixley F, Condeelis J. Invadopodia and podosomes in tumor invasion. Eur J Cell Biol 85: 213–218, 2006. [DOI] [PubMed] [Google Scholar]
- 166.Yamazaki D, Kurisu S, Takenawa T. Regulation of cancer cell motility through actin reorganization. Cancer Sci 96: 379–386, 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Yeung TL, Leung CS, Mok SC. CAF reprogramming inhibits ovarian cancer progression. Cell Cycle 13: 3783–3784, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Yeung TL, Leung CS, Wong KK, Samimi G, Thompson MS, Liu J, Zaid TM, Ghosh S, Birrer MJ, Mok SC. TGF-β modulates ovarian cancer invasion by upregulating CAF-derived versican in the tumor microenvironment. Cancer Res 73: 5016–5028, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Zaid TM, Yeung TL, Thompson MS, Leung CS, Harding T, Co NN, Schmandt RS, Kwan SY, Rodriguez-Aguay C, Lopez-Berestein G, Sood AK, Wong KK, Birrer MJ, Mok SC. Identification of FGFR4 as a potential therapeutic target for advanced-stage, high-grade serous ovarian cancer. Clin Cancer Res 19: 809–820, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Zhang L, Yang N, Garcia JR, Mohamed A, Benencia F, Rubin SC, Allman D, Coukos G. Generation of a syngeneic mouse model to study the effects of vascular endothelial growth factor in ovarian carcinoma. Am J Pathol 161: 2295–2309, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]


