Abstract
Malignant ascites presents a considerable clinical challenge to the management of ovarian cancer, but also provides a wealth of opportunities for translational research. The accessibility of ascitic fluid and its cellular components make it an excellent source of tumour tissue for the investigation of prognostic and predictive biomarkers, pharmacodynamic markers and for molecular profiling analysis. In this Opinion article, we discuss recent advances in our understanding of its pathophysiology, the development of new methods to characterize its molecular features and how these findings can be used to improve the treatment of malignant ascites, particularly in the context of ovarian cancer.
Ascites is the excess accumulation of fluid in the peritoneal cavity. Derived from the Greek word askites, meaning bag-like, it is caused by many pathologies; the most common is hepatic cirrhosis, which accounts for 81% of cases1. Others causes include heart failure (3%) and tuberculosis (2%), and a substantial minority (10%) is associated with malignancy (TABLE 1). The composition of ascitic fluid is dependent on the disease with which it is associated (BOX 1).
Table 1. Malignant ascites: clinical features and investigations.
| Investigations and indications |
Signs (% of patients)2,3 |
|---|---|
| Clinical examination | Dullness to percussion, a shifting dullness and fluid wave. This requires >1,500 mls of ascites to be present for detection. This means of diagnosis has a 50% to 94% sensitivity154 |
| Patient symptoms | Anorexia (36%), nausea (37%), abdominal swelling (55%), abdominal pain (53%), early satiety (6%), weight change (5%), dyspnoea (11%), vomiting (25%) and fatigue (17%) |
| Imaging | Ultrasound is used to assess and to mark the area most suitable for drainage. Using this method 100 mls of ascitic fluid can be detected155, whereas >20 mls can be detected using computer-aided tomography or magnetic resonance imaging. Disease status can also be assessed. It is not possible to distinguish between malignant and benign ascites on the basis of examination or radiological imaging alone |
| Cytological characteristics |
This is a standard technique for diagnosing malignant ascites. It is essential for differentiating between malignant, infective and inflammatory causes of ascites (97% sensitivity). Cellular content of ascitic fluid represents shedding of tumour cells from the tumour into the peritoneal fluid. The yield of cytology is greater with primary peritoneal tumours156 |
| Immunohistochemical staining |
Immunohistochemical staining of the ascites is important in determining the primary site of the tumour. CA125 is a glycoprotein secreted by and overexpressed on the cell surface of ovarian cancer cells. Other markers include oestrogen receptor, cytokeratin 7, Wilm’s tumor suppressor gene, cytokeratin 20, progesterone receptor, calretinin, carcinoembryonic antigen, gross cystic disease fluid protein, and thyroid transcription factor 1 (REF. 157) |
Box 1. The composition of ascites.
Ascites fluid contains variable proportions of suspended cells and debris depending on the pathogenesis.
Fluid composition
The protein content of ascites can be useful in determining the cause of ascites. Ascites that is secondary to portal hypertension (most commonly secondary to liver failure) can be readily differentiated from ascites that is due to other causes by the serum–ascites albumin gradient (SAAG = [albumin] serum – [albumin] ascites). If the SAAG is greater than or equal to 1.1 g per dl (or 11 g per L), ascites is ascribed to portal hypertension with an approximate 97% accuracy151. A lower SAAG strongly suggests an alternative diagnosis, such as malignant ascites or tuberculosis. Total glucose and lactic dehydrogenase levels also vary, typically measuring <3.33 mmol per L and >200 international units (IU) per L, respectively, in ascites secondary to malignancy and tuberculosis. With pancreatic ascites, the levels of ascitic fluid amylase may exceed 2,000 IU per L.
Cellular composition
In non-malignant ascites, the cell count and white cell differential is useful in determining an infective or inflammatory cause. With spontaneous bacterial peritonitis, the upper limit of the neutrophil count of 250 cells per μL is exceeded, and represents >50% of the total white cell count in the ascitic fluid. In malignant ascites, the cellular population consists of differing proportions of tumour cells, mesothelial cells, fibroblasts, macrophages, white blood cells and red blood cells152. Analysis of ascites samples from patients with ovarian cancer has shown considerable variation in the cell populations66, with a higher proportion of red blood cells characteristically seen when ascites has rapidly developed. A typical distribution of the cellular components comprising the ascitic fluid of women with ovarian cancer is 37% lymphocytes, 29% mesothelial cells, 32% macrophages and <0.1% adenocarcinoma cells152.
The most common primary site of cancer that is associated with ascites is ovarian, accounting for 38% of malignant ascites occurring in females2. Patients with other abdominal epithelial malignancies, including pancreatobiliary and gastric cancer, develop ascites in 21% and 18.3% of cases, respectively3. Malignant ascites can also develop secondary to extra-abdominal tumours, such as breast and lung cancer, as well as lymphoma.
In many forms of malignancy, ascites is a sign of advanced disease and poor prognosis3,4, with only 11% of patients surviving longer than 6 months2. Epithelial ovarian cancer (EOC) is the exception. The majority of women with ovarian cancer present with advanced disease (stage III or stage IV) that may include ascites. In these patients, treatment with surgery together with combination chemotherapy has resulted in a median progression-free survival of 16–22 months and a 5-year survival rate of 27%5, although better results are now being reported with improvements in therapy. One study has suggested that ascites at presentation is an independent prognostic factor for time to relapse6 and, overall, more than one-third of women with ovarian cancer develop ascites during the course of their disease3, and this is not limited to any specific histological subtype. Although some oncologists consider that the incidence of ascites may be reducing with the widespread use of combination chemotherapy that incorporates taxanes, there are no published supporting data.
Ascites can cause debilitating symptoms as a substantial volume of fluid can cause pain, early satiety and respiratory compromise7. The gastrointestinal and urinary systems can also be affected, resulting in significant morbidity (TABLE 1).
In patients with ovarian cancer, ascites is essentially treated through the treatment of the underlying disease; that is, using platinum-based intravenous chemotherapy. However, once chemoresistant disease has developed, intractable ascites can be a major problem and the majority of patients are subjected to frequent paracentesis to temporarily alleviate symptoms. Newer strategies to control ascites, including anti-vascular endothelial growth factor (VEGF) therapy, are proving promising, but there is an urgent need to further understand the pathophysiology of ascites so that more effective therapy can be developed. In this Opinion article, we discuss recent findings on the biology of ascites. Ascitic fluid provides a rich source of tumour cells that can be used to study various biological aspects of the underlying tumour, including drug resistance and mechanisms of tumour progression. Consequently, these studies have the potential to enhance our insight into effective therapeutic strategies to prevent the formation of ascites and to overcome platinum resistance in ovarian cancer.
Pathophysiology of ascites
The transition of epithelial cells to a more mesenchymal phenotype (epithelial to mesenchymal transition (EMT)) is associated with motility and, in cancer biology, with invasive growth8. Epithelial ovarian cells convert to a mesenchymal phenotype as part of a normal physiological process, which increases the motility and proliferation of these cells to allow repair of the surface epithelium when an oocyte is released9. Therefore, it is not surprising that EOC is frequently associated with the early invasion of the surrounding peritoneum. Adhesion to the mesothelin and hyaluronic acid expressed by mesothelial cells, which line the peritoneal cavity, is facilitated by CD44 (REFS 10,11), β-integrins12 and CA125 (REFS 13,14) expressed on the ovarian cancer cell surface. Once attached to the peritoneal surface, cells proliferate and invade the mesothelium (the outer layer of the peritoneal membrane). It has been suggested that transcoelomic metastasis (the seeding of tumour cells onto the surface of the peritoneum) is directly associated with ascites production. During embryogenesis, surface epithelial cells of the ovary originate from the coelomic mesothelium9, which might partly explain why ovarian cancer disseminates through the transcoelomic route. Reduction in peritoneal tumour bulk as a result of surgery and chemotherapy is mostly associated with a reduction in ascites; supporting the concept that transcoelomic metastases are involved in ascites production.
Anatomy of the peritoneal membrane
The peritoneal membrane consists of five layers and covers the visceral organs, as well as the abdominal and pelvic cavities (FIG. 1). The first layer of the peritoneal membrane comprises endothelial cells that line the intravascular space of capillaries. These cells have an extracellular glycocalyx and fixed anionic charge that makes it difficult for large blood plasma proteins, such as albumin, to pass through. Tight junctions link endothelial cells together, and so intracellular pores provide transport through the layer15. Separating the endothelial cells from the interstitial space (the third layer) is the endothelial basement membrane. Proteoglycans anchored in the basement membrane also exert a negative charge, which constitutes a selective barrier for albumin. Loose connective tissue in the interstitial space, comprised of fibroblasts, collagen and hyaluronic acid, blocks the diffusion of macromolecules before the submesothelial basement membrane. The final layer consists of mesothelial cells. Bound by tight junctions and secreting surface glycosaminoglycans (hyaluronan)16 into the abdominal space, mesothelial cells provide an effective anti-adhesive surface and a protective barrier against physical damage. In response to injury or insult mesothelial cells release chemokines, such as CCL2 (also known as MCP1) and interleukin-8 (IL-8; also known as CXCL8) and they also increase the expression of intracellular adhesion molecule 1 (ICAM1) and vascular cell adhesion protein 1 (VCAM1), to which leukocytes attach17.
Figure 1. The peritoneum, peritoneal membrane and ascites.
a ∣ The peritoneal membrane covers the visceral organs, as well as the abdominal and pelvic cavities, and consists of five layers. The first layer is made up of endothelial cells that line the intravascular space of capillaries. These cells have an extracellular glycocalyx and fixed anionic charge that makes it difficult for large blood plasma proteins, such as albumin, to pass through. Intracellular pores provide transport through this layer. The endothelial cell basement membrane provides the second layer. The interstitial space is the third layer and contains fibroblasts, collagen and hyaluronic acid, and it can block the diffusion of macromolecules before the submesothelial basement membrane (the fourth layer). The final layer consists of mesothelial cells. Bound by tight junctions and secreting surface glycosaminoglycans (hyaluronan) into the abdominal space, mesothelial cells provide an effective anti-adhesive surface and a protective barrier against physical damage. In physiological conditions, the difference in oncotic pressure across the peritoneal membrane (high at the endothelial layer and low at the mesothelial layer) limits capillary fluid filtration and prevents oedema that is due to water reabsorption into the capillaries from the interstitial space. b ∣ In patients with tumours in the abdominal cavity, the cross-sectional area of microvessels lining the peritoneal cavity is increased, and this results in an increased filtration surface for fluid. In addition, malignant ascites has a high protein concentration that is secondary to increased capillary permeability. Inflammatory cytokines and chemokines, as well as reduced lymphatic flow, all contribute to alterations of the peritoneal membrane. These changes decrease the plasma to peritoneal oncotic pressure difference, so the direction of flow of fluid is into the peritoneal cavity, obeying Starling’s hypothesis of capillary haemodynamics153. This leads to the build up of pathological volumes of fluid in the peritoneal cavity. c ∣ A computed tomography image of a patient with ovarian cancer showing the accumulation of ascites in the abdomen, leading to the compression of organs and tissues in the abdominal cavity. Image in part c courtesy of N. Tunariu, Royal Marsden Hospital, UK.
However, in malignancies, such as ovarian cancer, this balance of fluid production and reabsorption is disrupted and pathological volumes of fluid — that is, ascites — accumulate in the peritoneal cavity. The pathophysiological mechanisms for this have been attributed to impaired fluid drainage and/or to increased net filtration.
Impaired drainage from the peritoneal cavity
The peritoneal membrane is rich in lymphatic tissue. The lymphatic system circulates fluid, proteins and other macromolecules back into the vascular circulation. Lymphatic stomata, first described by von Recklinghause in 1863 (REF. 18), are triple-layered structures comprising mesothelium, a loose network of connective tissue and endothelium that permit the flow of fluid between the peritoneal surface and the submesothelial lymphatics. These lymphatic portals are particularly abundant on the omental and sub-diaphragmatic peritoneal surfaces19, and their obstruction by tumour cells has been suggested as a mechanism for ascites. This hypothesis was first proposed almost 50 years ago20. Experiments showed that intraperitoneal-injected Indian ink was not transported in the lymphatic system in mice with malignant ascites, suggesting obstruction. More substantial evidence was published in 1972 (REF. 21) when intraperitoneal-injected radioactive erythrocytes were not found in the peripheral bloodstream within 5 hours in mice that were inoculated with ovarian tumour cells that result in ascites. A similarly designed study using radioactive albumin also demonstrated reduced efflux from the peritoneum following tumour injection22.
Although the involvement of the lymphatic system in the pathogenesis of ascites is not disputed, it is unlikely to be a major contributing factor as there are indirect effects of malignancy on the anatomy and physiology of the peritoneal membrane. In mouse models23, ascites formation can occur in the absence of a tumour obstructing the draining lymphatics of the peritoneal cavity, and high concentrations of protein are evident in malignant ascites, indicative of alterations in peritoneal membrane permeability. In addition, in the clinic, ascites formation can be seen in the absence of tumour bulk, suggesting a prominent role for non-obstructive mechanisms, including those which involve cytokines and chemokines that are secreted by the tumour cells and the associated cells that constitute the tumour microenvironment.
Increased filtration into the peritoneal cavity
In physiological conditions, the difference in oncotic pressure (a form of osmotic pressure exerted by proteins, such as albumin, in blood plasma that increases water uptake) across the peritoneal membrane (high at the endothelial layer and low at the mesothelial layer) limits capillary fluid filtration and prevents oedema owing to water reabsorption into the capillaries from the interstitial space (FIG. 1). In patients with tumours in the abdominal cavity, the cross-sectional area of microvessels that line the peritoneal cavity is increased22 (discussed below), and this results in an increased filtration surface for fluid. In addition, malignant ascites has a high protein concentration that is secondary to increased capillary permeability. This decreases the plasma to peritoneal oncotic pressure difference, so that the direction of fluid flow is into the peritoneal cavity, obeying Starling’s hypothesis of capillary haemodynamics. As only a minor increase in portal vein pressure in patients with ovarian cancer with or without ascites has been observed, increased hydraulic pressure is not thought to contribute to the pathogenesis of ascites in ovarian cancer24.
Ascitic fluid that is generated as a result of malignancy profoundly affects microvascular permeability25, and in 1986 it was shown to cause the leak of proteins from a healthy omentum4. VEGF was subsequently identified as being instrumental in altering the permeability of the peritoneal membrane, and therefore in the pathogenesis of ascites. VEGF is a glycosylated mitogen that specifically acts on endothelial cells, inducing cell proliferation, migration and inhibition of apoptosis. Moreover, VEGF induces angiogenesis and the permeabilization of blood vessels26.
VEGF is crucial for normal ovarian function27 and overexpression of VEGF in patients with EOC has been shown to correlate with the development of malignant ascites28,29 and a poorer prognosis30,31. VEGF selectively accumulates in ascites secondary to EOC at much higher levels than in ascites resulting from non-malignant causes32, and it is predominantly associated with the advanced stages of ovarian cancer32 and higher grade disease33. Preclinical experiments have demonstrated that overexpression of VEGF can induce ascites production from normal ovarian epithelium34 and enforced expression of VEGF by ovarian carcinoma cells has been shown to dramatically reduce the time to onset of ascites formation35. In animal models, VEGF inhibition has been shown to suppress the formation and re-accumulation of ascites in mice with intraperitoneal ovarian tumours36,37.
One mechanism by which VEGF increases peritoneal permeability is through the downregulation of the tight junction protein claudin 5 in the peritoneal endothelium38,39. Claudin 5 is a transmembrane protein in the tight junction that, together with occludin40, mediates adhesion by promoting homophillic interactions along the cell border41,42. Studies in transgenic mice have shown that the absence of tight junction proteins affects the control of vascular permeability to fluids43. Angiogenesis is known to be associated with increased vessel permeability44, and a further mechanism contributing to this is tyrosine phosphorylation of cadherin–catenin complexes, which is mediated by VEGF45. The resulting complexes cause decreased junctional strength and increased permeability41.
There are several factors that are thought to influence VEGF production by ovarian cancer cells, including hypoxia46, lysophosphatidic acid (LPA)47, tumour necrosis factor (TNF)48, endothelin 1 (REF. 49), cyclooxygenase 1 (REF. 50), IL-1β51, matrix metalloprotein ases (MMPs)52, insulin-like growth factor 1 (REF. 53), epidermal growth factor (EGF)54, platelet-derived growth factor55 and transforming growth factor-β (TGFβ)56. Blockade of TGFβ inhibits ascites production through the inhibition of VEGF expression in orthotopic human ovarian carcinoma models56. Interestingly, peritoneal drainage of ascites was also shown to increase secondary to the normalization of lymphatic vessel morphology and a reduction in lymphangiogenesis.
LPA is a biologically active phospholipid that is present in high levels in ascites and in serum from patients with ovarian cancer57,58, and a LPA-induced gene expression signature has been shown to correlate with a worse prognosis59. Produced extracellularly by the phosphodiesterase autotaxin (ATX)60, LPA signals through G protein-dependent cell-surface receptors, resulting in the production of unsaturated fatty acyl chains. These G protein-dependent receptors are upregulated during the malignant transformation of ovarian surface epithelial cells61. The effects of LPA are diverse and include increased mRNA levels and transcriptional regulation58 of VEGFA, urokinase plasminogen activator (UPA), IL-6 and IL-8. This results in increased endothelial permeability and in the inhibition of gap junctional communication between adjacent cells62 and, therefore, in ascites production.
Additional properties of ascites
There is evidence to suggest that EOC cells, including those in ascitic fluid, might have evolved to evade immunological surveillance7. Ascites-derived epithelial ovarian cancer cells have been shown to constitutively release microvesicles that contain CD95 ligand (CD95L; also known as FAS ligand) rather than expressing CD95L on their cell surface. CD95L expression or secretion can result in the apoptosis of immune cells that express CD95 (REF. 63). High concentrations of complement C1 complex inhibitor and the alternative complement pathway inhibitors factor H and factor H-like protein 1 have been noted on isolated metastatic EOC cells in ascitic fluid, suggesting a mechanism for the evasion of complement-induced immune destruction64. Furthermore, regulatory T cells that suppress tumour-specific T cell immunity promote tumour growth in vivo, and a correlation between the presence of regulatory T cells in ascitic fluid and reduced survival has also been found7. In addition, a ganglio-side, GD3, has recently been identified in malignant ascites as an inhibitory factor that prevents the innate immune activation of natural killer T (NKT) cells65.
Despite these immune suppressive effects, ascites is rich in lymphocytes, cytokines and chemokines66. Cytokines, including IL-1β, IL-6, IL-8 and IL-10 (REFS 67–69), are present at concentrations that are two to three logs greater than serum concentrations69, indicating that ascites provides an environment that is conducive to cell growth. It is interesting that the expression of IL-6 is significantly higher in ascites secondary to ovarian cancer when compared with ascites resulting from other primary tumours70. Therefore, cytokine expression may generate a specific microenvironment that enables the characteristic pattern of metastatic spread in ovarian cancer. In animal models, the expression of IL-8 is associated with increased tumorigenicity and ascites formation71,72. IL-6 has been shown to promote growth70, chemoresistance, tumour invasion73 and angiogenesis74,75. High levels of IL-6 (REFS 69,76,77) and IL-10 (REF. 68) expression in ascites have been shown to be associated with shorter progression-free survival77, poor overall survival, poor initial response to chemotherapy69 and drug resistance. As well as the secretion of these cytokines by tumour and stromal cells that are present in the ascites microenvironment, Naldini et al.78 showed that healthy donor peripheral blood mononuclear cells cultured in the presence of ascitic fluid from patients with ovarian cancer significantly increased the release of cytokines and chemokines, such as IL-6 and IL8, which are known to support tumour progression, and inhibited the production of anti-inflammatory cytokines, such as interferon-γ and IL-12. In addition, MUC16 expressed on the surface of ovarian cancer cells has been shown to provide immune protection by inhibiting the interaction of ovarian cancer cells with natural killer cells79. Further characterization of cytokine profiles may provide a greater understanding of the role of ovarian cancer cells in the pathogenesis of ascites, as well as the role of ascites in ovarian cancer tumour progression.
Other chemokines also have a role in the pathophysiology of ascites. CXCR4, the receptor for CXCL12, seems to be selectively expressed on EOC cells80 and contributes to the proliferation81 and migration82 of EOC. The level of chemokine ligand CXCL4 is increased in ascites from patients with EOC and, together with CXCL6, CXCL8, CXCL14 and CCL13, CXCL4 has been shown to be upregulated in peritoneal specimens that lack detectable cancer cells from patients with ovarian cancer compared with patients with benign ovarian disease83, suggesting the upregulation of the chemokine network throughout the peritoneum. Proteinases, including MMPs, are crucial for invasive cancer growth and the pathogenesis of ascites. MMPs, mainly MMP9, have a role in the release of biologically active VEGF and consequently in the formation of ascites52 and have been shown to be an independent predictor of decreased survival84.
In vitro and in vivo research has suggested that the activation of the EGF receptor may partly induce a pro-inflammatory microenvironment76 through the upregulation of nuclear factor-κB (NF-κB). The NF-κB transcription factor is constitutively active in >50% of ovarian cancers and results in the upregulation of multiple genes that include IL6 and IL8. Inhibition of NF-κB signalling significantly inhibits the expression of VEGF and IL-8, resulting in decreased vascularization of lesions, decreased formation of malignant ascites and prolonged survival in mice71. Autocrine stimulation of the IL-6 receptor on ovarian cancer cells facilitates the activation of signal transducer and activator of transcription 3 (STAT3) through Janus kinase 2 (JAK2), resulting in the inhibition of apoptosis and the induction of angiogenesis85,86.
The activation of the PI3K pathway is deregulated in up to 70% of ovarian cancers61 through mechanisms that include amplification of PIK3CA and AKT or inactivating mutations of PTEN. Activation of AKT87,88 and ERK1 or ERK2–ELK1 signalling89 by survival factors in ascitic fluid has been shown to promote resistance to TNF-related apoptosis-inducing ligand (TRAIL)-induced apoptosis, highlighting the contribution of the tumour microenvironment to cell survival in ovarian cancer. AKT is also functionally required for bone morphogenetic protein (BMP) signalling in tumour spheroids and the adhesion and dispersion90 of epithelial ovarian cancer cells. The mechanisms underlining the interaction between AKT and BMP pathways, however, remain to be fully elucidated.
The treatment of ascites
Pharmacological
At presentation, patients with ovarian cancer and ascites are treated with surgery together with first-line combination chemotherapy of carboplatin and paclitaxel. There is an increasing trend for surgery to be deferred until at least three cycles of chemotherapy have been given, with the aim of reducing the problems associated with surgery, such as bowel resection. The clinical response to first-line chemotherapy exceeds 60%. However, the majority of patients relapse and the addition of a third cytotoxic drug, such as gemcitabine, topotecan, caelyx or epirubicin, to platinum and paclitaxel has yielded disappointing results91–93. The VEGF-binding antibody bevacizumab has been added in Phase III trials (Gynecologic Oncology Group 218 study (GOG218; clinical trial ID: NCT00262847) and ICON7 (NCT00483782)) to standard treatment during and up to 15 months after chemotherapy, with reports of improved progression-free survival94,95, as well as increased overall survival in high-risk subgroups (patients with stage IV disease or suboptimally debulked stage III disease)96. Randomized trials have indicated that bevacizumab, concurrent with and following chemotherapy, can also provide benefit in terms of delaying further recurrence in both platinum-sensitive (OCEANS trial (NCT00434642)97) and platinum-resistant (AURELIA trial (NCT00976911)98) disease. Although in these trials bevacizumab was not introduced specifically to treat ascites, its increasing utility should have an effect. However, resistance to anti-VEGF therapy will remain a key problem, and efforts to understand the underlying mechanisms and to develop further anti-angiogenic strategies are a high priority99.
For women with relapsed ovarian cancer, resistance to conventional chemotherapy is also a crucial factor and it develops in almost all cases. If this resistance is associated with ascites, pharmacological therapeutic options specific to the control of ascites are limited. Randomized controlled studies are few, with the majority of treatments having been studied in a small series; including the use of diuretics, octreotide, intraperitoneal therapy, immunological modulators, metalloproteinase inhibitors and anti-angiogenics.
Diuretics are frequently used in the treatment of ascites that has arisen as a result of non-malignant disease100. However, no randomized trials have assessed the efficacy of diuretics in malignant ascites101 and evidence for their use is weak102. Phase II data suggest that they are effective in approximately one-third of patients with malignancy103, and side effects of treatment include nausea, hypotension and electrolyte disturbance. Efficacy is thought to decline with tumour progression104 and, interestingly, patients with portal hypertension2 or massive hepatic metastases103 are most likely to respond to diuretics, which may account for the lack of efficacy in relation to ascites arising in patients with ovarian cancer. Octreotide, a safe, well-tolerated somatostatin analogue increases the glomerular filtration rate and the sodium and water excretion of patients with non-malignant ascites, mainly through the suppression of an activated renin–aldosterone axis105. In a pilot study106, monthly intramuscular injections of long-acting octreotide were shown to delay the need for repeat paracentesis from a mean of 14 days to 28 days in patients with malignant ascites. Although there may have been a degree of clinical benefit, this delay was not significant (P = 0.17).
The pharmacological model for intraperitoneal chemotherapy was published more than 30 years ago107. High concentrations of chemotherapy drugs in the peritoneum resulted in a prolonged exposure to these agents compared with intravenous administration. More recently, intraperitoneal chemotherapy has been explored for the first-line treatment of optimally debulked women with stage III ovarian cancer108. Despite a 16-month survival advantage, the complication rate, 34% of which was due to catheter-related problems, was significantly greater in the experimental group, and only 42% of women in the trial completed the recommended six cycles of intraperitoneal chemotherapy with cisplatin. Patients in the experimental group also suffered from more pain and fatigue, as well as higher rates of haematological, gastrointestinal and metabolic toxicities. The dose of cisplatin used in the experimental group (100 mg per m2) has not been widely used for these reasons.
Although the use of first-line intraperitoneal chemotherapy remains controversial, it has been the route of choice in many early phase trials in which patients with malignant ascites from any primary tumour were recruited to study chemotherapy in the control of ascites. Intraperitoneal administration of bleomycin109,110, mitoxantrone110,111 or fluorouracil in combination with cisplatin112 have all been reported to improve the control of ascites. However, there are no standardized criteria for assessment and further studies would therefore be required for full evaluation.
Evidence to suggest that an immunological approach to treat malignant ascites in EOC may be effective has previously been observed in small studies of intraperitoneal triamcinolone113, a long-acting synthetic corticosteroid, intraperitoneal interferon114 and TNF115. More recently, a prospective randomized Phase II/III trial was conducted using intraperitoneal catumaxomab (Removab; Fresenius Biotech)116. This is a trifunctional monoclonal antibody with two different antigen-binding sites (one that binds to tumour cells via the epithelial cell-adhesion molecule and another to T cells through CD3), as well as a functional Fc domain that activates Fcγ-receptor I-, Fcγ-receptor IIa- and Fcγ-receptor III-positive immune cells. Therefore, different immune effector cells are simultaneously recruited and activated, resulting in tumoricidal activity through different immunological killing mechanisms. The study showed that, at appropriate doses, treatment with catumaxomab led to significantly longer paracentesis-free survival and improved palliation following intraperitoneal administration in patients with malignant ascites compared with controls. These data led to the approval of catumaxomab for the treatment of malignant ascites by the European regulatory authorities in 2009; this was the first therapeutic agent to be specifically approved for the treatment of malignant ascites. Ott et al.117 demonstrated a strong correlation between humoral response to catumaxomab and clinical outcome by measuring human antimouse antibodies (HAMAs) 8 days after the fourth treatment, suggesting that HAMA development (with associated symptoms of cytokine release) may be a biomarker for catumaximab response. The effective harnessing of the immune system by catumaxomab suggests that this treatment may be beneficial to patients with earlier stage disease.
MMPs have a crucial role in tumour invasion and angiogenesis. Preclinical research in this area led to the development of batimatstat, an MMP inhibitor. Patients with ovarian cancer were included in a Phase I study of intraperitoneal batimatstat118 that found the main adverse effect to be nausea and vomiting. Interestingly, five of the 23 patients neither reaccumulated ascites nor died up to 112 days after dosing, and agents of this type may warrant further study.
"with the advances in our understanding of its pathophysiology … it is anticipated that more rational, targeted and effective treatment strategies for malignant ascites and ovarian cancer will emerge"
There is strong preclinical evidence for the dependence of ascites formation on abnormal tumour vascularity and permeability. Therefore, research on VEGF inhibitors has specifically studied their use to palliate the symptoms of ascites in heavily pretreated patients. Numnum et al.119 reported that repeated paracentesis could be discontinued because of dramatically reduced levels of ascites after the initiation of therapy with bevacizumab (15 mg per kg intravenously, every 3 weeks). Intraperitoneal administration of bevacizumab has also been explored. The largest series only included nine patients with refractory ascites, which resulted from colorectal, breast, uterine or ovarian cancer120. However, in all the patients, the ascites resolved without reaccumulation or repeat paracentesis over a median observation period of >2 months. In addition, no grade 2–5 adverse events were observed. Despite these results, and other positive case reports121,122, no randomized trials using bevacizumab specifically for the palliation of ascites have yet been reported.
Composed of entirely human sequences, aflibercept (also known as VEGF-Trap) binds to and neutralizes VEGFA, VEGFB and placental growth factor123. Preclinical models found that aflibercept prevented ascites accumulation and inhibited the growth of disseminated cancer35. A pilot study was carried out in 12 patients in whom time to repeat paracentesis was increased following treatment with aflibercept (4 mg per kg, every 2 weeks)124, and the effectiveness of VEGF blockade using aflibercept was confirmed in a Phase II randomized, double-blind trial conducted by Gotlieb et al.125 in patients with ovarian cancer. However, the authors acknowledge that the limitation of this treatment is the risk of significant morbidity associated with bowel perforation, which was higher in the treatment group. Therefore, the advantages of aflibercept over bevacizumab in this context are unclear.
Non-pharmacological
Paracentesis via percutaneous drainage remains the most regularly used intervention for providing immediate short-term palliation of symptoms. Complications of paracentesis include continuous leakage from the drainage site and, occasionally, bowel perforation. Procedures often require a hospital stay and may have to be frequently repeated. To reduce the need for multiple procedures, and so admissions, peritoneovenous shunts (PVSs) are used in the treatment of ascites126,127 to direct ascitic fluid through a one-way valve into the vena cava, palliating symptoms in up to 70% of patients102. Encouragingly, the primary patency of shunts averages 87 ± 57 days128. However, major complications that occur in 6% of patients include pulmonary oedema, pulmonary embolus and infection102.
Catheter drainage is an alternative form of management. Catheters, made from flexible silicone with a polyester cuff, are tunnelled subcutaneously towards the peritoneal cavity129 and are easy to self-drain, thus enhancing patient autonomy while negating the need for repeated paracentesis. A review assessing the safety and efficacy of indwelling intraperitoneal catheters (Tenckhoff and PleurX) for the management of refractory malignant ascites reported a safe and effective palliative strategy despite an 11% infection rate130. A recent study131 reported a 100% technical success rate for the insertion of a PleurX drain with an associated low complication rate, thus advocating their use as a first-line approach in patients with refractory malignant ascites. On the basis of this evidence, the UK National Institute for Clinical Excellence have recommended that the PleurX peritoneal catheter drainage system should be considered for use in patients with treatment-resistant, recurrent malignant ascites132.
Bridging the translational gap
As is evident from the discussions above, ascites is accessible and often therapeutically removed from patients; it is therefore a valuable source of tumour material, and consequently of information on the molecular perturbations of the tumour. The ability to isolate and analyse malignant ascites and its cellular components may thus facilitate the identification of therapeutic targets, as well as prognostic and predictive biomarkers. In addition, evidence to support clonal evolution, as well as molecular heterogeneity, throughout a patient’s progress during treatment may be obtained. Various high-throughput techniques are now available to characterize the cellular and acellular components of ascites at the genetic, transcriptomic and proteomic levels. These include microarray-based gene expression profiling133, proteomic methods134 and next-generation sequencing technologies135.
Translational research using cellular components in ascites
Do cancer cells in ascites harbour molecular changes that render them phenotypically distinct from those retained in the primary tumour? Indeed, it would seem that a degree of clonal evolution is necessary for the transcoelomic metastatic process and that a dynamic regulation of cell adhesion molecules, such as E-cadherin, throughout the process may also be required7. Nonetheless, the frequent presence of ascites at first presentation, and subsequent relapse in EOC, provides an accessible pool of tumour cells that, in most current studies, are assumed to mirror some or most of the molecular characteristics of cells in the primary tumour or metastatic deposits. In order to study the cellular component of ascites, malignant cells from ascites can be preserved in cell blocks for immunohistochemical testing136,137 or can be frozen for further molecular analysis137. Molecular and functional analysis of malignant ascitic cells has been greatly facilitated by the ability to isolate and culture cells derived from the ascitic fluid of patients with advanced EOC138,139, and this presents further opportunities for the identification and validation of therapeutic targets and predictive biomarkers.
The potential utility of this approach was demonstrated by Mukhopadhyay et al.140 where a functional assay for cells deficient in homologous recombination (HR)-mediated DNA repair was developed using primary cell cultures that were derived from malignant ascitic cells. This was used to identify patients who were most likely to benefit from poly(ADP-ribose) polymerase (PARP) inhibitors, a novel class of targeted therapy effective in tumour cells that are deficient in HR repair141. In another study by Elattar et al.142, primary ovarian cancer cultures were established from the ascitic fluid of 11 patients to assess the expression of androgen receptor (AR) mRNA and response to androgenic stimulation. The study revealed a strong correlation between increased nuclear AR expression by immunohistochemistry and increased S phase fraction changes in primary cultures, suggesting that intervention with anti-androgenic agents may be effective at an earlier stage of the disease142.
In an effort to elucidate the role of PI3K pathway activation in drug-resistant ovarian cancer, Carden et al.143 immuno-magnetically separated ovarian cancer cells from ascites before quantifying the levels of phosphorylated AKT, p70S6K (also known as RPS6Kβ1) and glycogen synthase kinase 3β (GSK3β) by enzyme-linked immunosorbent assay (ELISA) and sequencing PIK3CA and AKT2 by PCR-amplified mass spectroscopy detection methods. In addition, PIK3CA and AKT2 amplifications and PTEN deletions were analysed by fluorescence in situ hybridization (FISH). The study observed that mutations of PIK3CA and amplification of PIK3CA and AKT2 or deletion of PTEN did not correlate with levels of phosphorylation of AKT, p70S6K and GSK3β. However, significantly higher levels of phosphorylated p70S6K levels were present in ascitic tumour cells from patients who did not respond to subsequent chemotherapy compared with samples of patients who did respond (P = 0.01). This suggested that other as yet unidentified factors that are independent of PI3K and that influence p70S6K activation may be important for clinical drug resistance.
Ascitic cells from patients with EOC have also been used to study subpopulations of ovarian tumour cells with stem cell-like properties — the side population of cells that take up low levels of Hoechst 33342 dye in flow cytometry analyses139,144. Side-population cells are characterized by prolonged anchorage-independent growth as spheroids, tumorigenicity when injected at low cell numbers into mice, inherent drug resistance leading to tumour recurrence following chemotherapy and ABC transporter-mediated efflux of the Hoechst 33342 dye144,145. Side-population cells that are capable of forming tumours in xenogeneic mice have been isolated from ascites of patients with ovarian cancer139, and a higher proportion of side-population cells has also been observed in ascites from patients who have relapsed following chemotherapy compared with chemo-naive patients144. Further characterization of these cells has suggested that the histone methyltransferase EZH2, a key component of the Polycomb-repressive complex 2, may be crucial for maintaining the stem cell state, as small interfering RNA-mediated knockdown of EZH2 results in the loss of side-population cells in ovarian tumour models, reduced anchorage-independent growth and reduced tumour growth in vivo144. Recent evidence suggests that interferon-α (IFNα) may have specific antitumour effects in side-population cells145, as might Muellerian-inhibiting factor146. The presence of these cells provides circumstantial evidence of the molecular heterogeneity in the population of malignant ascitic cells, and their clinical relevance is supported by a recent study that suggests that tumour cells isolated from ascites of chemoresistant patients had mRNA profiles that were characteristic of cancer stem cells147. However, it remains unclear how much of this is reflected in the population of malignant cells in the primary tumour.
Translational research using ascitic fluid
As discussed above, ascitic fluid harbours factors such as VEGF, epidermal growth factor and LPA7, which provide a supportive microenvironment for malignant cells to overcome anoikis and to survive in the hypoxic, but otherwise rich, liquid milieu7. Identification of additional crucial signals that promote and support cellular survival in ascites should permit the identification of new treatment strategies to control ascites. Methods used to elucidate the molecular characteristics of malignant ascitic fluid have included proteomic studies134 using mass spectroscopy (MS)-based techniques such as liquid chromatography (LC), surface enhanced laser desorption and ionization (SELDI), and matrix-assisted laser desorption and ionization (MALDI). More recently, high-throughput automated array-based proteomics techniques have been used such as reverse phase protein arrays (RPPAs) in which only very small amounts of tissue lysates are deposited onto slides covered with nitrocellulose, and proteins of interest are subsequently detected using specific antibodies148.
In a study using RPPA analysis by Davidson et al.148, malignant effusions (both ascitic and pleural effusions) from patients with EOC were found to have significantly higher expression levels of AKT, cAMP-responsive element binding protein (CREB) and JUN N-terminal kinase (JNK) than benign effusions (following RPPA analysis). Furthermore, high levels of p38, an increase in the ratio of phosphorylated EGFR and increased levels of phosphorylated JNK were associated with worse outcome in these patients148. Given that upregulation of the AKT–PI3K pro-survival and invasion pathways has been demonstrated in ovarian cancer149, it would seem that the proteomic profile of the ascitic milieu may reflect the protein expression profile of the underlying tumour. The results of this small study are an indication of the potential use of this technique for the discovery of diagnostic and prognostic biomarkers, but it will nonetheless require further validation in independent cohorts. Analysis of serial samples, such as pre- and post-chemotherapy levels, from each patient could also be compared to assess other molecular changes that may be predictive of treatment response and, in the case of clinical trials with targeted agents, indicative of target validation and inhibition134,148,150. It might be anticipated that ascitic fluid would have a generally stimulatory effect on the invasion and growth of EOC cells, but a study by Puiffe et al.133 demonstrated that the protein content of ascitic fluid can be either stimulatory or inhibitory to EOC cells in vitro, suggesting that both positive and negative regulators of tumour progression may be present in ascitic fluid. A wide variability of cytokine expression profile between ascites from different patients was recently demonstrated by Matte et al.68 using a multiplex cytokine array technique that simultaneously measured the level of 120 cytokines in ascites from patients with EOC, further illustrating the intrapatient heterogeneity of ascitic fluid content. Among the cytokines assayed, higher levels of osteoprotegerin (OPG), IL-10 and leptin in ascitic fluid were associated with worse outcome, and IL-10 was also found to mediate the survival-promoting activity of ascites against TRAIL-induced apoptosis68.
Conclusions
Malignant ascites presents a substantial clinical challenge, but also provides a wealth of potential opportunities for translational research in ovarian cancer. The accessibility of ascites translates into a readily available source of tumour tissue during the course of a patient’s treatment. In addition, it provides the opportunity for the identification of prognostic and predictive biomarkers, pharmacodynamic information, as well as options for molecular profiling and systems-biology-based analyses. In the current zeitgeist of personalized cancer medicine, the emerging picture from studies on malignant ascites seems to suggest that its molecular profile is subject to considerable inter-patient variability and that the ability to provide accurate molecular characterization will be crucial in designing the most appropriate intervention for patients in the era of targeted therapy.
Undeniably, although the best way to treat malignant ascites is to eradicate the underlying malignancy, the limits of our current therapeutic armamentarium are such that, in many cases of advanced ovarian cancer, tumours will inevitably become resistant to treatment, with the focus of care at that point being shifted to palliation of symptoms. In this context, given its significant associated morbidity, the ability to successfully manage the problem of recurrent ascites associated with treatment-resistant cancer through pharmacological or non-pharmacological methods with minimal risk, toxicity and discomfort, has the potential to vastly improve the level of palliative benefit and quality of life for patients. Therefore, it will be important to also include methods to assess ascites formation, such as time to paracentesis, as an end point in future clinical trials.
Inevitably, what we now know about malignant ascites has only served to highlight the considerable gap in our knowledge that still remains. Nonetheless, with the advances in our understanding of its pathophysiology, coupled with the development of new methods to delineate the molecular features of its various cellular and acellular components, it is anticipated that more rational, targeted and effective treatment strategies for malignant ascites and ovarian cancer will emerge in the future.
Acknowledgements
The Drug Development Unit, the Institute of Cancer Research and the Royal Marsden Hospital NHS Foundation Trust acknowledge support from Cancer Research UK, through core support and an Experimental Cancer Medicine Centre grant and from the National Institute of Health Research, as a Biomedical Research Centre. E.K. acknowledges support from the Wellcome Trust as a clinical research fellow.
Glossary
- Omentum
A fold of peritoneum connecting or supporting abdominal structures, such as the stomach and liver.
- Paracentesis
The procedure to remove fluid that has accumulated in the abdominal cavity (peritoneal fluid) by inserting a wide-bore needle through the abdominal wall.
- Satiety
A condition of being full beyond capacity.
- Starling’s hypothesis of capillary haemodynamics
The direction and rate of fluid transfer between blood plasma in the capillary and fluid in the tissue spaces depend on the hydrostatic pressure on each side of the capillary wall, on the osmotic pressure of protein in plasma and in tissue fluid, and on the properties of the capillary walls as a filtering membrane.
Footnotes
Competing interests statement
The authors declare no competing financial interests.
Contributor Information
Emma Kipps, The Institute of Cancer Research/Royal Marsden Hospital, Medicine, Downs Road, Sutton SM2 5PT, UK..
David S. P. Tan, The Drug Development Program, Princess Margaret Hospital, 610 University Avenue, Toronto M5G 2M9, Canada.
Stan B. Kaye, The Drug Development Unit, Sycamore House, Royal Marsden NHS Foundation Trust, Downs Road, Sutton SM2 5PT, UK.
References
- 1.Runyon BA. Care of patients with ascites. N. Engl. J. Med. 1994;330:337–342. doi: 10.1056/NEJM199402033300508. [DOI] [PubMed] [Google Scholar]
- 2.Parsons SL, Lang MW, Steele RJ. Malignant ascites: a 2-year review from a teaching hospital. Eur. J. Surg. Oncol. 1996;22:237–239. doi: 10.1016/s0748-7983(96)80009-6. [DOI] [PubMed] [Google Scholar]
- 3.Ayantunde A, Parsons S. Pattern and prognostic factors in patients with malignant ascites: a retrospective study. Ann. Oncol. 2007;18:945–949. doi: 10.1093/annonc/mdl499. [DOI] [PubMed] [Google Scholar]
- 4.Garrison RN, Kaelin LD, Galloway RH, Heuser LS. Malignant ascites. Clinical and experimental observations. Ann. Surg. 1986;203:644–651. doi: 10.1097/00000658-198606000-00009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Howlader N, et al. SEER Cancer Statistics Review, 1975–2009 (Vintage 2009 Populations) National Cancer Institute; 2012. [online], http://seer.cancer.gov/csr/1975_2009_pops09/ [Google Scholar]
- 6.Lopez RI, et al. Prognostic factor analysis, for patients with no evidence of disease after initial chemotherapy for advanced epithelial ovarian carcinoma. Int. J. Gynecol. Cancer. 1996;6:8–14. [Google Scholar]
- 7.Tan DS, Agarwal R, Kaye SB. Mechanisms of transcoelomic metastasis in ovarian cancer. Lancet Oncol. 2006;7:925–934. doi: 10.1016/S1470-2045(06)70939-1. [DOI] [PubMed] [Google Scholar]
- 8.Bagnato A, Rosano L. Epithelial-mesenchymal transition in ovarian cancer progression: a crucial role for the endothelin axis. Cells Tissues Organs. 2007;185:85–94. doi: 10.1159/000101307. [DOI] [PubMed] [Google Scholar]
- 9.Auersperg N, Wong AS, Choi KC, Kang SK, Leung PC. Ovarian surface epithelium: biology, endocrinology, and pathology. Endocr. Rev. 2001;22:255–288. doi: 10.1210/edrv.22.2.0422. [DOI] [PubMed] [Google Scholar]
- 10.Gardner MJ, Catterall JB, Jones LM, Turner GA. Human ovarian tumour cells can bind hyaluronic acid via membrane CD44: a possible step in peritoneal metastasis. Clin. Exp. Metastasis. 1996;14:325–334. doi: 10.1007/BF00123391. [DOI] [PubMed] [Google Scholar]
- 11.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. 1997;57:1228–1232. [PubMed] [Google Scholar]
- 12.Wagner BJ, et al. Simvastatin reduces tumor cell adhesion to human peritoneal mesothelial cells by decreased expression of VCAM-1 and β1 integrin. Int. J. Oncol. 2011;39:1593–1600. doi: 10.3892/ijo.2011.1167. [DOI] [PubMed] [Google Scholar]
- 13.Rump A, et al. Binding of ovarian cancer antigen CA125/MUC16 to mesothelin mediates cell adhesion. J. Biol. Chem. 2004;279:9190–9198. doi: 10.1074/jbc.M312372200. [DOI] [PubMed] [Google Scholar]
- 14.Gubbels JA, et al. Mesothelin-MUC16 binding is a high affinity, N-glycan dependent interaction that facilitates peritoneal metastasis of ovarian tumors. Mol. Cancer. 2006;566:50. doi: 10.1186/1476-4598-5-50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Renkin EM. Some consequences of capillary permeability to macromolecules: Starling’s hypothesis reconsidered. Am. J. Physiol. 1986;250:H706–710. doi: 10.1152/ajpheart.1986.250.5.H706. [DOI] [PubMed] [Google Scholar]
- 16.Mutsaers SE. The mesothelial cell. Int. J. Biochem. Cell Biol. 2004;36:9–16. doi: 10.1016/s1357-2725(03)00242-5. [DOI] [PubMed] [Google Scholar]
- 17.Mutsaers SE. Mesothelial cells: their structure, function and role in serosal repair. Respirology. 2002;7:171–191. doi: 10.1046/j.1440-1843.2002.00404.x. [DOI] [PubMed] [Google Scholar]
- 18.von Recklinghausen FT. Zur fettre sorption. Arch. Pathol. Anat. Physiol. 1863;2666:172. [Google Scholar]
- 19.Sodek KL, Murphy KJ, Brown TJ, Ringuette MJ. Cell-cell and cell-matrix dynamics in intraperitoneal cancer metastasis. Cancer Metastasis Rev. 2012;31:397–414. doi: 10.1007/s10555-012-9351-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Holm-Nielsen P. Pathogenesis of ascites in peritoneal carcinomatosis. Acta Pathol. Microbiol. Scand. 1953;33:10–21. doi: 10.1111/j.1699-0463.1953.tb04805.x. [DOI] [PubMed] [Google Scholar]
- 21.Feldman GB, Knapp RC, Order SE, Hellman S. The role of lymphatic obstruction in the formation of ascites in a murine ovarian carcinoma. Cancer Res. 1972;32:1663–1666. [PubMed] [Google Scholar]
- 22.Nagy JA, Herzberg KT, Dvorak JM, Dvorak HF. Pathogenesis of malignant ascites formation: initiating events that lead to fluid accumulation. Cancer Res. 1993;53:2631–2643. [PubMed] [Google Scholar]
- 23.Garrison RN, Galloway RH, Heuser LS. Mechanisms of malignant ascites production. J. Surg. Res. 1987;42:126–132. doi: 10.1016/0022-4804(87)90109-0. [DOI] [PubMed] [Google Scholar]
- 24.Hirabayashi K, Graham J. Genesis of ascites in ovarian cancer. Am. J. Obstet. Gynecol. 1970;106:492–497. doi: 10.1016/0002-9378(70)90031-1. [DOI] [PubMed] [Google Scholar]
- 25.Senger DR, et al. Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid. Science. 1983;219:983–985. doi: 10.1126/science.6823562. [DOI] [PubMed] [Google Scholar]
- 26.Neufeld G, Cohen T, Gengrinovitch S, Poltorak Z. Vascular endothelial growth factor (VEGF) and its receptors. FASEB J. 1999;13:9–22. [PubMed] [Google Scholar]
- 27.Geva E, Jaffe RB. Role of vascular endothelial growth factor in ovarian physiology and pathology. Fertil. Steril. 2000;74:429–438. doi: 10.1016/s0015-0282(00)00670-1. [DOI] [PubMed] [Google Scholar]
- 28.Zebrowski BK, et al. Markedly elevated levels of vascular endothelial growth factor in malignant ascites. Ann. Surg. Oncol. 1999;6:373–378. doi: 10.1007/s10434-999-0373-0. [DOI] [PubMed] [Google Scholar]
- 29.Barton DP, et al. Angiogenic protein expression in advanced epithelial ovarian cancer. Clin. Cancer Res. 1997;3:1579–1586. [PubMed] [Google Scholar]
- 30.Kassim SK, et al. Vascular endothelial growth factor and interleukin-8 are associated with poor prognosis in epithelial ovarian cancer patients. Clin. Biochem. 2004;37:363–369. doi: 10.1016/j.clinbiochem.2004.01.014. [DOI] [PubMed] [Google Scholar]
- 31.Paley PJ, et al. Vascular endothelial growth factor expression in early stage ovarian carcinoma. Cancer. 1997;80:98–106. doi: 10.1002/(sici)1097-0142(19970701)80:1<98::aid-cncr13>3.0.co;2-a. [DOI] [PubMed] [Google Scholar]
- 32.Bamias A, et al. Correlation of NK T-like CD3+CD56+ cells and CD4+CD25+(hi) regulatory T cells with VEGF and TNFα in ascites from advanced ovarian cancer: association with platinum resistance and prognosis in patients receiving first-line, platinum-based chemotherapy. Gynecol. Oncol. 2008;108:421–427. doi: 10.1016/j.ygyno.2007.10.018. [DOI] [PubMed] [Google Scholar]
- 33.Santin AD, et al. Secretion of vascular endothelial growth factor in ovarian cancer. Eur. J. Gynaecol. Oncol. 1999;20:177–181. [PubMed] [Google Scholar]
- 34.Schumacher JJ, et al. Modulation of angiogenic phenotype alters tumorigenicity in rat ovarian epithelial cells. Cancer Res. 2007;67:3683–3690. doi: 10.1158/0008-5472.CAN-06-3608. [DOI] [PubMed] [Google Scholar]
- 35.Byrne AT, et al. Vascular endothelial growth factor-trap decreases tumor burden, inhibits ascites, and causes dramatic vascular remodeling in an ovarian cancer model. Clin. Cancer Res. 2003;9:5721–5728. [PubMed] [Google Scholar]
- 36.Mesiano S, Ferrara N, Jaffe RB. Role of vascular endothelial growth factor in ovarian cancer: inhibition of ascites formation by immunoneutralization. Am. J. Pathol. 1998;153:1249–1256. doi: 10.1016/S0002-9440(10)65669-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Yukita A, Asano M, Okamoto T, Mizutani S, Suzuki H. Suppression of ascites formation and re-accumulation associated with human ovarian cancer by an anti-VPF monoclonal antibody in vivo. Anticancer Res. 2000;20:155–160. [PubMed] [Google Scholar]
- 38.Herr D, et al. VEGF induces ascites in ovarian cancer patients via increasing peritoneal permeability by downregulation of Claudin 5. Gynecol. Oncol. 2012;127:210–216. doi: 10.1016/j.ygyno.2012.05.002. [DOI] [PubMed] [Google Scholar]
- 39.Rodewald M, et al. Regulation of tight junction proteins occludin and claudin 5 in the primate ovary during the ovulatory cycle and after inhibition of vascular endothelial growth factor. Mol. Hum. Reprod. 2007;13:781–789. doi: 10.1093/molehr/gam066. [DOI] [PubMed] [Google Scholar]
- 40.Saitou M, et al. Complex phenotype of mice lacking occludin, a component of tight junction strands. Mol. Biol. Cell. 2000;11:4131–4142. doi: 10.1091/mbc.11.12.4131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Dejana E. Endothelial cell-cell junctions: happy together. Nature Rev. Mol. Cell Biol. 2004;5:261–270. doi: 10.1038/nrm1357. [DOI] [PubMed] [Google Scholar]
- 42.Takahashi A, Kondoh M, Kodaka M, Yagi K. Peptides as tight junction modulators. Curr. Pharm. Des. 2011;17:2699–2703. doi: 10.2174/138161211797416084. [DOI] [PubMed] [Google Scholar]
- 43.Nitta T, et al. Size-selective loosening of the blood-brain barrier in claudin-5-deficient mice. J. Cell Biol. 2003;161:653–660. doi: 10.1083/jcb.200302070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Dvorak HF, Nagy JA, Feng D, Brown LF, Dvorak AM. Vascular permeability factor/vascular endothelial growth factor and the significance of microvascular hyperpermeability in angiogenesis. Curr. Top. Microbiol. Immunol. 1999;237:97–132. doi: 10.1007/978-3-642-59953-8_6. [DOI] [PubMed] [Google Scholar]
- 45.Esser S, Lampugnani MG, Corada M, Dejana E, Risau W. Vascular endothelial growth factor induces VE-cadherin tyrosine phosphorylation in endothelial cells. J. Cell Sci. 1998;111:1853–1865. doi: 10.1242/jcs.111.13.1853. [DOI] [PubMed] [Google Scholar]
- 46.Horiuchi A, et al. Hypoxia-induced changes in the expression of VEGF, HIF-1 α and cell cycle-related molecules in ovarian cancer cells. Anticancer Res. 2002;22:2697–2702. [PubMed] [Google Scholar]
- 47.Hu YL, et al. Lysophosphatidic acid induction of vascular endothelial growth factor expression in human ovarian cancer cells. J. Natl Cancer Inst. 2001;93:762–768. doi: 10.1093/jnci/93.10.762. [DOI] [PubMed] [Google Scholar]
- 48.Kulbe H, et al. The inflammatory cytokine tumor necrosis factor-α generates an autocrine tumor-promoting network in epithelial ovarian cancer cells. Cancer Res. 2007;67:585–592. doi: 10.1158/0008-5472.CAN-06-2941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Salani D, et al. Role of endothelin-1 in neovascularization of ovarian carcinoma. Am. J. Pathol. 2000;157:1537–1547. doi: 10.1016/S0002-9440(10)64791-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Gupta RA, et al. Cyclooxygenase-1 is overexpressed and promotes angiogenic growth factor production in ovarian cancer. Cancer Res. 2003;63:906–911. [PubMed] [Google Scholar]
- 51.Stadlmann S, et al. 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. 2005;97:784–789. doi: 10.1016/j.ygyno.2005.02.017. [DOI] [PubMed] [Google Scholar]
- 52.Belotti D, et al. Matrix metalloproteinases (MMP9 and MMP2) induce the release of vascular endothelial growth factor (VEGF) by ovarian carcinoma cells: implications for ascites formation. Cancer Res. 2003;63:5224–5229. [PubMed] [Google Scholar]
- 53.Chen Y, Gou X, Ke X, Cui H, Chen Z. Human tumor cells induce angiogenesis through positive feedback between CD147 and insulin-like growth factor-I. PLoS ONE. 2012;766:e40965. doi: 10.1371/journal.pone.0040965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Liu LZ, et al. Reactive oxygen species regulate epidermal growth factor-induced vascular endothelial growth factor and hypoxia-inducible factor-1α expression through activation of AKT and P70S6K1 in human ovarian cancer cells. Free Radic. Biol. Med. 2006;41:1521–1533. doi: 10.1016/j.freeradbiomed.2006.08.003. [DOI] [PubMed] [Google Scholar]
- 55.Matei D, et al. PDGF BB induces VEGF secretion in ovarian cancer. Cancer Biol. Ther. 2007;6:1951–1959. doi: 10.4161/cbt.6.12.4976. [DOI] [PubMed] [Google Scholar]
- 56.Liao S, et al. TGF-β blockade controls ascites by preventing abnormalization of lymphatic vessels in orthotopic human ovarian carcinoma models. Clin. Cancer Res. 2011;17:1415–1424. doi: 10.1158/1078-0432.CCR-10-2429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Mills GB, May C, McGill M, Roifman CM, Mellors AA. Putative new growth factor in ascitic fluid from ovarian cancer patients: identification, characterization, and mechanism of action. Cancer Res. 1988;48:1066–1071. [PubMed] [Google Scholar]
- 58.Fang X, et al. Mechanisms for lysophosphatidic acid-induced cytokine production in ovarian cancer cells. J. Biol. Chem. 2004;279:9653–9661. doi: 10.1074/jbc.M306662200. [DOI] [PubMed] [Google Scholar]
- 59.Murph MM, et al. Lysophosphatidic acid-induced transcriptional profile represents serous epithelial ovarian carcinoma and worsened prognosis. PLoS ONE. 2009;4:e5583. doi: 10.1371/journal.pone.0005583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Jankowski M. Autotaxin: its role in biology of melanoma cells and as a pharmacological target. Enzyme Res. 2011;19:48–57. doi: 10.4061/2011/194857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Bast RC, Jr, Hennessy B, Mills GB. The biology of ovarian cancer: new opportunities for translation. Nature Rev. Cancer. 2009;9:415–428. doi: 10.1038/nrc2644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Mills GB, Moolenaar WH. The emerging role of lysophosphatidic acid in cancer. Nature Rev. Cancer. 2003;3:582–591. doi: 10.1038/nrc1143. [DOI] [PubMed] [Google Scholar]
- 63.Abrahams VM, et al. Epithelial ovarian cancer cells secrete functional Fas ligand. Cancer Res. 2003;63:5573–5581. [PubMed] [Google Scholar]
- 64.Bjorge L, et al. Ascitic complement system in ovarian cancer. Br. J. Cancer. 2005;92:895–905. doi: 10.1038/sj.bjc.6602334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Webb TJ, et al. Molecular identification of GD3 as a suppressor of the innate immune response in ovarian cancer. Cancer Res. 2012;72:3744–3752. doi: 10.1158/0008-5472.CAN-11-2695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Milliken D, Scotton C, Raju S, Balkwill F, Wilson J. Analysis of chemokines and chemokine receptor expression in ovarian cancer ascites. Clin. Cancer Res. 2002;8:1108–1114. [PubMed] [Google Scholar]
- 67.Guo Y, et al. Effects of siltuximab on the IL-6-induced signaling pathway in ovarian cancer. Clin. Cancer Res. 2010;16:5759–5769. doi: 10.1158/1078-0432.CCR-10-1095. [DOI] [PubMed] [Google Scholar]
- 68.Matte I, Lane D, Laplante C, Rancourt C, Piche A. Profiling of cytokines in human epithelial ovarian cancer ascites. Am. J. Cancer Res. 2012;2:566–580. [PMC free article] [PubMed] [Google Scholar]
- 69.Penson RT, et al. Cytokines IL-1β, IL-2, IL-6, IL-8, MCP-1, GM-CSF and TNFα in patients with epithelial ovarian cancer and their relationship to treatment with paclitaxel. Int. J. Gynecol. Cancer. 2000;10:33–41. doi: 10.1046/j.1525-1438.2000.00003.x. [DOI] [PubMed] [Google Scholar]
- 70.Kryczek I, Grybos M, Karabon L, Klimczak A, Lange A. IL-6 production in ovarian carcinoma is associated with histiotype and biological characteristics of the tumour and influences local immunity. Br. J. Cancer. 2000;82:621–628. doi: 10.1054/bjoc.1999.0973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Huang S, Robinson JB, DeGuzman A, Bucana CD, Fidler IJ. Blockade of nuclear factor-κB signaling inhibits angiogenesis and tumorigenicity of human ovarian cancer cells by suppressing expression of vascular endothelial growth factor and interleukin 8. Cancer Res. 2000;60:5334–5339. [PubMed] [Google Scholar]
- 72.Yoneda J, et al. Expression of angiogenesis-related genes and progression of human ovarian carcinomas in nude mice. J. Natl Cancer Institute. 1998;90:447–454. doi: 10.1093/jnci/90.6.447. [DOI] [PubMed] [Google Scholar]
- 73.Obata N, Tamakoshi K, Shibata K, Kikkawa F, Tomoda Y. Effects of interleukin-6 on in vitro cell attachment, migration and invasion of human carcinoma. Anticancer Res. 1997;17:337–342. [PubMed] [Google Scholar]
- 74.Woolery KT, Kruk PA. Ovarian epithelial-stromal interactions: role of interleukins 1 and 6. Obstet. Gynecol. Int. 2011;35:84–93. doi: 10.1155/2011/358493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Nilsson MB, Langley RR, Fidler IJ. Interleukin-6, secreted by human ovarian carcinoma cells, is a potent proangiogenic cytokine. Cancer Res. 2005;65:10794–10800. doi: 10.1158/0008-5472.CAN-05-0623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Alberti C, et al. Ligand-dependent EGFR activation induces the co-expression of IL-6 and PAI-1 via the NFkB pathway in advanced-stage epithelial ovarian cancer. Oncogene. 2011;31:4139–4149. doi: 10.1038/onc.2011.572. [DOI] [PubMed] [Google Scholar]
- 77.Lane D, Matte I, Rancourt C, Piche A. Prognostic significance of IL-6 and IL-8 ascites levels in ovarian cancer patients. BMC Cancer. 2011;1166:210. doi: 10.1186/1471-2407-11-210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Naldini A, et al. Identification of thrombin-like activity in ovarian cancer associated ascites and modulation of multiple cytokine networks. Thromb. Haemost. 2011;106:705–711. doi: 10.1160/TH11-05-0311. [DOI] [PubMed] [Google Scholar]
- 79.Gubbels JA, et al. MUC16 provides immune protection by inhibiting synapse formation between NK and ovarian tumor cells. Mol. Cancer. 2010;966:11. doi: 10.1186/1476-4598-9-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Sica A, Saccani A, Mantovani A. Tumor-associated macrophages: a molecular perspective. Int. Immunopharmacol. 2002;2:1045–1054. doi: 10.1016/s1567-5769(02)00064-4. [DOI] [PubMed] [Google Scholar]
- 81.Hagemann T, et al. Ovarian cancer cells polarize macrophages toward a tumor-associated phenotype. J. Immunol. 2006;176:5023–5032. doi: 10.4049/jimmunol.176.8.5023. [DOI] [PubMed] [Google Scholar]
- 82.Balkwill F. Cancer and the chemokine network. Nature Rev. Cancer. 2004;4:540–550. doi: 10.1038/nrc1388. [DOI] [PubMed] [Google Scholar]
- 83.Wang E, et al. Peritoneal and subperitoneal stroma may facilitate regional spread of ovarian cancer. Clin. Cancer Res. 2005;11:113–122. [PubMed] [Google Scholar]
- 84.Lin YG, et al. EphA2 overexpression is associated with angiogenesis in ovarian cancer. Cancer. 2007;109:332–340. doi: 10.1002/cncr.22415. [DOI] [PubMed] [Google Scholar]
- 85.Niu G, et al. Constitutive Stat3 activity up-regulates VEGF expression and tumor angiogenesis. Oncogene. 2002;21:2000–2008. doi: 10.1038/sj.onc.1205260. [DOI] [PubMed] [Google Scholar]
- 86.Rosen DG, et al. The role of constitutively active signal transducer and activator of transcription 3 in ovarian tumorigenesis and prognosis. Cancer. 2006;107:2730–2740. doi: 10.1002/cncr.22293. [DOI] [PubMed] [Google Scholar]
- 87.Lane D, Robert V, Grondin R, Rancourt C, Piche A. Malignant ascites protect against TRAIL-induced apoptosis by activating the PI3K/Akt pathway in human ovarian carcinoma cells. Int. J. Cancer. 2007;121:1227–1237. doi: 10.1002/ijc.22840. [DOI] [PubMed] [Google Scholar]
- 88.Lane D, Goncharenko-Khaider N, Rancourt C, Piche A. Ovarian cancer ascites protects from TRAIL-induced cell death through αvβ5 integrin-mediated focal adhesion kinase and Akt activation. Oncogene. 2010;29:3519–3531. doi: 10.1038/onc.2010.107. [DOI] [PubMed] [Google Scholar]
- 89.Goncharenko-Khaider N, Matte I, Lane D, Rancourt C, Piche A. Ovarian cancer ascites increase Mcl-1 expression in tumor cells through ERK1/2-Elk-1 signaling to attenuate TRAIL-induced apoptosis. Mol. Cancer. 2012;1166:84. doi: 10.1186/1476-4598-11-84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Peart TM, Correa RJ, Valdes YR, Dimattia GE, Shepherd TG. BMP signalling controls the malignant potential of ascites-derived human epithelial ovarian cancer spheroids via AKT kinase activation. Clin. Exp. Metastasis. 2012;29:293–313. doi: 10.1007/s10585-011-9451-3. [DOI] [PubMed] [Google Scholar]
- 91.Bookman MA, et al. Evaluation of new platinum-based treatment regimens in advanced-stage ovarian cancer: a Phase III Trial of the Gynecologic Cancer Intergroup. J. Clin. Oncol. 2009;27:1419–1425. doi: 10.1200/JCO.2008.19.1684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.De Placido S, et al. Topotecan compared with no therapy after response to surgery and carboplatin/paclitaxel in patients with ovarian cancer: Multicenter Italian Trials in Ovarian Cancer (MITO-1) randomized study. J. Clin. Oncol. 2004;22:2635–2642. doi: 10.1200/JCO.2004.09.088. [DOI] [PubMed] [Google Scholar]
- 93.du Bois A, et al. Addition of epirubicin as a third drug to carboplatin-paclitaxel in first-line treatment of advanced ovarian cancer: a prospectively randomized gynecologic cancer intergroup trial by the Arbeitsgemeinschaft Gynaekologische Onkologie Ovarian Cancer Study Group and the Groupe d’Investigateurs Nationaux pour l’Etude des Cancers Ovariens. J. Clin. Oncol. 2006;24:1127–1135. doi: 10.1200/JCO.2005.03.2938. [DOI] [PubMed] [Google Scholar]
- 94.Perren TJ, et al. A phase 3 trial of bevacizumab in ovarian cancer. New Engl. J. Med. 2011;365:2484–2496. doi: 10.1056/NEJMoa1103799. [DOI] [PubMed] [Google Scholar]
- 95.Burger RA, et al. Incorporation of bevacizumab in the primary treatment of ovarian cancer. New Engl. J. Med. 2011;365:2473–2483. doi: 10.1056/NEJMoa1104390. [DOI] [PubMed] [Google Scholar]
- 96.Kristensen GP, et al. Result of interim analysis of overall survival in the GCIG ICON7 phase III randomized trial of bevacizumab in women with newly diagnosed ovarian cancer. J. Clin. Oncol. Abstr. 2011;29:LBA5006. [Google Scholar]
- 97.Aghajanian C, et al. OCEANS: a randomized, double-blind, placebo-controlled phase III trial of chemotherapy with or without bevacizumab in patients with platinum-sensitive recurrent epithelial ovarian, primary peritoneal, or fallopian tube cancer. J. Clin. Oncol. 2012;30:2039–2045. doi: 10.1200/JCO.2012.42.0505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Pujade-Lauraine E, et al. AURELIA: a randomized phase III trial evaluating bevacizumab (BEV) plus chemotherapy (CT) for platinum (PT)-resistant recurrent ovarian cancer (OC) J. Clin. Oncol. Abstr. 2012;30:LBA5002. [Google Scholar]
- 99.Sennino B, McDonald DM. Controlling escape from angiogenesis inhibitors. Nature Rev. Cancer. 2012;12:699–709. doi: 10.1038/nrc3366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Runyon BA. Management of adult patients with ascites due to cirrhosis: an update. Hepatology. 2009;49:2087–2107. doi: 10.1002/hep.22853. [DOI] [PubMed] [Google Scholar]
- 101.Lee CW, Bociek G, Faught W. A survey of practice in management of malignant ascites. J. Pain Symptom Manage. 1998;16:96–101. doi: 10.1016/s0885-3924(98)00037-2. [DOI] [PubMed] [Google Scholar]
- 102.Becker G, Galandi D, Blum HE. Malignant ascites: systematic review and guideline for treatment. Eur. J. Cancer. 2006;42:589–597. doi: 10.1016/j.ejca.2005.11.018. [DOI] [PubMed] [Google Scholar]
- 103.Pockros PJ, Esrason KT, Nguyen C, Duque J, Woods S. Mobilization of malignant ascites with diuretics is dependent on ascitic fluid characteristics. Gastroenterology. 1992;103:1302–1306. doi: 10.1016/0016-5085(92)91520-e. [DOI] [PubMed] [Google Scholar]
- 104.Cavazzoni E, Bugiantella W, Graziosi L, Franceschini MS, Donini A. Malignant ascites: pathophysiology and treatment. Int. J. Clin. Oncol. 2012 Mar 31; doi: 10.1007/s10147-012-0396-6. (doi:10.1007/s10147-012-0396-6) [DOI] [PubMed] [Google Scholar]
- 105.Kalambokis G, et al. Renal effects of treatment with diuretics, octreotide or both, in non-azotemic cirrhotic patients with ascites. Nephrol. Dial. Transplant. 2005;20:1623–1629. doi: 10.1093/ndt/gfh871. [DOI] [PubMed] [Google Scholar]
- 106.Jatoi A, et al. A pilot study of long-acting octreotide for symptomatic malignant ascites. Oncology. 2012;82:315–320. doi: 10.1159/000337246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Dedrick RL, Myers CE, Bungay PM, DeVita VT., Jr. Pharmacokinetic rationale for peritoneal drug administration in the treatment of ovarian cancer. Cancer Treat. Rep. 1978;62:1–11. [PubMed] [Google Scholar]
- 108.Armstrong DK, et al. Intraperitoneal cisplatin and paclitaxel in ovarian cancer. N. Engl. J. Med. 2006;354:34–43. doi: 10.1056/NEJMoa052985. [DOI] [PubMed] [Google Scholar]
- 109.Ostrowski MJ. An assessment of the long-term results of controlling the reaccumulation of malignant effusions using intracavity bleomycin. Cancer. 1986;57:721–727. doi: 10.1002/1097-0142(19860215)57:4<721::aid-cncr2820570407>3.0.co;2-5. [DOI] [PubMed] [Google Scholar]
- 110.Maiche AG. Management of peritoneal effusions with intracavitary mitoxantrone or bleomycin. Anticancer Drugs. 1994;5:305–308. doi: 10.1097/00001813-199406000-00007. [DOI] [PubMed] [Google Scholar]
- 111.Link KH, et al. Intraperitoneal chemotherapy with mitoxantrone in malignant ascites. Surg. Oncol. Clin. N. Am. 2003;12:865–872. doi: 10.1016/s1055-3207(03)00050-4. [DOI] [PubMed] [Google Scholar]
- 112.Schilsky RL, et al. Phase I clinical and pharmacologic study of intraperitoneal cisplatin and fluorouracil in patients with advanced intraabdominal cancer. J. Clin. Oncol. 1990;8:2054–2061. doi: 10.1200/JCO.1990.8.12.2054. [DOI] [PubMed] [Google Scholar]
- 113.Mackey JR, Wood L, Nabholtz J, Jensen J, Venner P. A phase II trial of triamcinolone hexacetanide for symptomatic recurrent malignant ascites. J. Pain Symptom Manage. 2000;19:193–199. doi: 10.1016/s0885-3924(00)00106-8. [DOI] [PubMed] [Google Scholar]
- 114.Stuart GC, Nation JG, Snider DD, Thunberg P. Intraperitoneal interferon in the management of malignant ascites. Cancer. 1993;71:2027–2030. doi: 10.1002/1097-0142(19930315)71:6<2027::aid-cncr2820710617>3.0.co;2-c. [DOI] [PubMed] [Google Scholar]
- 115.Rath U, et al. Effect of intraperitoneal recombinant human tumour necrosis factor α on malignant ascites. Eur. J. Cancer. 1991;27:121–125. doi: 10.1016/0277-5379(91)90467-r. [DOI] [PubMed] [Google Scholar]
- 116.Heiss MM, et al. The trifunctional antibody catumaxomab for the treatment of malignant ascites due to epithelial cancer: results of a prospective randomized phase II/III trial. Int. J. Cancer. 2010;127:2209–2221. doi: 10.1002/ijc.25423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Ott MG, et al. Humoral response to catumaxomab correlates with clinical outcome: results of the pivotal phase II/III study in patients with malignant ascites. Int. J. Cancer. 2012;130:2195–2203. doi: 10.1002/ijc.26258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Beattie GJ, Smyth JF. Phase I study of intraperitoneal metalloproteinase inhibitor BB94 in patients with malignant ascites. Clin. Cancer Res. 1998;4:1899–1902. [PubMed] [Google Scholar]
- 119.Numnum TM, Rocconi RP, Whitworth J, Barnes MN. The use of bevacizumab to palliate symptomatic ascites in patients with refractory ovarian carcinoma. Gynecol. Oncol. 2006;102:425–428. doi: 10.1016/j.ygyno.2006.05.018. [DOI] [PubMed] [Google Scholar]
- 120.El-Shami K, Elsaid A, El-Kerm Y. Open-label safety and efficacy pilot trial of intraperitoneal bevacizumab as palliative treatment in refractory malignant ascites. J. Clin. Oncol. 2007;25:9043. [Google Scholar]
- 121.Hamilton CA, et al. Intraperitoneal bevacizumab for the palliation of malignant ascites in refractory ovarian cancer. Gynecol. Oncol. 2008;111:530–532. doi: 10.1016/j.ygyno.2008.04.028. [DOI] [PubMed] [Google Scholar]
- 122.Bellati F, et al. Complete remission of ovarian cancer induced intractable malignant ascites with intraperitoneal bevacizumab. Immunological observations and a literature review. Invest. New Drugs. 2010;28:887–894. doi: 10.1007/s10637-009-9351-4. [DOI] [PubMed] [Google Scholar]
- 123.Holash J, et al. VEGF-Trap: a VEGF blocker with potent antitumor effects. Proc. Natl Acad. Sci. 2002;99:11393–11398. doi: 10.1073/pnas.172398299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Colombo N, et al. A phase II study of aflibercept in patients with advanced epithelial ovarian cancer and symptomatic malignant ascites. Gynecol. Oncol. 2012;125:42–47. doi: 10.1016/j.ygyno.2011.11.021. [DOI] [PubMed] [Google Scholar]
- 125.Gotlieb WH, et al. Intravenous aflibercept for treatment of recurrent symptomatic malignant ascites in patients with advanced ovarian cancer: a phase 2, randomised, double-blind, placebo-controlled study. Lancet Oncol. 2012;13:154–162. doi: 10.1016/S1470-2045(11)70338-2. [DOI] [PubMed] [Google Scholar]
- 126.LeVeen HH, et al. Peritoneo-venous shunting for ascites. Ann. Surg. 1974;180:580–591. doi: 10.1097/00000658-197410000-00023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Mamada Y, et al. Peritoneovenous shunts for palliation of malignant ascites. J. Nippon Med. Sch. 2007;74:355–358. doi: 10.1272/jnms.74.355. [DOI] [PubMed] [Google Scholar]
- 128.White MA, Agle SC, Padia RK, Zervos EE. Denver peritoneovenous shunts for the management of malignant ascites: a review of the literature in the post LeVeen Era. Am. Surg. 2011;77:1070–1075. doi: 10.1177/000313481107700830. [DOI] [PubMed] [Google Scholar]
- 129.Saiz-Mendiguren R, et al. Permanent tunneled drainage for malignant ascites: initial experience with the PleurX® catheter. Radiologia. 2010;52:541–545. doi: 10.1016/j.rx.2010.06.005. [DOI] [PubMed] [Google Scholar]
- 130.Fleming ND, Alvarez-Secord A, Von Gruenigen V, Miller MJ, Abernethy AP. Indwelling catheters for the management of refractory malignant ascites: a systematic literature overview and retrospective chart review. J. Pain Symptom Manage. 2009;38:341–349. doi: 10.1016/j.jpainsymman.2008.09.008. [DOI] [PubMed] [Google Scholar]
- 131.Tapping CR, Ling L, Razack A. PleurX drain use in the management of malignant ascites: safety, complications, long-term patency and factors predictive of success. Br. J. Radiol. 2012;85:623–628. doi: 10.1259/bjr/24538524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Kmietowicz Z. Cancer patients should have access to device to treat fluid retention at home, says NICE. BMJ. 2012;344:e2272. doi: 10.1136/bmj.e2272. [DOI] [PubMed] [Google Scholar]
- 133.Puiffe M-L, et al. Characterization of ovarian cancer ascites on cell. Invasion, proliferation, spheroid formation, and gene expression in an in vitro model of epithelial ovarian cancer. Neoplasia. 2007;9:820–829. doi: 10.1593/neo.07472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Lee J. m., Kohn EC. Proteomics as a guiding tool for more effective personalized therapy. Ann. Oncol. 2010;21:205–210. doi: 10.1093/annonc/mdq375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Morozova O, Marra MA. Applications of next-generation sequencing technologies in functional genomics. Genomics. 2008;92:255–264. doi: 10.1016/j.ygeno.2008.07.001. [DOI] [PubMed] [Google Scholar]
- 136.Hetland TE, et al. Class III β-tubulin expression in advanced-stage serous ovarian carcinoma effusions is associated with poor survival and primary chemoresistance. Hum. Pathol. 2011;42:1019–1026. doi: 10.1016/j.humpath.2010.10.025. [DOI] [PubMed] [Google Scholar]
- 137.Gillet J-P, et al. Clinical relevance of multidrug resistance gene expression in ovarian serous carcinoma effusions. Mol. Pharm. 2011;8:2080–2088. doi: 10.1021/mp200240a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Shepherd TG, Thériault BL, Campbell EJ, Nachtigal MW. Primary culture of ovarian surface epithelial cells and ascites-derived ovarian cancer cells from patients. Nature Protoc. 2007;1:2643–2649. doi: 10.1038/nprot.2006.328. [DOI] [PubMed] [Google Scholar]
- 139.Hu L, McArthur C, Jaffe RB. Ovarian cancer stem-like side-population cells are tumourigenic and chemoresistant. Br. J. Cancer. 2010;102:1276–1283. doi: 10.1038/sj.bjc.6605626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Mukhopadhyay A, et al. Development of a functional assay for homologous recombination status in primary cultures of epithelial ovarian tumor and correlation with sensitivity to poly(ADP-ribose) polymerase inhibitors. Clin. Cancer Res. 2010;16:2344–2351. doi: 10.1158/1078-0432.CCR-09-2758. [DOI] [PubMed] [Google Scholar]
- 141.Fong PC, et al. Poly(ADP)-ribose polymerase inhibition: frequent durable responses in BRCA carrier ovarian cancer correlating with platinum-free interval. J. Clin. Oncol. 2010;28:2512–2519. doi: 10.1200/JCO.2009.26.9589. [DOI] [PubMed] [Google Scholar]
- 142.Elattar A, et al. Androgen receptor expression is a biological marker for androgen sensitivity in high grade serous epithelial ovarian cancer. Gynecol. Oncol. 2012;124:142–147. doi: 10.1016/j.ygyno.2011.09.004. [DOI] [PubMed] [Google Scholar]
- 143.Carden CP, et al. The association of PI3 kinase signaling and chemoresistance in advanced ovarian cancer. Mol. Cancer Ther. 2012;11:1609–1617. doi: 10.1158/1535-7163.MCT-11-0996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Rizzo S, et al. Ovarian cancer stem cell-like side populations are enriched following chemotherapy and overexpress EZH2. Mol. Cancer Ther. 2011;10:325–335. doi: 10.1158/1535-7163.MCT-10-0788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Moserle L, et al. The side population of ovarian cancer cells is a primary target of IFN-α antitumor effects. Cancer Res. 2008;68:5658–5668. doi: 10.1158/0008-5472.CAN-07-6341. [DOI] [PubMed] [Google Scholar]
- 146.Meirelles K, et al. Human ovarian cancer stem/progenitor cells are stimulated by doxorubicin but inhibited by Mullerian inhibiting substance. Proc. Natl Acad. Sci. USA. 2012;109:2358–2363. doi: 10.1073/pnas.1120733109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Latifi A, et al. Isolation and characterization of tumor cells from the ascites of ovarian cancer patients: molecular phenotype of chemoresistant ovarian tumors. PLoS ONE. 2012;766:e46858. doi: 10.1371/journal.pone.0046858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Davidson B. Proteomic analysis of malignant ovarian cancer effusions as a tool for biologic and prognostic profiling. Clin. Cancer Res. 2006;12:791–799. doi: 10.1158/1078-0432.CCR-05-2516. [DOI] [PubMed] [Google Scholar]
- 149.Yap TA, Carden CP, Kaye SB. Beyond chemotherapy: targeted therapies in ovarian cancer. Nature Rev. Cancer. 2009;9:167–181. doi: 10.1038/nrc2583. [DOI] [PubMed] [Google Scholar]
- 150.Lee J-M, Han JJ, Altwerger G, Kohn EC. Proteomics and biomarkers in clinical trials for drug development. J. Proteomics. 2011;74:2632–2641. doi: 10.1016/j.jprot.2011.04.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Runyon BA, et al. The serum-ascites albumin gradient is superior to the exudate-transudate concept in the differential diagnosis of ascites. Ann. Intern. Med. 1992;117:215–220. doi: 10.7326/0003-4819-117-3-215. [DOI] [PubMed] [Google Scholar]
- 152.Sheid B. Angiogenic effects of macrophages isolated from ascitic fluid aspirated from women with advanced ovarian cancer. Cancer Lett. 1992;62:153–158. doi: 10.1016/0304-3835(92)90186-y. [DOI] [PubMed] [Google Scholar]
- 153.Starling EH. On the absorption of fluids from the connective tissue spaces. J. Physiol. 1896;19:312–326. doi: 10.1113/jphysiol.1896.sp000596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Cattau EL, Jr, Benjamin SB, Knuff TE, Castell DO. The accuracy of the physical examination in the diagnosis of suspected ascites. JAMA. 1982;247:1164–1166. [PubMed] [Google Scholar]
- 155.Inadomi J, Cello JP, Koch J. Ultrasonographic determination of ascitic volume. Hepatology. 1996;24:549–551. doi: 10.1002/hep.510240314. [DOI] [PubMed] [Google Scholar]
- 156.Akriviadis EA. Hemoperitoneum in patients with ascites. Am. J. Gastroenterol. 1997;92:567–575. [PubMed] [Google Scholar]
- 157.von Riedenauer WB, Janjua SA, Kwon DS, Zhang Z, Velanovich V. Immunohistochemical identification of primary peritoneal serous cystadenocarcinoma mimicking advanced colorectal carcinoma: a case report. J. Med. Case Rep. 2007;166:150. doi: 10.1186/1752-1947-1-150. [DOI] [PMC free article] [PubMed] [Google Scholar]

