Abstract
Malignant pleural effusions (MPEs) are a significant source of cancer-related morbidity. Over 150,000 patients in the United States suffer from breathlessness and diminished quality of life due to MPE each year. Current management strategies are of mostly palliative value and focus on symptom control; they do not address the pathobiology of the effusion, nor do they improve survival. Further elucidation of the pathophysiological mechanisms, coupled with the development of novel treatments such as intrapleural chemotherapeutics targeting this process, has the potential to greatly improve the efficacy of our current management options. Vascular endothelial growth factor-A (VEGF-A) has been implicated as a critical cytokine in the formation of malignant pleural effusions. Elevated levels of VEGF produced by tumor cells, mesothelial cells and infiltrating immune cells, result in increased vascular permeability, cancer cell transmigration, and angiogenesis. Therefore anti-angiogenic therapies such as Bevacizumab, a monoclonal antibody targeting VEGF-A, may have a potential role in the management of malignant pleural effusions. Herein we review the pathogenesis and potential treatment strategies of malignant pleural effusions, with a focus on angiogenesis and anti-angiogenic therapeutics.
Keywords: Pleural Effusions, Angiogenesis, Vascular Endothelial Growth Factor, Lung Cancer
Introduction
Malignant pleural effusions (MPEs) are a significant source of cancer-related morbidity, debilitating patients by impairing respiratory function and decreasing quality of life dramatically in over 150,000 patients in the US yearly [1]. MPEs are common complications in cancer patients. During the course of their disease approximately 50% of all patients with metastatic cancer develop a MPE. Virtually any cancer can cause an MPE, although greater than 75% are caused by lung -, breast -, ovarian cancer, or by malignant lymphomas [2]. MPEs are especially common complications of lung cancer, with 15% of lung cancer patients having an MPE at presentation and 50% developing an MPE during the course of their disease [3, 4]. Unfortunately, MPEs are associated with a bleak prognosis, heralding a rapid deterioration with a median survival of 3 months [5]. This time is typically plagued by numerous hospitalizations and multiple interventions for symptom control [6]. Furthermore, the presence of an MPE can decrease the patient’s overall performance status and thereby affect their candidacy to receive potentially life-extending anticancer therapies.
Most of our current management strategies for MPE do not improve patient survival, largely fail to address the underlying cause of the effusion and consequently are predominantly palliative. Further elucidation of the pathogenic mechanisms, coupled with novel local and/or systemic treatments targeting these pathways, has the potential to improve the efficacy of our current management strategies. Herein we present a review of the pathophysiology, diagnosis and management of MPEs focusing on the role of vascular endothelial growth factor (VEGF) in the formation of MPEs and the rationale for VEGF-targeted treatment modalities.
VEGF in the Pathogenesis of Malignant Pleural Effusions
As far back as 1939, Ide and colleagues hypothesized the production of pro-angiogenic factors by tumors [7]. In 1971, Folkman postulated that a tumors are dependent on increasing vascular supply, and suggested that antiangiogenic therapies could serve a role in cancer treatment [8]. Subsequent research led to our increasing understanding of the mechanisms of VEGF in malignancy and the development of a number of anti-angiogenic therapeutic strategies.
Among the various mediators found in malignant effusions, VEGF has drawn interest for its central role in pleural fluid accumulation [9] and for its potential as a therapeutic target [10, 11]. VEGF is a family of endothelial growth factors which includes VEGF-A –B –C –D –E and placental growth factor [12]. This family of peptides has been the focus of extensive research with applications in effusions, cancer, hypoxic injury and normal growth and development. VEGF possesses critical functions in angiogenesis [13], exerting a number of effects on the vascular endothelium including survival, proliferation, differentiation, sprouting and tube formation [14-16]. VEGF not only possesses potent vasodilatory effects [17], but also the ability to increase vascular [18] and mesothelial permeability [19]. Increased permeability as a result of VEGF stimulation is mediated by several mechanisms (see Bates, 2010[20]for an excellent review of this subject) including induction of endothelial fenestrations [21-23], loss of junctional integrity [24] and the formation of transcellular gaps [25].
Many cancers have been shown to over-express VEGF, a finding associated with a poor prognosis in at least pancreatic [26, 27], gastric [28, 29] and colonic carcinomas [30, 31], as well as in lung [32], breast [33, 34] and prostate [35] cancers and melanoma [36]. The factors influencing the expression of VEGF include hypoxia; several growth factors such as epidermal growth factor, transforming growth factor, insulin-like growth factor, and others; a variety of hormones; and oncogenic mechanisms leading to the activation of proto-oncogenes and the dysfunction of tumor suppressor genes [37, 38]. Hypoxia is a well-established inducer of angiogenesis, which activates hypoxia-inducible factor-1, a transcription factor responsible for the regulation of a number of hypoxia-responsive genes [39]. Transcription of VEGF mRNA is initiated upon binding of the hypoxia-inducible factor-1/aryl hydrocarbon nuclear translocator complex to the promoter region [40]. The molecular target of rapamycin (mTOR) has been shown to play a role in the expression of VEGF through its ability to increase the expression of hypoxia-inducible factor-1 in hematologic and various solid malignancies [41-43]. Cancer cells may produce VEGF through autocrine signaling mediated by interleukin-6, as well, indicating that there are likely a number of upstream mechanisms for initiating VEGF production in MPEs [44]. Alternative mRNA splicing of the VEGF gene produces at least six splice variants (VEGF121-206) which have varying biologic effects, ranging from pro-angiogenic to anti-angiogenic [45, 46].
Several receptor tyrosine kinases, including VEGFR-1 -2 and -3 (also known as Flt-1, KDR/Flk-1 and Flt-4 respectively) mediate the biological response to VEGF [15]. A number of additional factors have also been shown to play a role in the cellular response to VEGF, including heparin and heparan sulfate proteoglycans, as well as co-receptors known as neuropilin-1 and -2 [46-48]. The signal transduction cascade downstream of the VEGF receptors is complex and includes at least the mitogen-activated protein kinase (MAPK), phosphatidylinositol 3-kinase/protein kinase-B (also referred to as PI3K/AKT) and protein kinase-C (PKC) pathways, which are central in actuating the array of VEGF effects [12]. VEGFR-2 mediates the majority of the angiogenic response to VEGF, including increased permeability [49], migration, invasion, proliferation and survival of the vascular endothelium [50, 51].
The role of VEGF in MPE formation is under investigation, with current data strongly implicating VEGF as a critical cytokine in MPE pathogenesis. Elevated levels of VEGF have been observed in pleural effusions due to both malignant and benign processes [52], but higher levels of VEGF are consistently found in pleural effusions of malignant origin [53-61, 49-51]. VEGF production by intrathoracic lung cancer cells has been shown to contribute to pleural effusion formation, tumor dissemination, and angiogenesis [62, 63]. Prager and colleagues have demonstrated that VEGF causes an increase in the permeability of an endothelial monolayer and induces transmigration of primary and cell-line derived cancer cells, effects which were both blocked by the administration of the anti-VEGF monoclonal antibody bevacizumab [43]. The addition of 20 nM rapamycin (Sirolimus) to culture medium also decreased the production of VEGF by both primary and cell line-derived cancer cells, a finding which suggests a potential benefit from co-administration of rapamycin and bevacizumab in MPE [43]. Interestingly, cancer stem cells have been isolated from malignant pleural effusions [153], a finding which prompted the hypothesis that VEGF may serve a chemotactic function for cancer stem cells [43]. It should be noted that VEGF does not function in isolation and a variety of other vasoactive mediators, such as osteopontin [64], chemokine ligand 2 [65], interleukin 5 [66], matrix metalloprotease 9 and tumor necrosis factor-alpha [67] have also been implicated in the formation of MPEs. An eloquent model of malignant pleural effusion pathogenesis which integrates tumor and host interactions has recently been presented by Stathopoulos and Kalomenidis [68].
Given the important functions of VEGF in cancer and a number of other conditions, multiple compounds capable of antagonizing the effects of VEGF have been developed for use in the clinical setting [69]. These include several tyrosine kinase inhibitors [70] and bevacizumab, a recombinant humanized monoclonal antibody targeted to VEGF that inhibits the binding of VEGF to its receptors VEGFR-1 and -2 [71]. Aflibercept is a fusion antibody protein with affinity for VEGF-A, VEGF-B and placental growth factor that has recently been approved by the FDA for use in colorectal cancer [72]. Multiple studies have carefully followed plasma levels of VEGF after administration of bevacizumab and have shown that >97% of circulating VEGF is bound by bevacizumab within hours of administration [73, 74]. Bevacizumab has been approved for the treatment of several malignancies, including advanced colorectal and non-small cell lung carcinomas, as well as advanced renal cell carcinoma [75].
Evidence is mounting in support of the hypothesis that the VEGF found in malignant effusions is produced within the pleural space. VEGF levels in malignant effusions are consistently much higher than serum levels; and multiple studies have shown no correlation between the levels of VEGF in malignant effusions and plasma [76-78, 57, 54]. While many cancers over-express VEGF, virtually all cells have this capability, and accumulating data suggest that non-neoplastic cells may contribute to the increased levels of VEGF observed in malignant states, including platelets [79] and tumor-associated stromal and immune cells [80-83]. Mesothelial cells have been shown to express VEGF in response to TGF-beta stimulation, both in vitro and in vivo, suggesting a potential role in malignant effusion [84]. These observations support the merit of intrapleural therapeutics targeting angiogenic pathways (see below).
Diagnosis of MPE
It is critical to accurately diagnose MPEs, as the diagnosis generally precludes the possibility of a curative resection (the seventh edition of the American Joint Committee on Cancer classifies patients with an MPE as M1a, stage IV disease) [85]. As only approximately 50% of pleural effusions in the setting of cancer are MPEs, distinguishing between malignant and para-malignant effusions significantly impacts management. Para-malignant pleural effusions can develop from such processes as lymphatic obstruction, atelectasis, pulmonary embolus or post-obstructive pneumonitis. There are several features of pleural fluid suggestive of MPE. These include lymphocytic predominance with lymphocytes representing 50-70% of nucleated cells, >10% eosinophils, the presence of erythrocytes, pH < 7.3 and glucose < 60 mg/dL [2, 86]. While the majority of MPEs are exudative effusions, it should be noted that 3-10% are transudates, according to Light’s criteria [87-89]. However, definitive diagnosis of MPE requires the identification of malignant cells within the fluid or positive pleural biopsy, as the aforementioned features are relatively non-specific [2]. Roughly 50-60% of MPEs are detected by cytologic examination after a single thoracentesis [90-92, 55], and the diagnostic yield improves with up to two additional thoracenteses. Increasing the volume of a single thoracentesis does not increase the diagnostic accuracy, however, and 50 mL is likely a sufficient volume [93, 94]. As the diagnostic yield of a single thoracentesis is insufficient to rule out MPE with a single negative result, in practice three negative thoracenteses are generally required to exclude MPE.
Diagnostic and Prognostic Implications of Biomarkers in MPE
A variety of biomarkers have been investigated as potential diagnostic and prognostic indicators of MPE including (among others) cystatin-C [95], D-dimer [96], epididymal secretory protein E1 precursor [97], lung surfactant protein-A [98], carcinoembryonic antigen [98], pigment epithelium-derived factor [99-101], pro-calcitonin and c-reactive protein [102], vascular endothelial growth factor [103, 54], and insulin-like growth factor binding protein-2 (which may also play a role in cell transmigration) [104]. While many of the biomarker levels differ between malignant and benign pleural effusions, none have yet demonstrated sufficient sensitivity and specificity to garner acceptance in the clinical setting.
The diagnostic yield of thoracentesis may be increased by the additional measurement of VEGF in pleural fluid, although as expected, the sensitivity and specificity vary with different cut-off concentrations [55]. A recent meta-analysis reviewed the potential role of pleural fluid VEGF levels in the diagnosis of MPEs and found modest maximum joint sensitivity and specificity of 0.72 with an area under the curve of 0.82 [105]. The authors concluded that the detection of pleural fluid VEGF may play some role in MPE diagnosis, but with unsatisfactory diagnostic value to be used in isolation, requiring interpretation in the context of clinical findings and conventional diagnostic studies. Notably, benign exudative effusions also demonstrate elevated VEGF levels, albeit less elevated than in MPEs, and there is some evidence to support the use of VEGF in differentiating between the two [55].
Few prognostic indicators have demonstrated strong predictive value. Prognostic indicators currently include the type of cancer, cell type, tumor stage, extent of pleural tumor involvement, presence of adhesions and performance status [106-110]. Several features of pleural fluid, including low pH and low glucose are associated with a poor prognosis [111-113], as are failed attempts at pleurodesis [114, 115]. Pleural effusion fluid levels of VEGF have been shown to carry prognostic significance in malignant pleural mesothelioma, with levels greater than or equal to 2000 pg/mL associated with a poor prognosis [116].
Standard Treatment Options in MPE
Following the diagnosis of MPE, management decisions are based on the volume and symptomatic impact of the effusion. Whereas small, asymptomatic effusions can be observed, if the patient is suffering from breathlessness, drainage of the effusion by thoracocentesis should be pursued. If there is rapid reaccumulation of a symptomatic effusion (less than 1 month), the risks and benefits of permanent pleural drainage should be considered. Treatment modalities include placement of a tunneled in-dwelling pleural catheter (TIPC, PleurX®), chemical or physical pleurodesis. Uncommon management strategies include surgical pleurectomy or the placement of a pleuroperitoneal shunt.
While talc pleurodesis has long been the preferred treatment modality, its safety and efficacy have recently been challenged [117]. A systematic review of the Cochrane database demonstrated rates of approximately 90% successful pleurodesis in patients treated with chemical pleurodesis, with talc the preferred modality [118]. However, this may be an optimistic measure, as a recently published study instituting current pleurodesis guidelines and analyzed by intention-to treat revealed that only approximately one-third of patients clearly benefit from pleurodesis [119].
Increasingly, TIPCs have drawn attention for their potential value in the management of MPEs. A recently published prospective, randomized clinical trial comparing talc pleurodesis to placement of a tunneled in-dwelling pleural catheter demonstrated improved survival, effusion control and activity without dyspnea in the catheter-treated group [120]. While use as a first-line modality continues to be debated among some clinicians, TIPCs have also been shown to be cost-effective in patients with a life expectancy of six weeks or less [121]. A retrospective review of 355 patients with 418 tunneled pleural catheters demonstrated suboptimal control of MPEs, however, with only 75% and 50% control rates at one and six months, respectively [122]. And while a recently published systematic review indicated that TIPC appear to be effective in ameliorating symptoms (95.6%) with a low rate of complications, spontaneous pleurodesis was only achieved in 45.6% and the authors note that the evidence was low-quality, being primarily based on one case series [123]. These data indicate the continued need for the development of improved treatment modalities in MPEs.
In this context, eligibility criteria for currently available clinical trials should be considered as some of these therapeutic modalities may disqualify patients from enrollment into certain trials. In patients whose effusions re-accumulate very slowly, repeated thoracocentesis represents a viable treatment option. While these treatments can provide symptomatic relief [124], they do not address the underlying cause of the MPE.
The value of systemic treatment for MPE depends upon the underlying tumor type and the expected patient survival. While for some patients systemic chemotherapy may result in increased survival—particularly if the underlying malignancy is breast-, ovarian cancer or malignant lymphoma—for others these strategies are of limited palliative value. Consequently, there is a great need for the development of improved treatments, which may have the potential to improve the currently dismal survival in patients thus afflicted.
Rationale for anti-VEGF therapy in MPE
Intrapleural therapy, as an alternate route of administration of traditional or novel chemotherapeutics and targeted agents, represents a potential modality for the management of MPEs. Studies investigating intrapleural 5-fluorouracil, taxanes, bleomycin, cytarabine, anthracyclines, platinum agents, etoposide, adenoviral-mediated interferon-beta and OK-432 (a product of heat-killed Streptococcus pyogenes) have been performed in patients with MPEs with mixed results and toxicity profiles according to the agents under investigation [125-137]. A recent trial in which staphylococcal superantigen of the enterotoxin gene cluster was instilled directly into the pleural space of 14 unselected patients with non-small cell lung carcinoma demonstrated an improvement in survival compared to talc pleurodesis (median survival of 7.9 months compared to 2.0 months respectively), with minimal toxicity [138]. Studies such as these have validated the feasibility of intrapleural delivery, and have paved the way for the application of more targeted therapeutics via the intrapleural route.
Multiple pre-clinical studies have been performed investigating the potential benefit of VEGF blockade in patients with malignant effusions (Table 1; see Gerber, 2005 [10] for a focused review). Several studies have demonstrated a significant decrease in ascites formation in mice with implanted with ovarian tumor cells that were treated with bevacizumab or A4.6.1, the murine equivalent of bevacizumab [139-141]. Virally-encoded murine A4.6.1 introduced directly into the pleural space in mice demonstrated a significant decrease in metastatic lung tumor volume and improved survival with VEGF antibody undetectable outside of the pleura and lung [142]. Studies in which mice were implanted with mouse breast cancer cells and subsequently treated with AF-493-NA, a goat anti-mouse VEGF antibody, also demonstrate significant reduction in malignant ascites formation [143, 144]. Mice given intraperitoneal injections of human colon cancer cells and treated with intraperitoneal DC101 (a mouse anti-VEGFR-2 antibody) demonstrated similarly positive results with reduction in ascites as assessed by ascites grading [145]. Yano and colleagues demonstrated decreased incidence and reduced formation of MPE in a mouse model of MPE secondary to human lung adenocarcinoma cell injection [146]. New Zealand rabbits with inflammatory pleural effusions that were treated with intrapleural bevacizumab combined with talc or silver nitrate pleurodesis demonstrated a significant reduction in pleural effusion formation compared to rabbits that did not receive bevacizumab [147]. However, it should be noted that several studies in animals have suggested that treatment with anti-angiogenic agents prior to attempted pleurodesis may reduce the success of pleural symphysis [148, 149].
Table 1.
Model (n) | Antiangiogenic Treatment | Outcome |
---|---|---|
Rabbits NMPE (50) [147] | Bev | Reduced effusion |
Murine ovarian MA (35) [140] | Bev | Virtually no formation |
Murine ovarian MA (21) [139] | A4.6.1 | No formation in 20/21 |
Murine ovarian MA (6) [141] | A4.6.1 | Reduced formation in 6/6 |
Murine breast MA (12) [143] | IP AF-493-NA | Reduced formation |
Murine colon MA (10) [145] | IP DC101 | Reduced formation |
Murine lung MPE (18) [146] | PTK 787 | No formation in 9/18 Reduced in remaining 9 |
Bev: Bevacizumab; IP: intrapleural; MA: malignant ascites; MPE: malignant pleural effusion; NMPE: nonmalignant pleural effusion. A4.6.1: murine equivalent of bevacizumab; AF-493-NA = Goat anti-mouse VEGF monoclonal antibody; DC101 = anti-mouse VEGFR-2 antibody; PTK 787 = VEGFR-2 tyrosine kinase inhibitor.
Early clinical studies investigating the role of antiangiogenic therapy in MPEs are promising (Table 2). A case report by Pichelmayer and colleagues describes a patient with a massive non-malignant pleural effusion which responded dramatically to treatment with a single dose of 5 mg/kg intravenous bevacizumab (one of the approved doses for patients with colorectal carcinoma) [150]. Pichelmayer and colleagues also report another set of cases in which patients with malignant effusions were treated with the same dose of bevacizumab with no significant effusion reduction, however [151]. One of these patients was found to have significantly elevated levels of plasma VEGF even after treatment with bevacizumab. This observation, combined with the elevated levels of VEGF known to be present in both plasma and effusion fluid of patients with MPE, prompted the treatment of two other patients with higher doses of bevacizumab (15 mg/kg). Both patients treated with this elevated dose experienced successful resolution of their effusions and dramatic reduction of serum and plasma VEGF levels (serum levels were evaluated in order to assess total VEGF load, including VEGF stored in platelets). Numnum and colleagues treated four patients with malignant ascites with 15 mg/kg bevacizumab every three weeks for palliative purposes. All four patients experienced symptomatic ascites relief with no grade 3 toxicities [11]. One of these patients also had a pleural effusion, although response of the pleural effusion to bevacizumab cannot be evaluated as this patient was treated with concomitant pleurodesis [personal correspondence]. More recently, a retrospective review of bevacizumab plus chemotherapy in patients with non-small cell lung cancer and MPE was published [152]. In this study, twelve of thirteen patients achieved MPE control for greater than 8 weeks, with a median progression-free survival time without effusion reaccumulation of 312 days. Such studies have demonstrated the clinical merit of targeted antiangiogenic therapies in MPEs and prospective clinical trials are currently in development.
Table 2.
Patients (n) | Antiangiogenic Treatment | Outcome |
---|---|---|
NMPE (1) [150] | Bev 5 mg/kgA | Reduced in 1/1 |
NMPE (1) [154] | Bev 5 mg/kgB | Resolved in 1/1 |
MPE (2) [151] | Bev 5 mg/kgC | Not Resolved in 2/2 |
MPE (13) [152] | Bev 15 mg/kgB | Controlled in 12/13 (92.3%) |
MA (4) [11] | Bev 15 mg/kgB | Controlled in 4/4 |
MA (2) [151] | Bev 15 mg/kgA | Resolved or Reduced in 2/2 |
MA (1) [155] | Bev 15 mg/kgB | Resolved in 1/1 |
MA (1) [156] | IP Bev 5 mg/kgD | Reduced in 1/1 |
MA (9) [157] | IP Bev 5 mg/kgD | Resolved in 9//9 |
Bev: Bevacizumab; IP: intraperitoneal; MA: malignant ascites; MPE: malignant pleural effusion; NMPE: nonmalignant pleural effusion. Treatment schedule as follows:
once;
every three weeks;
5 mg/kg initial dose was followed by one more dose of 5 mg/kg in one patient and 10 mg/kg in the other;
monthly.
Conclusions
In conclusion, accumulating evidence implicates VEGF in the formation of MPEs. The preclinical data described herein suggest that there may be a potential benefit of intrapleural anti-VEGF therapeutics in the treatment of malignant pleural effusions.
Footnotes
Conflict of Interest
Michael Bradshaw declares no conflict of interest.
Aaron Mansfield declares no conflict of interest.
Tobias Peikert declares no conflict of interest.
References
- 1.Haas AR, Sterman DH, Musani AI. Malignant pleural effusions: management options with consideration of coding, billing, and a decision approach. Chest. 2007;132(3):1036–41. doi: 10.1378/chest.06-1757. [DOI] [PubMed] [Google Scholar]
- 2.Heffner JE, Klein JS. Recent advances in the diagnosis and management of malignant pleural effusions. Mayo Clin Proc. 2008;83(2):235–50. doi: 10.4065/83.2.235. [DOI] [PubMed] [Google Scholar]
- 3.Anderson CB, Philpott GW, Ferguson TB. The treatment of malignant pleural effusions. Cancer. 1974;33(4):916–22. doi: 10.1002/1097-0142(197404)33:4<916::aid-cncr2820330405>3.0.co;2-u. [DOI] [PubMed] [Google Scholar]
- 4.Memon A, Zawadzki ZA. Malignant effusions: diagnostic evaluation and therapeutic strategy. Curr Probl Cancer. 1981;5(8):1–30. doi: 10.1016/s0147-0272(81)80012-8. [DOI] [PubMed] [Google Scholar]
- 5.Morgensztern D, Waqar S, Subramanian J, Trinkaus K, Govindan R. Prognostic impact of malignant pleural effusion at presentation in patients with metastatic non-small-cell lung cancer. J Thorac Oncol. 2012;7(10):1485–9. doi: 10.1097/JTO.0b013e318267223a. [DOI] [PubMed] [Google Scholar]
- 6.Antunes G, Neville E, Duffy J, Ali N. BTS guidelines for the management of malignant pleural effusions. Thorax. 2003;58(Suppl 2):ii29–38. doi: 10.1136/thorax.58.suppl_2.ii29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Ide AG, Baker NH, SL W. Vascularization of the Brown Pearce rabbit epithelioma transplant as seen in the transparent ear chamber. Am J Roentgenol. 1939;42:891–9. [Google Scholar]
- 8.Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med. 1971;285(21):1182–6. doi: 10.1056/NEJM197111182852108. [DOI] [PubMed] [Google Scholar]
- 9.Hamed EA, El-Noweihi AM, Mohamed AZ, Mahmoud A. Vasoactive mediators (VEGF and TNF-alpha) in patients with malignant and tuberculous pleural effusions. Respirology. 2004;9(1):81–6. doi: 10.1111/j.1440-1843.2003.00529.x. [DOI] [PubMed] [Google Scholar]
- 10.Gerber HP, Ferrara N. Pharmacology and pharmacodynamics of bevacizumab as monotherapy or in combination with cytotoxic therapy in preclinical studies. Cancer Res. 2005;65(3):671–80. [PubMed] [Google Scholar]
- 11.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(3):425–8. doi: 10.1016/j.ygyno.2006.05.018. [DOI] [PubMed] [Google Scholar]
- 12.Zachary I. Signaling mechanisms mediating vascular protective actions of vascular endothelial growth factor. Am J Physiol Cell Physiol. 2001;280(6):C1375–86. doi: 10.1152/ajpcell.2001.280.6.C1375. [DOI] [PubMed] [Google Scholar]
- 13.Senger DR, Galli SJ, Dvorak AM, Perruzzi CA, Harvey VS, Dvorak HF. Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid. Science. 1983;219(4587):983–5. doi: 10.1126/science.6823562. [DOI] [PubMed] [Google Scholar]
- 14.Ferrara N, Henzel WJ. Pituitary follicular cells secrete a novel heparin-binding growth factor specific for vascular endothelial cells. Biochem Biophys Res Commun. 1989;161(2):851–8. doi: 10.1016/0006-291x(89)92678-8. [DOI] [PubMed] [Google Scholar]
- 15.Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med. 2003;9(6):669–76. doi: 10.1038/nm0603-669. [DOI] [PubMed] [Google Scholar]
- 16.Hicklin DJ, Ellis LM. Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. J Clin Oncol. 2005;23(5):1011–27. doi: 10.1200/JCO.2005.06.081. [DOI] [PubMed] [Google Scholar]
- 17.Ku DD, Zaleski JK, Liu S, Brock TA. Vascular endothelial growth factor induces EDRF-dependent relaxation in coronary arteries. Am J Physiol. 1993;265(2 Pt 2):H586–92. doi: 10.1152/ajpheart.1993.265.2.H586. [DOI] [PubMed] [Google Scholar]
- 18.Brown LF, Detmar M, Claffey K, Nagy JA, Feng D, Dvorak AM, et al. Vascular permeability factor/vascular endothelial growth factor: a multifunctional angiogenic cytokine. Exs. 1997;79:233–69. doi: 10.1007/978-3-0348-9006-9_10. [DOI] [PubMed] [Google Scholar]
- 19.Mohammed KA, Nasreen N, Hardwick J, Logie CS, Patterson CE, Antony VB. Bacterial induction of pleural mesothelial monolayer barrier dysfunction. Am J Physiol Lung Cell Mol Physiol. 2001;281(1):L119–25. doi: 10.1152/ajplung.2001.281.1.L119. [DOI] [PubMed] [Google Scholar]
- 20.Bates DO. Vascular endothelial growth factors and vascular permeability. Cardiovasc Res. 2010;87(2):262–71. doi: 10.1093/cvr/cvq105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Roberts WG, Palade GE. Increased microvascular permeability and endothelial fenestration induced by vascular endothelial growth factor. J Cell Sci. 1995;108(Pt 6):2369–79. doi: 10.1242/jcs.108.6.2369. [DOI] [PubMed] [Google Scholar]
- 22.Roberts WG, Palade GE. Neovasculature induced by vascular endothelial growth factor is fenestrated. Cancer Res. 1997;57(4):765–72. [PubMed] [Google Scholar]
- 23.Satchell SC, Braet F. Glomerular endothelial cell fenestrations: an integral component of the glomerular filtration barrier. Am J Physiol Renal Physiol. 2009;296(5):F947–56. doi: 10.1152/ajprenal.90601.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Michel CC, Neal CR. Openings through endothelial cells associated with increased microvascular permeability. Microcirculation. 1999;6(1):45–54. [PubMed] [Google Scholar]
- 25.Feng D, Nagy JA, Hipp J, Pyne K, Dvorak HF, Dvorak AM. Reinterpretation of endothelial cell gaps induced by vasoactive mediators in guinea-pig, mouse and rat: many are transcellular pores. J Physiol. 1997;504(Pt 3):747–61. doi: 10.1111/j.1469-7793.1997.747bd.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Fujimoto K, Hosotani R, Wada M, Lee JU, Koshiba T, Miyamoto Y, et al. Expression of two angiogenic factors, vascular endothelial growth factor and platelet-derived endothelial cell growth factor in human pancreatic cancer, and its relationship to angiogenesis. Eur J Cancer. 1998;34(9):1439–47. doi: 10.1016/s0959-8049(98)00069-0. [DOI] [PubMed] [Google Scholar]
- 27.Ikeda N, Adachi M, Taki T, Huang C, Hashida H, Takabayashi A, et al. Prognostic significance of angiogenesis in human pancreatic cancer. Br J Cancer. 1999;79(9-10):1553–63. doi: 10.1038/sj.bjc.6690248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Maeda K, Chung YS, Ogawa Y, Takatsuka S, Kang SM, Ogawa M, et al. Prognostic value of vascular endothelial growth factor expression in gastric carcinoma. Cancer. 1996;77(5):858–63. doi: 10.1002/(sici)1097-0142(19960301)77:5<858::aid-cncr8>3.0.co;2-a. [DOI] [PubMed] [Google Scholar]
- 29.Takahashi Y, Cleary KR, Mai M, Kitadai Y, Bucana CD, Ellis LM. Significance of vessel count and vascular endothelial growth factor and its receptor (KDR) in intestinal-type gastric cancer. Clin Cancer Res. 1996;2(10):1679–84. [PubMed] [Google Scholar]
- 30.Lee JC, Chow NH, Wang ST, Huang SM. Prognostic value of vascular endothelial growth factor expression in colorectal cancer patients. Eur J Cancer. 2000;36(6):748–53. doi: 10.1016/s0959-8049(00)00003-4. [DOI] [PubMed] [Google Scholar]
- 31.Takahashi Y, Kitadai Y, Bucana CD, Cleary KR, Ellis LM. Expression of vascular endothelial growth factor and its receptor, KDR, correlates with vascularity, metastasis, and proliferation of human colon cancer. Cancer Res. 1995;55(18):3964–8. [PubMed] [Google Scholar]
- 32.Fontanini G, Lucchi M, Vignati S, et al. Angiogenesis as a prognostic indicator of survival in non-small-cell lung carcinoma: A prospective study. J Natl Cancer Inst. 1997;89:881–6. doi: 10.1093/jnci/89.12.881. [DOI] [PubMed] [Google Scholar]
- 33.Berns EM, Klijn JG, Look MP, Grebenchtchikov N, Vossen R, Peters H, et al. Combined vascular endothelial growth factor and TP53 status predicts poor response to tamoxifen therapy in estrogen receptor-positive advanced breast cancer. Clin Cancer Res. 2003;9(4):1253–8. [PubMed] [Google Scholar]
- 34.Manders P, Beex LV, Tjan-Heijnen VC, Geurts-Moespot J, Van Tienoven TH, Foekens JA, et al. The prognostic value of vascular endothelial growth factor in 574 node-negative breast cancer patients who did not receive adjuvant systemic therapy. Br J Cancer. 2002;87(7):772–8. doi: 10.1038/sj.bjc.6600555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.George DJ, Halabi S, Shepard TF, Vogelzang NJ, Hayes DF, Small EJ, et al. Prognostic significance of plasma vascular endothelial growth factor levels in patients with hormone-refractory prostate cancer treated on Cancer and Leukemia Group B 9480. Clin Cancer Res. 2001;7(7):1932–6. [PubMed] [Google Scholar]
- 36.Gorski DH, Leal AD, Goydos JS. Differential expression of vascular endothelial growth factor-A isoforms at different stages of melanoma progression. J Am Coll Surg. 2003;197(3):408–18. doi: 10.1016/S1072-7515(03)00388-0. [DOI] [PubMed] [Google Scholar]
- 37.Ferrara N. Vascular endothelial growth factor: basic science and clinical progress. Endocr Rev. 2004;25(4):581–611. doi: 10.1210/er.2003-0027. [DOI] [PubMed] [Google Scholar]
- 38.Voelkel NF, Vandivier RW, Tuder RM. Vascular endothelial growth factor in the lung. Am J Physiol Lung Cell Mol Physiol. 2006;290(2):L209–21. doi: 10.1152/ajplung.00185.2005. [DOI] [PubMed] [Google Scholar]
- 39.Wang GL, Semenza GL. Purification and characterization of hypoxia-inducible factor 1. J Biol Chem. 1995;270(3):1230–7. doi: 10.1074/jbc.270.3.1230. [DOI] [PubMed] [Google Scholar]
- 40.Semenza GL. Targeting HIF-1 for cancer therapy. Nat Rev Cancer. 2003;3(10):721–32. doi: 10.1038/nrc1187. [DOI] [PubMed] [Google Scholar]
- 41.Mayerhofer M, Valent P, Sperr WR, Griffin JD, Sillaber C. BCR/ABL induces expression of vascular endothelial growth factor and its transcriptional activator, hypoxia inducible factor-1alpha, through a pathway involving phosphoinositide 3-kinase and the mammalian target of rapamycin. Blood. 2002;100(10):3767–75. doi: 10.1182/blood-2002-01-0109. [DOI] [PubMed] [Google Scholar]
- 42.Mayerhofer M, Aichberger KJ, Florian S, Krauth MT, Hauswirth AW, Derdak S, et al. Identification of mTOR as a novel bifunctional target in chronic myeloid leukemia: dissection of growth-inhibitory and VEGF-suppressive effects of rapamycin in leukemic cells. Faseb J. 2005;19(8):960–2. doi: 10.1096/fj.04-1973fje. [DOI] [PubMed] [Google Scholar]
- 43*.Prager GW, Lackner EM, Krauth MT, Unseld M, Poettler M, Laffer S, et al. Targeting of VEGF-dependent transendothelial migration of cancer cells by bevacizumab. Mol Oncol. 2010;4(2):150–60. doi: 10.1016/j.molonc.2010.01.002. VEGF induces endothelial permeability and tumor cell transmigration in vitro, effects which are mitigated by rapamycin or bevacizumab. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Yeh HH, Lai WW, Chen HH, Liu HS, Su WC. Autocrine IL-6-induced Stat3 activation contributes to the pathogenesis of lung adenocarcinoma and malignant pleural effusion. Oncogene. 2006;25(31):4300–9. doi: 10.1038/sj.onc.1209464. [DOI] [PubMed] [Google Scholar]
- 45.Pritchard-Jones RO, Dunn DB, Qiu Y, Varey AH, Orlando A, Rigby H, et al. Expression of VEGF(xxx)b, the inhibitory isoforms of VEGF, in malignant melanoma. Br J Cancer. 2007;97(2):223–30. doi: 10.1038/sj.bjc.6603839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Robinson CJ, Stringer SE. The splice variants of vascular endothelial growth factor (VEGF) and their receptors. J Cell Sci. 2001;114(Pt 5):853–65. doi: 10.1242/jcs.114.5.853. [DOI] [PubMed] [Google Scholar]
- 47.Soker S, Fidder H, Neufeld G, Klagsbrun M. Characterization of novel vascular endothelial growth factor (VEGF) receptors on tumor cells that bind VEGF165 via its exon 7-encoded domain. J Biol Chem. 1996;271(10):5761–7. doi: 10.1074/jbc.271.10.5761. [DOI] [PubMed] [Google Scholar]
- 48.Soker S, Takashima S, Miao HQ, Neufeld G, Klagsbrun M. Neuropilin-1 is expressed by endothelial and tumor cells as an isoform-specific receptor for vascular endothelial growth factor. Cell. 1998;92(6):735–45. doi: 10.1016/s0092-8674(00)81402-6. [DOI] [PubMed] [Google Scholar]
- 49.Dvorak HF. Vascular permeability factor/vascular endothelial growth factor: a critical cytokine in tumor angiogenesis and a potential target for diagnosis and therapy. J Clin Oncol. 2002;20(21):4368–80. doi: 10.1200/JCO.2002.10.088. [DOI] [PubMed] [Google Scholar]
- 50.Millauer B, Wizigmann-Voos S, Schnurch H, Martinez R, Moller NP, Risau W, et al. High affinity VEGF binding and developmental expression suggest Flk-1 as a major regulator of vasculogenesis and angiogenesis. Cell. 1993;72(6):835–46. doi: 10.1016/0092-8674(93)90573-9. [DOI] [PubMed] [Google Scholar]
- 51.Zeng H, Dvorak HF, Mukhopadhyay D. Vascular permeability factor (VPF)/vascular endothelial growth factor (VEGF) peceptor-1 down-modulates VPF/VEGF receptor-2-mediated endothelial cell proliferation, but not migration, through phosphatidylinositol 3-kinase-dependent pathways. J Biol Chem. 2001;276(29):26969–79. doi: 10.1074/jbc.M103213200. [DOI] [PubMed] [Google Scholar]
- 52.Grove CS, Lee YC. Vascular endothelial growth factor: the key mediator in pleural effusion formation. Curr Opin Pulm Med. 2002;8(4):294–301. doi: 10.1097/00063198-200207000-00009. [DOI] [PubMed] [Google Scholar]
- 53.Cheng D, Lee YC, Rogers JT, Perkett EA, Moyers JP, Rodriguez RM, et al. Vascular endothelial growth factor level correlates with transforming growth factor-beta isoform levels in pleural effusions. Chest. 2000;118(6):1747–53. doi: 10.1378/chest.118.6.1747. [DOI] [PubMed] [Google Scholar]
- 54.Croghan GA, Nichols F, Cassivi S, et al. VEGF A, C, and D levels in malignant pleural effusions. J Clin Oncol. 2008;26(15S) abst. 22126. [Google Scholar]
- 55.Fiorelli A, Vicidomini G, Di Domenico M, Napolitano F, Messina G, Morgillo F, et al. Vascular endothelial growth factor in pleural fluid for differential diagnosis of benign and malignant origin and its clinical applications. Interact Cardiovasc Thorac Surg. 2011;12(3):420–4. doi: 10.1510/icvts.2010.250357. [DOI] [PubMed] [Google Scholar]
- 56.Lieser E, Bradshaw M, Croghan G, Mullon J, Shen KR, Nichols F. Proangiogenesis factors and cell phenotyping in the etiology and diagnosis of malignant pleural effusions. J Thorac Oncol. 2011;6(6):S750–51. [Google Scholar]
- 57.Sack U, Hoffmann M, Zhao XJ, Chan KS, Hui DS, Gosse H, et al. Vascular endothelial growth factor in pleural effusions of different origin. Eur Respir J. 2005;25(4):600–4. doi: 10.1183/09031936.05.00037004. [DOI] [PubMed] [Google Scholar]
- 58.Thickett DR, Armstrong L, Millar AB. Vascular endothelial growth factor (VEGF) in inflammatory and malignant pleural effusions. Thorax. 1999;54(8):707–10. doi: 10.1136/thx.54.8.707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Yanagawa H, Takeuchi E, Suzuki Y, Ohmoto Y, Bando H, Sone S. Vascular endothelial growth factor in malignant pleural effusion associated with lung cancer. Cancer Immunol Immunother. 1999;48(7):396–400. doi: 10.1007/s002620050592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Zebrowski BK, Liu W, Ramirez K, Akagi Y, Mills GB, Ellis LM. Markedly elevated levels of vascular endothelial growth factor in malignant ascites. Ann Surg Oncol. 1999;6(4):373–8. doi: 10.1007/s10434-999-0373-0. [DOI] [PubMed] [Google Scholar]
- 61.Zebrowski BK, Yano S, Liu W, Shaheen RM, Hicklin DJ, Putnam JB, Jr, et al. Vascular endothelial growth factor levels and induction of permeability in malignant pleural effusions. Clin Cancer Res. 1999;5(11):3364–8. [PubMed] [Google Scholar]
- 62.Ishii H, Yazawa T, Sato H, Suzuki T, Ikeda M, Hayashi Y, et al. Enhancement of pleural dissemination and lymph node metastasis of intrathoracic lung cancer cells by vascular endothelial growth factors (VEGFs) Lung Cancer. 2004;45(3):325–37. doi: 10.1016/j.lungcan.2004.02.021. [DOI] [PubMed] [Google Scholar]
- 63.Yano S, Shinohara H, Herbst RS, Kuniyasu H, Bucana CD, Ellis LM, et al. Production of experimental malignant pleural effusions is dependent on invasion of the pleura and expression of vascular endothelial growth factor/vascular permeability factor by human lung cancer cells. Am J Pathol. 2000;157(6):1893–903. doi: 10.1016/S0002-9440(10)64828-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Cui R, Takahashi F, Ohashi R, Yoshioka M, Gu T, Tajima K, et al. Osteopontin is involved in the formation of malignant pleural effusion in lung cancer. Lung Cancer. 2009;63(3):368–74. doi: 10.1016/j.lungcan.2008.06.020. [DOI] [PubMed] [Google Scholar]
- 65.Stathopoulos GT, Psallidas I, Moustaki A, Moschos C, Kollintza A, Karabela S, et al. A central role for tumor-derived monocyte chemoattractant protein-1 in malignant pleural effusion. J Natl Cancer Inst. 2008;100(20):1464–76. doi: 10.1093/jnci/djn325. [DOI] [PubMed] [Google Scholar]
- 66.Stathopoulos GT, Sherrill TP, Karabela SP, Goleniewska K, Kalomenidis I, Roussos C, et al. Host-derived interleukin-5 promotes adenocarcinoma-induced malignant pleural effusion. Am J Respir Crit Care Med. 2010;182(10):1273–81. doi: 10.1164/rccm.201001-0001OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Stathopoulos GT, Kollintza A, Moschos C, Psallidas I, Sherrill TP, Pitsinos EN, et al. Tumor necrosis factor-alpha promotes malignant pleural effusion. Cancer Res. 2007;67(20):9825–34. doi: 10.1158/0008-5472.CAN-07-1064. [DOI] [PubMed] [Google Scholar]
- 68**.Stathopoulos GT, Kalomenidis I, et al. Malignant pleural effusion: tumor-host interactions unleashed. Am J Respir Crit Care Med. 2012;186(6):487–92. doi: 10.1164/rccm.201203-0465PP. Presents a model of malignant pleural effusion pathogenesis integrating the interactions between tumor and host. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Korpanty G, Smyth E, Sullivan LA, Brekken RA, Carney DN. Antiangiogenic therapy in lung cancer: focus on vascular endothelial growth factor pathway. Exp Biol Med (Maywood) 2010;235(1):3–9. doi: 10.1258/ebm.2009.009191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Ferrara N, Kerbel RS. Angiogenesis as a therapeutic target. Nature. 2005;438(7070):967–74. doi: 10.1038/nature04483. [DOI] [PubMed] [Google Scholar]
- 71.Wang Y, Fei D, Vanderlaan M, Song A. Biological activity of bevacizumab, a humanized anti-VEGF antibody in vitro. Angiogenesis. 2004;7(4):335–45. doi: 10.1007/s10456-004-8272-2. [DOI] [PubMed] [Google Scholar]
- 72.Mitchell EP. Targeted Therapy for Metastatic Colorectal Cancer: Role of Aflibercept. Clin Colorectal Cancer. 2012 doi: 10.1016/j.clcc.2012.08.001. [DOI] [PubMed] [Google Scholar]
- 73.Gordon MS, Margolin K, Talpaz M, Sledge GW, Jr, Holmgren E, Benjamin R, et al. Phase I safety and pharmacokinetic study of recombinant human anti-vascular endothelial growth factor in patients with advanced cancer. J Clin Oncol. 2001;19(3):843–50. doi: 10.1200/JCO.2001.19.3.843. [DOI] [PubMed] [Google Scholar]
- 74.Karp JE, Gojo I, Pili R, Gocke CD, Greer J, Guo C, et al. Targeting vascular endothelial growth factor for relapsed and refractory adult acute myelogenous leukemias: therapy with sequential 1-beta-d-arabinofuranosylcytosine, mitoxantrone, and bevacizumab. Clin Cancer Res. 2004;10(11):3577–85. doi: 10.1158/1078-0432.CCR-03-0627. [DOI] [PubMed] [Google Scholar]
- 75.Kazazi-Hyseni F, Beijnen JH, Schellens JH. Bevacizumab. Oncologist. 2010;15(8):819–25. doi: 10.1634/theoncologist.2009-0317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Kraft A, Weindel K, Ochs A, Marth C, Zmija J, Schumacher P, et al. Vascular endothelial growth factor in the sera and effusions of patients with malignant and nonmalignant disease. Cancer. 1999;85(1):178–87. [PubMed] [Google Scholar]
- 77.Strizzi L, Catalano A, Vianale G, Orecchia S, Casalini A, Tassi G, et al. Vascular endothelial growth factor is an autocrine growth factor in human malignant mesothelioma. J Pathol. 2001;193(4):468–75. doi: 10.1002/path.824. [DOI] [PubMed] [Google Scholar]
- 78.Yeo KT, Wang HH, Nagy JA, Sioussat TM, Ledbetter SR, Hoogewerf AJ, et al. Vascular permeability factor (vascular endothelial growth factor) in guinea pig and human tumor and inflammatory effusions. Cancer Res. 1993;53(12):2912–8. [PubMed] [Google Scholar]
- 79.Caine GJ, Lip GY, Blann AD. Platelet-derived VEGF, Flt-1, angiopoietin-1 and P-selectin in breast and prostate cancer: further evidence for a role of platelets in tumour angiogenesis. Ann Med. 2004;36(4):273–7. doi: 10.1080/07853890410026098. [DOI] [PubMed] [Google Scholar]
- 80.Gerber HP, Kowalski J, Sherman D, Eberhard DA, Ferrara N. Complete inhibition of rhabdomyosarcoma xenograft growth and neovascularization requires blockade of both tumor and host vascular endothelial growth factor. Cancer Res. 2000;60(22):6253–8. [PubMed] [Google Scholar]
- 81.Fukumura D, Xavier R, Sugiura T, Chen Y, Park EC, Lu N, et al. Tumor induction of VEGF promoter activity in stromal cells. Cell. 1998;94(6):715–25. doi: 10.1016/s0092-8674(00)81731-6. [DOI] [PubMed] [Google Scholar]
- 82.Tsuzuki Y, Fukumura D, Oosthuyse B, Koike C, Carmeliet P, Jain RK. Vascular endothelial growth factor (VEGF) modulation by targeting hypoxia-inducible factor-1alpha--> hypoxia response element--> VEGF cascade differentially regulates vascular response and growth rate in tumors. Cancer Res. 2000;60(22):6248–52. [PubMed] [Google Scholar]
- 83.Kishimoto J, Ehama R, Ge Y, Kobayashi T, Nishiyama T, Detmar M, et al. In vivo detection of human vascular endothelial growth factor promoter activity in transgenic mouse skin. Am J Pathol. 2000;157(1):103–10. doi: 10.1016/S0002-9440(10)64522-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Gary Lee YC, Melkerneker D, Thompson PJ, Light RW, Lane KB. Transforming growth factor beta induces vascular endothelial growth factor elaboration from pleural mesothelial cells in vivo and in vitro. Am J Respir Crit Care Med. 2002;165(1):88–94. doi: 10.1164/ajrccm.165.1.2104006. [DOI] [PubMed] [Google Scholar]
- 85.Edge SB American Joint Committee on Cancer. AJCC cancer staging manual. 7. New York: Springer; 2010. [DOI] [PubMed] [Google Scholar]
- 86.Heffner JE. Diagnosis and management of malignant pleural effusions. Respirology. 2008;13(1):5–20. doi: 10.1111/j.1440-1843.2007.01154.x. [DOI] [PubMed] [Google Scholar]
- 87.Ryu JS, Ryu ST, Kim YS, Cho JH, Lee HL. What is the clinical significance of transudative malignant pleural effusion? Korean J Intern Med. 2003;18(4):230–3. doi: 10.3904/kjim.2003.18.4.230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Porcel JM, Alvarez M, Salud A, Vives M. Should a cytologic study be ordered in transudative pleural effusions? Chest. 1999;116(6):1836–7. doi: 10.1378/chest.116.6.1836. [DOI] [PubMed] [Google Scholar]
- 89.Light RW, Macgregor MI, Luchsinger PC, Ball WC., Jr Pleural effusions: the diagnostic separation of transudates and exudates. Ann Intern Med. 1972;77(4):507–13. doi: 10.7326/0003-4819-77-4-507. [DOI] [PubMed] [Google Scholar]
- 90.Prakash UBS, Reiman HM. Comparison of Needle-Biopsy with Cytologic Analysis for the Evaluation of Pleural Effusion - Analysis of 414 Cases. Mayo Clinic Proceedings. 1985;60(3):158–64. doi: 10.1016/s0025-6196(12)60212-2. [DOI] [PubMed] [Google Scholar]
- 91.Sahn SA. Malignant pleural effusions. Clin Chest Med. 1985;6(1):113–25. [PubMed] [Google Scholar]
- 92.Escudero Bueno C, Garcia Clemente M, Cuesta Castro B, Molinos Martin L, Rodriguez Ramos S, Gonzalez Panizo A, et al. Cytologic and bacteriologic analysis of fluid and pleural biopsy specimens with Cope’s needle. Study of 414 patients. Arch Intern Med. 1990;150(6):1190–4. doi: 10.1001/archinte.150.6.1190. [DOI] [PubMed] [Google Scholar]
- 93.Abouzgheib W, Bartter T, Dagher H, Pratter M, Klump W. A prospective study of the volume of pleural fluid required for accurate diagnosis of malignant pleural effusion. Chest. 2009;135(4):999–1001. doi: 10.1378/chest.08-2002. [DOI] [PubMed] [Google Scholar]
- 94.Sallach SM, Sallach JA, Vasquez E, Schultz L, Kvale P. Volume of pleural fluid required for diagnosis of pleural malignancy. Chest. 2002;122(6):1913–7. doi: 10.1378/chest.122.6.1913. [DOI] [PubMed] [Google Scholar]
- 95.Domej W, Tilz GP, Foldes-Papp Z, Demel U, Rabold T, Holzer H. Cystatin C of pleural effusion as a novel diagnostic aid in pleural diseases of different aetiologies. Clin Sci (Lond) 2002;102(3):373–80. [PubMed] [Google Scholar]
- 96.Matveychuk A, Rashid G, Fridman Z, Guber A, Shitrit D. Pleural ELFA D-dimer assay: a surrogate marker for malignant pleural effusion. Thromb Res. 2012;129(5):648–51. doi: 10.1016/j.thromres.2011.07.036. [DOI] [PubMed] [Google Scholar]
- 97.Pernemalm M, De Petris L, Eriksson H, Branden E, Koyi H, Kanter L, et al. Use of narrow-range peptide IEF to improve detection of lung adenocarcinoma markers in plasma and pleural effusion. Proteomics. 2009;9(13):3414–24. doi: 10.1002/pmic.200800814. [DOI] [PubMed] [Google Scholar]
- 98.Shijubo N, Honda Y, Fujishima T, Takahashi H, Kodama T, Kuroki Y, et al. Lung surfactant protein-A and carcinoembryonic antigen in pleural effusions due to lung adenocarcinoma and malignant mesothelioma. Eur Respir J. 1995;8(3):403–6. doi: 10.1183/09031936.95.08030403. [DOI] [PubMed] [Google Scholar]
- 99.Bard MP, Hegmans JP, Hemmes A, Luider TM, Willemsen R, Severijnen LA, et al. Proteomic analysis of exosomes isolated from human malignant pleural effusions. Am J Respir Cell Mol Biol. 2004;31(1):114–21. doi: 10.1165/rcmb.2003-0238OC. [DOI] [PubMed] [Google Scholar]
- 100.Hsieh WY, Chen MW, Ho HT, You TM, Lu YT. Identification of differentially expressed proteins in human malignant pleural effusions. Eur Respir J. 2006;28(6):1178–85. doi: 10.1183/09031936.06.00135405. [DOI] [PubMed] [Google Scholar]
- 101.Rodriguez-Pineiro AM, Blanco-Prieto S, Sanchez-Otero N, Rodriguez-Berrocal FJ, de la Cadena MP. On the identification of biomarkers for non-small cell lung cancer in serum and pleural effusion. J Proteomics. 2010;73(8):1511–22. doi: 10.1016/j.jprot.2010.03.005. [DOI] [PubMed] [Google Scholar]
- 102.Botana-Rial M, Casado-Rey P, Leiro-Fernandez V, Andrade-Olivie M, Represas-Represas C, Fernandez-Villar A. Validity of procalcitonin and C-reactive protein measurement when differentiating between benign and malignant pleural effusion. Clin Lab. 2011;57(5-6):373–8. [PubMed] [Google Scholar]
- 103.Cheng D, Rodriguez RM, Perkett EA, Rogers J, Bienvenu G, Lappalainen U, et al. Vascular endothelial growth factor in pleural fluid. Chest. 1999;116(3):760–5. doi: 10.1378/chest.116.3.760. [DOI] [PubMed] [Google Scholar]
- 104.Yu CJ, Wang CL, Wang CI, Chen CD, Dan YM, Wu CC, et al. Comprehensive proteome analysis of malignant pleural effusion for lung cancer biomarker discovery by using multidimensional protein identification technology. J Proteome Res. 2011;10(10):4671–82. doi: 10.1021/pr2004743. [DOI] [PubMed] [Google Scholar]
- 105.Shen YC, Liu MQ, Wan C, Chen L, Wang T, Wen FQ. Diagnostic accuracy of vascular endothelial growth factor for malignant pleural effusion: A meta-analysis. Exp Ther Med. 2012;3(6):1072–6. doi: 10.3892/etm.2012.514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Bielsa S, Martin-Juan J, Porcel JM, Rodriguez-Panadero F. Diagnostic and prognostic implications of pleural adhesions in malignant effusions. J Thorac Oncol. 2008;3(11):1251–6. doi: 10.1097/JTO.0b013e318189f53d. [DOI] [PubMed] [Google Scholar]
- 107.Bernard A, de Dompsure RB, Hagry O, Favre JP. Early and late mortality after pleurodesis for malignant pleural effusion. Ann Thorac Surg. 2002;74(1):213–7. doi: 10.1016/s0003-4975(02)03599-3. [DOI] [PubMed] [Google Scholar]
- 108.Burrows CM, Mathews WC, Colt HG. Predicting survival in patients with recurrent symptomatic malignant pleural effusions: an assessment of the prognostic values of physiologic, morphologic, and quality of life measures of extent of disease. Chest. 2000;117(1):73–8. doi: 10.1378/chest.117.1.73. [DOI] [PubMed] [Google Scholar]
- 109.Dresler CM, Olak J, Herndon JE, 2nd, Richards WG, Scalzetti E, Fleishman SB, et al. Phase III intergroup study of talc poudrage vs talc slurry sclerosis for malignant pleural effusion. Chest. 2005;127(3):909–15. doi: 10.1378/chest.127.3.909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Schulze M, Boehle AS, Kurdow R, Dohrmann P, Henne-Bruns D. Effective treatment of malignant pleural effusion by minimal invasive thoracic surgery: thoracoscopic talc pleurodesis and pleuroperitoneal shunts in 101 patients. Ann Thorac Surg. 2001;71(6):1809–12. doi: 10.1016/s0003-4975(01)02586-3. [DOI] [PubMed] [Google Scholar]
- 111.Rodriguez-Panadero F, Lopez-Mejias J. Survival time of patients with pleural metastatic carcinoma predicted by glucose and pH studies. Chest. 1989;95(2):320–4. doi: 10.1378/chest.95.2.320. [DOI] [PubMed] [Google Scholar]
- 112.Sahn SA, Good JT., Jr Pleural fluid pH in malignant effusions. Diagnostic, prognostic, and therapeutic implications. Ann Intern Med. 1988;108(3):345–9. doi: 10.7326/0003-4819-108-3-345. [DOI] [PubMed] [Google Scholar]
- 113.Sanchez-Armengol A, Rodriguez-Panadero F. Survival and talc pleurodesis in metastatic pleural carcinoma, revisited. Report of 125 cases. Chest. 1993;104(5):1482–5. doi: 10.1378/chest.104.5.1482. [DOI] [PubMed] [Google Scholar]
- 114.Love D, White D, Kiroff G. Thoracoscopic talc pleurodesis for malignant pleural effusion. ANZ J Surg. 2003;73(1-2):19–22. doi: 10.1046/j.1445-2197.2003.02616.x. [DOI] [PubMed] [Google Scholar]
- 115.Viallat JR, Rey F, Astoul P, Boutin C. Thoracoscopic talc poudrage pleurodesis for malignant effusions. A review of 360 cases. Chest. 1996;110(6):1387–93. doi: 10.1378/chest.110.6.1387. [DOI] [PubMed] [Google Scholar]
- 116.Hirayama N, Tabata C, Tabata R, Maeda R, Yasumitsu A, Yamada S, et al. Pleural effusion VEGF levels as a prognostic factor of malignant pleural mesothelioma. Respir Med. 2011;105(1):137–42. doi: 10.1016/j.rmed.2010.10.010. [DOI] [PubMed] [Google Scholar]
- 117.Davies HE, Steer H, Fysh E, et al. Diagnosis and management of malignant pleural effusions. Minerva Pneumonol. 2010;49:237–52. [Google Scholar]
- 118.Shaw P, Agarwal R. Pleurodesis for malignant pleural effusions. Cochrane Database Syst Rev. 2004;(1) doi: 10.1002/14651858.CD002916.pub2. CD002916. [DOI] [PubMed] [Google Scholar]
- 119.Burgers JA, Kunst PW, Koolen MG, Willems LN, Burgers JS, van den Heuvel M. Pleural drainage and pleurodesis: implementation of guidelines in four hospitals. Eur Respir J. 2008;32(5):1321–7. doi: 10.1183/09031936.00165607. [DOI] [PubMed] [Google Scholar]
- 120.Demmy TL, Gu L, Burkhalter JE, Toloza EM, D’Amico TA, Sutherland S, et al. Optimal management of malignant pleural effusions (results of CALGB 30102) J Natl Compr Canc Netw. 2012;10(8):975–82. doi: 10.6004/jnccn.2012.0102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Olden AM, Holloway R. Treatment of malignant pleural effusion: PleuRx catheter or talc pleurodesis? A cost-effectiveness analysis. J Palliat Med. 2010;13(1):59–65. doi: 10.1089/jpm.2009.0220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Suzuki K, Servais EL, Rizk NP, Solomon SB, Sima CS, Park BJ, et al. Palliation and pleurodesis in malignant pleural effusion: the role for tunneled pleural catheters. J Thorac Oncol. 2011;6(4):762–7. doi: 10.1097/JTO.0b013e31820d614f. [DOI] [PubMed] [Google Scholar]
- 123.Van Meter ME, McKee KY, Kohlwes RJ. Efficacy and safety of tunneled pleural catheters in adults with malignant pleural effusions: a systematic review. J Gen Intern Med. 2011;26(1):70–6. doi: 10.1007/s11606-010-1472-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Tremblay A, Michaud G. Single-center experience with 250 tunnelled pleural catheter insertions for malignant pleural effusion. Chest. 2006;129(2):362–8. doi: 10.1378/chest.129.2.362. [DOI] [PubMed] [Google Scholar]
- 125.Bogliolo GV, Lerza R, Bottino GB, Mencoboni MP, Pannacciulli IM, Vannozzi M, et al. Regional pharmacokinetic selectivity of intrapleural cisplatin. Eur J Cancer. 1991;27(7):839–42. doi: 10.1016/0277-5379(91)90129-2. [DOI] [PubMed] [Google Scholar]
- 126.Figlin R, Mendoza E, Piantadosi S, Rusch V. Intrapleural chemotherapy without pleurodesis for malignant pleural effusions. LCSG Trial 861. Chest. 1994;106(6 Suppl):363S–6S. [PubMed] [Google Scholar]
- 127.Jones DR, Taylor MD, Petroni GR, Shu J, Burks SG, Daniel TM, et al. Phase I trial of intrapleural docetaxel administered through an implantable catheter in subjects with a malignant pleural effusion. J Thorac Oncol. 2010;5(1):75–81. doi: 10.1097/JTO.0b013e3181c07ddc. [DOI] [PubMed] [Google Scholar]
- 128.Lerza R, Vannozzi MO, Tolino G, Viale M, Bottino GB, Bogliolo G, et al. Carboplatin and cisplatin pharmacokinetics after intrapleural combination treatment in patients with malignant pleural effusion. Ann Oncol. 1997;8(4):385–91. doi: 10.1023/a:1008203100410. [DOI] [PubMed] [Google Scholar]
- 129.Masuno T, Kishimoto S, Ogura T, Honma T, Niitani H, Fukuoka M, et al. A comparative trial of LC9018 plus doxorubicin and doxorubicin alone for the treatment of malignant pleural effusion secondary to lung cancer. Cancer. 1991;68(7):1495–500. doi: 10.1002/1097-0142(19911001)68:7<1495::aid-cncr2820680705>3.0.co;2-6. [DOI] [PubMed] [Google Scholar]
- 130.Muraoka M, Oka T, Akamine S, Tagawa T, Morinaga M, Inoue M, et al. Modified intrapleural cisplatin treatment for lung cancer with positive pleural lavage cytology or malignant effusion. J Surg Oncol. 2006;93(4):323–9. doi: 10.1002/jso.20470. [DOI] [PubMed] [Google Scholar]
- 131.Perng RP, Wu MF, Lin SY, Chen YM, Lin JY, Whang-Peng J. A phase I feasibility and pharmacokinetic study of intrapleural paclitaxel in patients with malignant pleural effusions. Anticancer Drugs. 1997;8(6):565–73. doi: 10.1097/00001813-199707000-00003. [DOI] [PubMed] [Google Scholar]
- 132.Shoji T, Tanaka F, Yanagihara K, Inui K, Wada H. Phase II study of repeated intrapleural chemotherapy using implantable access system for management of malignant pleural effusion. Chest. 2002;121(3):821–4. doi: 10.1378/chest.121.3.821. [DOI] [PubMed] [Google Scholar]
- 133.Tohda Y, Iwanaga T, Takada M, Yana T, Kawahara M, Negoro S, et al. Intrapleural administration of cisplatin and etoposide to treat malignant pleural effusions in patients with non-small cell lung cancer. Chemotherapy. 1999;45(3):197–204. doi: 10.1159/000007183. [DOI] [PubMed] [Google Scholar]
- 134.Wang X, Zhou J, Wang Y, Zhu Z, Lu Y, Wei Y, et al. A phase I clinical and pharmacokinetic study of paclitaxel liposome infused in non-small cell lung cancer patients with malignant pleural effusions. Eur J Cancer. 2010;46(8):1474–80. doi: 10.1016/j.ejca.2010.02.002. [DOI] [PubMed] [Google Scholar]
- 135.Yoshida K, Sugiura T, Takifuji N, Kawahara M, Matsui K, Kudoh S, et al. Randomized phase II trial of three intrapleural therapy regimens for the management of malignant pleural effusion in previously untreated non-small cell lung cancer: JCOG 9515. Lung Cancer. 2007;58(3):362–8. doi: 10.1016/j.lungcan.2007.07.009. [DOI] [PubMed] [Google Scholar]
- 136.Sterman DH, Recio A, Haas AR, Vachani A, Katz SI, Gillespie CT, et al. A phase I trial of repeated intrapleural adenoviral-mediated interferon-beta gene transfer for mesothelioma and metastatic pleural effusions. Mol Ther. 2010;18(4):852–60. doi: 10.1038/mt.2009.309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Sterman DH, Recio A, Carroll RG, Gillespie CT, Haas A, Vachani A, et al. A phase I clinical trial of single-dose intrapleural IFN-beta gene transfer for malignant pleural mesothelioma and metastatic pleural effusions: high rate of antitumor immune responses. Clin Cancer Res. 2007;13(15 Pt 1):4456–66. doi: 10.1158/1078-0432.CCR-07-0403. [DOI] [PubMed] [Google Scholar]
- 138.Ren S, Terman DS, Bohach G, Silvers A, Hansen C, Colt H, et al. Intrapleural staphylococcal superantigen induces resolution of malignant pleural effusions and a survival benefit in non-small cell lung cancer. Chest. 2004;126(5):1529–39. doi: 10.1378/chest.126.5.1529. [DOI] [PubMed] [Google Scholar]
- 139.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(4):1249–56. doi: 10.1016/S0002-9440(10)65669-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Hu L, Hofmann J, Zaloudek C, Ferrara N, Hamilton T, Jaffe RB. Vascular endothelial growth factor immunoneutralization plus Paclitaxel markedly reduces tumor burden and ascites in athymic mouse model of ovarian cancer. Am J Pathol. 2002;161(5):1917–24. doi: 10.1016/S0002-9440(10)64467-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Gossmann A, Helbich TH, Mesiano S, Shames DM, Wendland MF, Roberts TP, et al. 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. 2000;183(4):956–63. doi: 10.1067/mob.2000.107092. [DOI] [PubMed] [Google Scholar]
- 142.Watanabe M, Boyer JL, Crystal RG. AAVrh.10-mediated genetic delivery of bevacizumab to the pleura to provide local anti-VEGF to suppress growth of metastatic lung tumors. Gene Ther. 2010;17(8):1042–51. doi: 10.1038/gt.2010.87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Luo JC, Toyoda M, Shibuya M. Differential inhibition of fluid accumulation and tumor growth in two mouse ascites tumors by an antivascular endothelial growth factor/permeability factor neutralizing antibody. Cancer Res. 1998;58(12):2594–600. [PubMed] [Google Scholar]
- 144.Shibuya M, Luo JC, Toyoda M, Yamaguchi S. Involvement of VEGF and its receptors in ascites tumor formation. Cancer Chemother Pharmacol. 1999;43(Suppl):S72–7. doi: 10.1007/s002800051102. [DOI] [PubMed] [Google Scholar]
- 145.Shaheen RM, Ahmad SA, Liu W, Reinmuth N, Jung YD, Tseng WW, et al. Inhibited growth of colon cancer carcinomatosis by antibodies to vascular endothelial and epidermal growth factor receptors. Br J Cancer. 2001;85(4):584–9. doi: 10.1054/bjoc.2001.1936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Yano S, Herbst RS, Shinohara H, Knighton B, Bucana CD, Killion JJ, et al. Treatment for malignant pleural effusion of human lung adenocarcinoma by inhibition of vascular endothelial growth factor receptor tyrosine kinase phosphorylation. Clin Cancer Res. 2000;6(3):957–65. [PubMed] [Google Scholar]
- 147.Ribeiro SC, Vargas FS, Antonangelo L, Marchi E, Genofre EH, Acencio MM, et al. Monoclonal anti-vascular endothelial growth factor antibody reduces fluid volume in an experimental model of inflammatory pleural effusion. Respirology. 2009;14(8):1188–93. doi: 10.1111/j.1440-1843.2009.01628.x. [DOI] [PubMed] [Google Scholar]
- 148.Teixeira LR, Vargas FS, Acencio MM, Ribeiro SC, Sales RK, Antonangelo L, et al. Blockage of vascular endothelial growth factor (VEGF) reduces experimental pleurodesis. Lung Cancer. 2011 doi: 10.1016/j.lungcan.2011.04.015. [DOI] [PubMed] [Google Scholar]
- 149.Guo YB, Kalomenidis I, Hawthorne M, Parman KS, Lane KB, Light RW. Pleurodesis is inhibited by anti-vascular endothelial growth factor antibody. Chest. 2005;128(3):1790–7. doi: 10.1378/chest.128.3.1790. [DOI] [PubMed] [Google Scholar]
- 150.Pichelmayer O, Zielinski C, Raderer M. Response of a nonmalignant pleural effusion to bevacizumab. N Engl J Med. 2005;353(7):740–1. doi: 10.1056/NEJM200508183530721. [DOI] [PubMed] [Google Scholar]
- 151.Pichelmayer O, Gruenberger B, Zielinski C, Raderer M. Bevacizumab is active in malignant effusion. Ann Oncol. 2006;17(12):1853. doi: 10.1093/annonc/mdl143. [DOI] [PubMed] [Google Scholar]
- 152.Kitamura K, Kubota K, Ando M, Takahashi S, Nishijima N, Sugano T, et al. Bevacizumab plus chemotherapy for advanced non-squamous non-small-cell lung cancer with malignant pleural effusion. Cancer Chemother Pharmacol. 2013;71(2):457–61. doi: 10.1007/s00280-012-2026-4. [DOI] [PubMed] [Google Scholar]
- 153.Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A. 2003;100(7):3983–8. doi: 10.1073/pnas.0530291100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Bae SH, Hwang JY, Kim WJ, Yoon HH, Kim JM, Nam YH, et al. A case of cardiac amyloidosis with diuretic-refractory pleural effusions treated with bevacizumab. Korean Circ J. 2010;40(12):671–6. doi: 10.4070/kcj.2010.40.12.671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Kesterson JP, Mhawech-Fauceglia P, Lele S. The use of bevacizumab in refractory ovarian granulosa-cell carcinoma with symptomatic relief of ascites: a case report. Gynecol Oncol. 2008;111(3):527–9. doi: 10.1016/j.ygyno.2008.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Hamilton CA, Maxwell GL, Chernofsky MR, Bernstein SA, Farley JH, Rose GS. Intraperitoneal bevacizumab for the palliation of malignant ascites in refractory ovarian cancer. Gynecol Oncol. 2008;111(3):530–2. doi: 10.1016/j.ygyno.2008.04.028. [DOI] [PubMed] [Google Scholar]
- 157.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(18 Suppl):9043. [Google Scholar]