With the exception of hematological malignancies, flow cytometry (FC) is infrequently applied as an ancillary tool in the diagnosis of malignant effusions in most institutions. However, FC may be effectively used to differentiate between epithelial cells, mesothelial cells and leukocytes using antibodies against both cell surface and intracellular proteins, offering the advantage of quantitative analysis. Additionally, FC may be applied to the quantitative detection of cancer-associated molecules, including stem cell markers, as well as assessment of critical cellular processes, such as proliferation and apoptosis. Some of the latter tests may have relevance for monitoring treatment response in the presence of metastatic disease, although this does not constitute routine practice to date. This review summarizes current knowledge regarding the application of FC to serous effusions in the diagnostic setting, as well as in research into cancer biology focusing on clinical specimens. The studies published to date suggest a role for this method in the clinical setting in the context of diagnosis, prediction and prognosis.

The serosal cavities are frequently affected by malignancy, most commonly in the form of metastatic disease originating from the lung, breast, female genital tract or gastrointestinal tract. Malignant mesothelioma (MM), the native tumor of this anatomic site, is an important differential diagnosis. Differentiating carcinoma cells from inflammatory cells (particularly macrophages) and reactive or malignant mesothelial cells based on morphology alone can be difficult. Immunohistochemistry (IHC) has in recent years become the most widely used ancillary method in effusion diagnosis and allows for the correct classification of the majority of tumors [1]. The use of cell blocks enables the application of protocols identical to the ones used in surgical pathology.

Flow cytometry (FC) is widely applied in the diagnosis of hematological disorders, but has been infrequently applied in the characterization of epithelial and mesothelial cells in the diagnostic setting. FC has the advantage of being a quantitative method that may be rapidly applied to viable cells in fresh specimens, thereby avoiding fixation artifacts. FC reliably detects cytoplasmic, nuclear and surface antigens, and multicolor FC allows for the assessment of multiple antigens simultaneously. Recent years have seen an expansion in the use of FC for research purposes, as it provides an accurate assessment of multiple biological processes. As effusions constitute metastatic disease that requires treatment by chemotherapy and/or targeted therapy, FC is of evident potential as an objective tool to assess predictive and prognostic endpoints.

The present review presents data from studies performed in the last 20 years in which FC has been applied alone or with other methods in both the diagnostic and research settings, the latter with the potential to advance treatment. Studies of hematological cancers are not discussed in this overview.

Earlier studies investigated the role of DNA ploidy by FC in the diagnosis of malignancy on effusions. These studies generally reported good sensitivity and specificity of this assay in differentiating benign from malignant effusions [2,3,4,5,6,7]. Some authors reported that image cytometry performed better than FC in this context [2], while others had the opposite experience [4]. Irrespective of this, the role of this method in effusion cytology has been limited in recent years.

The author's group applied FC as a diagnostic tool in several studies. In two of these, a panel consisting of Ber-EP4, N-Cadherin, CD45 and CD14 effectively identified carcinoma cells, mesothelial cells, leukocytes and macrophages, respectively [8,9]. A panel including Ber-EP4, CD45 and CD14 was used by Pillai et al. [10] in a study of 83 effusions, with sensitivity and specificity at 88.15 and 97.64% for detecting malignancy, respectively, compared to 73.68 and 100% for morphology.

A third study by the author included reactive and malignant effusions, the latter including both carcinomas and MM specimens, using the 4 abovementioned markers and an antiepithelial membrane antigen antibody, detecting the same protein as MUC1 antibodies, as well as 3 nonconjugated antibodies against carbohydrate/glycoprotein moieties on the cell surface of tumor cells B72.3, the AH6 antibody against the Lewisy antigen, and the HB-Tn antibody against the Tn carbohydrate antigen. Carcinoma cells expressed 3 of the 4 epithelial markers (Ber-EP4, B72.3, AH6 and HB-Tn) in 77% of cases compared to 7% of reactive specimens, and cells positive for all 4 markers were found in 39% of carcinoma effusions versus none in reactive effusions [11].

Hartman et al. [12] applied the 6E6 antibody against the MUC1/Y protein to ovarian and breast carcinoma effusions and reported a high sensitivity in detecting tumor cells. Several other proteins have been assessed for their diagnostic role by FC in effusions. Kentrou et al. [13] analyzed 125 effusions suspected of being malignant using a two-phase analysis applying a large panel. Specimens containing cells negative for CD45 and positive for CD71 (transferring receptor), suspected of being malignant, were analyzed for the expression of Ber-EP4, antiepithelial membrane antigen, desmin (expressed in reactive mesothelial cells), the myeloid markers CD66 and CD56 (NCAM), as well as ploidy. FC and conventional cytology had a sensitivity of 85.1 and 93.2%, and specificity of 97.8 and 95.6%, respectively.

In another study, a panel including CD45 and CD326, the latter as a tumor marker, was applied to 506 ascites and peritoneal washing specimens from 333 patients, and a high tumor-to-leukocyte ratio using these markers was significantly associated with poor survival [14].

The detection of EpCAM in microvesicles by FC was recently reported to be useful in diagnosing malignant pleural effusions in the analysis of 71 malignant and 14 benign specimens, and its addition to morphology was superior to morphology alone [15].

Folate receptor-α was shown to be a sensitive marker for ovarian carcinoma in a study combining an antibody against this protein with Ber-EP4 [16]. The FOLR1 gene, encoding for this protein, was found to be overexpressed in ovarian serous carcinoma compared to peritoneal MM in a gene expression array analysis [17], and folate receptor-α expression by FC was subsequently found to be useful in differentiating ovarian and breast carcinoma from MM in an analysis of 91 effusions [18]. PINCH-2, a protein with a cellular role in adhesion and signaling, was not differentially expressed in these 3 tumor types [19], despite overexpression of the gene in peritoneal MM in the abovementioned gene expression array analysis.

In recent years, FC has been applied to studies related to different aspects of tumor biology, including to analyses of the host response to cancer. These studies are discussed below.

The Immune Response

The host immune response to cancer is often ineffective or even tumor promoting and the presence of leukocytes does not necessarily imply an improved outcome. Characterization of leukocyte subclasses that modulate the immune response to cancer has been extensively studied in recent years in practically every cancer type. FC is highly informative in this respect as it provides a quantitative analysis of these cell populations.

The diagnostic value of specific leukocyte populations in effusions was assessed in 2 studies. Cornfield and Gheith [20] compared natural killer (NK) and T-cell parameters in 30 benign and 30 malignant effusions. CD16+/CD56+ NK cell counts were higher in malignant effusions, but the difference was only weakly significant (p = 0.04). T cells carrying α/β T-cell receptor rearrangement were far more numerous than those expressing T-cell receptor γ/δ in both specimens. In contrast, Wang et al. [21] found significantly higher numbers of CD14+/CD163+ tumor-infiltrating macrophages, considered to be tumor promoting, in malignant compared to benign effusions, with a cut-off at 3.65% of the macrophage population associated with sensitivity and specificity at 81.2 and 100%, respectively.

In another comparative study, effector memory CD8+ T cells were found to be present in significantly higher levels in blood and pleural fluid from healthy controls compared to patients with malignant pleural effusion, pleural metastases or benign asbestos-related lesions [22]. Breast carcinoma specimens, including malignant pleural effusions, were shown to contain nonconventional double-positive T cells expressing both CD4 and CD8 that produced IL-5 and IL-13 [23].

Leukocyte counts in effusions were shown to be associated with patient survival. Cytokine array analysis showed that ascites specimens in ovarian carcinoma have a suppressive immune environment. A low CD4/CD8 ratio, observed in specimens from 13 patients, was associated with longer survival [24]. In an analysis of 73 ovarian carcinoma effusions for the expression of leukocyte markers (CD3, CD4, CD8, CD4/CD8 ratio, CD16, CD19, and CD14), the presence of CD16-positive NK cells and more advanced (FIGO stage IV) disease was associated with a shorter overall survival (OS). Additionally, a higher percentage of CD19-positive B cells and stage IV disease were markers of poor survival for patients with postchemotherapy effusions [25].

The expression and clinical role of regulatory T cells (Tregs) have been investigated in several studies. The fraction of CD4+/CD25high/Foxp3+ regulatory T lymphocytes, which suppress the immune response, among CD4+ cells was increased in cultured cells obtained from ovarian carcinoma cells at diagnosis and at disease recurrence following expansion by interleukin-2 (IL-2), and was more pronounced in the latter group [26].

Tregs (CD4+/CD25+/CD127low/-) were found in ascites from gastric cancer patients, as well as at other tumor sites, and higher Foxp3 expression in these cells was associated with a more advanced TNM stage [27]. In another study [28],lymphocytes from lung cancer patients with malignant pleural effusions contained a higher number of CD4+/CD25+ cells strongly expressing Foxp3 and CTLA4 compared to pleural lavage specimens from patients with lung cancer but no effusion.

The chemokine CCL22 was overexpressed in malignant pleural effusion specimens from lung cancer patients compared to corresponding serum, and was shown to promote the influx of CD4+/CD25+ Tregs into the pleural cavity [29]. A higher ratio of Tregs/T helper IL-17-producing cells (Th17) by FC was found in malignant (n = 26) compared to parapneumonic (n = 12) pleural effusions and a higher ratio was associated with significantly shorter survival [30]. Conversely, another group reported that Th17 cells are increased whereas Tregs are decreased in effusions from patients with cancer and tuberculosis compared to benign specimens from patients with other conditions [31].

Chemokine receptor expression was analyzed in several studies using FC. Milliken et al. [32] found the frequent expression of CCR1, CCR2 and CCR5 on macrophages in ascites specimens from ovarian carcinoma patients. CCR1 was expressed by >60% of all T cells, whereas CCR2 and CCR5 expression was more frequent on CD4+ than CD8+ T cells.

Analysis of the expression of 5 chemokine receptors (CXCR1, CXCR4, CCR2, CCR5 and CCR7) by FC in ovarian carcinoma, breast carcinoma and MM effusions using FC showed the frequent expression of these proteins on leukocytes, while all receptors were rarely expressed on cancer cells. Chemokine receptor expression on lymphocytes and monocytes from ovarian carcinoma effusions had no prognostic role [25,33,34].

Analysis of 44 effusions constituting 27 malignant specimens and 17 specimens from patients with cirrhosis showed that malignant specimens were enriched for T cells with a ‘naïve phenotype' (CD62L+ and CD45RA+CCR7+), ‘central memory' (CD45RA-CCR7+) and type 2-polarized (CCR4+) cells, while being deficient in ‘effector'-type (CD45RA-CCR7+ or CD45RA+CCR7-) and presumably type 1-polarized T cells (CCR5+), as well as potentially cytotoxic CD56+ NK cells [35]. Macrophages isolated from malignant and reactive effusions were shown to have a different antigenic profile, with CCR7 expression being significantly higher in nonmalignant effusions [36].

Adhesion Molecules and Other Cell Membrane Proteins

FC has been applied to several studies of surface molecules involved in cell-cell adhesion or interaction between cells and the extracellular matrix. Integrins, a large family of proteins containing α- and β-subunits, have been the focus of several studies by the author's group. In the first two studies, frequent expression of the αv, α6 and β1 integrin subunits was observed at the protein and mRNA level in ovarian carcinoma effusions [37,38]. The absence of α6 integrin subunit protein expression using FC was associated with shorter OS, though not significantly [38]. The third study investigated whether these receptors are differentially expressed in carcinomas of various origins, MM and reactive mesothelial cells [39]. Analysis of 67 effusions (48 carcinomas, 7 MM and 12 RM specimens) for αv, α6, β1 and β3 integrin subunit expression using FC showed frequent expression of the αv, α6, and β1 subunits, with a significantly higher expression of the α6 subunit in MM. Expression of the β3 integrin subunit was not found in any of the specimens in our assay.

Quantitative analysis of two CD44 splice variants in gastric carcinoma showed low expression of the v6 variant, whereas expression of the v9 variant was high in carcinoma cells in ascites specimens [40]. Analysis of CD44 and its variants v4/5 and v6 in ascites specimens from patients with gynecological carcinomas showed variant expression in vesicles constituting membrane fragments [41].

Expression of matrix metalloproteinases (MMP-2, MMP-7, MMP-9, MMP-14/MT1-MMP) and their inhibitors TIMP-2 and TIMP-4, the latter family required for MMP activation, was observed on the cell surface of smooth muscle actin-positive cells, identified as tumor-associated myofibroblasts, in effusions from gastric cancer patients [42].

Analysis of Her2 expression in ovarian carcinoma specimens from multiple anatomic sites, including effusions, as well as short-term cultures, showed that this method was superior to IHC in terms of sensitivity in detecting this protein [43].

MM cells from 9 effusion specimens were shown to be consistently positive for mesothelin. However, upon short-term culturing the expression of this protein was variably lost, as assessed by IHC and FC. Tumors with high expression of this protein were susceptible to the antimesothelin immunotoxin SS1P [44].

Cancer Stem Cells

Cancer stem cells (CSC) are increasingly regarded as a chemoresistant cell population that is the cause of failure of antitumor therapy and cancer-related death. FC is ideal for identifying cell populations expressing postulated CSC markers, and has been used in this context in several studies.

Meng et al. [45] identified CSC by the high aldehyde dehydrogenase (ALDHhigh) phenotype by FC and the presence of these cells was associated with shorter progression-free survival (PFS) compared to patients with specimens that had an ALDHlow profile. A higher expression of ALDH1A1 was observed in ovarian carcinoma effusions compared to solid tumors, which decreased after treatment with niclosamide [46].

The detection of a side population by FC is another widely used method for isolating CSC, as in the study of ovarian carcinoma effusions by Hu et al. [47]. Yao et al. [48] analyzed the expression of 35 cancer-associated markers, including CSC markers, by FC in 12 malignant and 8 benign pleural effusions. EpCAM+ tumor cells were tumorigenic in mice and had a high CD24 expression.

The cell surface proteoglycan chondroitin sulfate proteoglycan 4 (CSPG4) was detected on breast carcinoma cells from primary carcinomas and malignant pleural effusions, and in the latter specimens was observed on CSC with a CD44+/CD24low/- profile by FC [49].

Apoptosis

FC is a highly valuable method for analyzing apoptosis, since it allows for the simultaneous detection and quantification of several apoptosis parameters, thus ensuring the reliability of the obtained results. The author's group published a protocol for the measurement of apoptosis on carcinoma cells in effusions. Cells were identified by Ber-EP4 and EpCAM expression and the absence of CD45, and apoptosis was quantified by FC using the intracellular/nuclear markers cleaved caspase-3, cleaved caspase-8 and incorporated dUTP [50].

A subsequent study of 76 ovarian carcinoma effusions analyzed the expression and clinical role of cleaved caspases and dUTP incorporation by FC. Caspase cleavage and dUTP incorporation were found in <10% of cells in the majority of effusions, with no significant difference between pre- and postchemotherapy effusions, suggesting little apoptosis occurs in tumor cells. Cleaved caspase-3 and cleaved caspase-8 levels, as well as the levels of both cleaved caspases and dUTP incorporation, were directly related. Higher than median cleaved caspase-3 levels correlated with a longer OS and PFS [51].

Exposure of phosphatidylserine at the cell surface, detected by annexin-V, is widely used as an assay for detecting apoptosis, but in fact occurs on stressed cells in general. Analysis of annexin-V expression in ovarian carcinoma effusions (n = 76) using FC showed a significantly higher percentage of cells expressing this marker compared to the previously studied cleaved caspases and dUTP incorporation. Additionally, a higher percentage of annexin-V-expressing cells in postchemotherapy specimens was significantly associated with poor OS and PFS, suggesting it is a survival rather than apoptosis marker [52]. Annexin-V assay measurement similarly correlated poorly with cleaved caspases and dUTP incorporation in a subsequent study of nonovarian malignant effusions and reactive effusions [53].

Expression of the antiapoptotic protein cellular FLICE-inhibitory protein (c-FLIP) was universally seen in the analysis of 69 ovarian carcinoma effusions using FC. c-FLIP expression was unrelated to clinicopathologic parameters, including chemoresponse and survival, but was significantly inversely related to that of cleaved caspase-3 [54].

Death receptor family members are most often regarded as proapoptotic markers, but mediate both apoptosis and survival in cancer cells. Analysis of 95 ovarian carcinoma effusions and a smaller series of breast carcinoma effusions for DR4, DR5, Fas, and the tumor necrosis factor (TNF) receptors TNFR1 and TNFR2 expression using FC showed DR4, DR5 and Fas expression on tumor cells in the majority of specimens, with less frequent expression of TNFR1 and TNFR2. Unexpectedly, higher DR4, DR5 and Fas expression was significantly related to a poor chemoresponse, and DR4 expression was associated with poor OS and PFS in both univariate and multivariate survival analysis [55].

In agreement with this observation, Koyama et al. [56] found frequent expression of DR4, DR5 and the decoy receptor DcR2 on gastric carcinoma in both the primary tumor and metastatic cells in effusions, with little apoptosis detected in these cells. Another TNFR family member, CD40, was reported to be frequently expressed on ovarian carcinoma cells and its activation by an agonist antibody in vitro resulted in reduced cell growth and increased apoptosis [57].

Other Molecules

Few additional studies have analyzed the expression of various cancer-associated molecules in effusions using FC. Equilibrative and concentrative nucleoside transporters (ENTs and CNTs) mediate the cellular uptake of nucleosides used as anticancer agents. Analysis of ENT1, ENT2, ENT4 and CNT3 expression in a series of 66 ovarian carcinoma effusions showed the frequent expression of all 4 transporters in tumor cells, but no association with previous exposure to chemotherapy and response to chemotherapy at diagnosis, and no significant relationship to survival [58]. Another study by the author's group documented frequent expression of the phosphatidylinositol-3-kinase pathway proteins AKT, mTOR and DJ-1 in ovarian carcinoma effusions using FC, Western blotting and IHC [59].

FC immunophenotyping of epithelial and mesothelial cells in effusions is a quantitative and highly informative method. This, however, requires knowledge of the cell populations found in these specimens and an understanding of their immunoprofile. Common errors, like equating cytokeratin, EpCAM or CA-125 expression with epithelial differentiation (all are present on mesothelial cells) or interpreting annexin-V staining as evidence of apoptosis, may skew results considerably. The future role of FC in the management of patients with malignant effusions rather than in a pure research setting is yet to be decided. The potential is nevertheless evident. FC may be an efficient tool in identifying isolated tumor cells in a reactive background using reliable epithelial markers. It may also aid in assessing therapy response, for example through quantitative analysis of apoptosis or proliferation. In the context of targeted therapy, FC may identify the fraction of the tumor cell population expressing the protein to be silenced or inhibited. All this, however, depends on the availability of instruments and expertise, which remains to be resolved in the future.

The research of Prof. Davidson is supported by the Inger and John Fredriksen Foundation for Ovarian Cancer Research.

1.
Davidson B, Firat P, Michael CW: Serous Effusions - Etiology, Diagnosis, Prognosis and Therapy. Springer, London, 2012.
2.
Friedman MT, Gentile P, Tarectecan A, Fuchs A: Malignant mesothelioma: immunohistochemistry and DNA ploidy analysis as methods to differentiate mesothelioma from benign reactive mesothelial cell proliferation and adenocarcinoma in pleural and peritoneal effusions. Arch Pathol Lab Med 1996;120:959-966.
3.
Chen LM, Lazcano O, Katzmann JA, Kimlinger TK, Li CY: The role of conventional cytology, immunocytochemistry, and flow cytometric DNA ploidy in the evaluation of body cavity fluids: a prospective study of 52 patients. Am J Clin Pathol 1998;109:712-721.
4.
Saha I, Dey P, Vhora H, Nijhawan R: Role of DNA flow cytometry and image cytometry on effusion fluid. Diagn Cytopathol 2000;22:81-85.
5.
Both CT, de Mattos AA, Neumann J, Reis MD: Flow cytometry in the diagnosis of peritoneal carcinomatosis. Am J Gastroenterol 2001;96:1605-1609.
6.
Krishan A, Ganjei-Azar P, Jorda M, Hamelik RM, Reis IM, Nadji M: Detection of tumor cells in body cavity fluids by flow cytometric and immunocytochemical analysis. Diagn Cytopathol 2006;34:528-541.
7.
Kundu R, Handa U, Mohan H: Role of DNA flow cytometry and immunocytochemical analysis in diagnosis of malignant effusions. Diagn Cytopathol 2012;40:887-892.
8.
Risberg B, Davidson B, Dong HP, Nesland JM, Berner A: Flow cytometric immunophenotyping of serous effusions and peritoneal washings: comparison with immunocytometry and morphological findings. J Clin Pathol 2000;53:513-517.
9.
Risberg B, Davidson B, Nielsen S, et al: Detection of monocyte/macrophage cell populations in effusions - a comparative study using flow cytometric immunophenotyping and immunocytochemistry. Diagn Cytopathol 2001;25:214-219.
10.
Pillai V, Cibas ES, Dorfman DM: A simplified flow cytometric immunophenotyping procedure for the diagnosis of effusions caused by epithelial malignancies. Am J Clin Pathol 2013;139:672-681.
11.
Davidson B, Dong HP, Berner A, et al: Detection of malignant epithelial cells in effusions using flow cytometric immunophenotyping: an analysis of 92 cases. Am J Clin Pathol 2002;118:85-92.
12.
Hartman M, Baruch A, Ron I, et al: MUC1 isoform specific monoclonal antibody 6E6/2 detects preferential expression of the novel MUC1/Y protein in breast and ovarian cancer. Int J Cancer 1999;82:256-267.
13.
Kentrou NA, Tsagarakis NJ, Tzanetou K, et al: An improved flow cytometric assay for detection and discrimination between malignant cells and atypical mesothelial cells, in serous cavity effusions. Cytometry B Clin Cytom 2011;80:324-334.
14.
Kitayama J, Emoto S, Yamaguchi H, et al: Flow cytometric quantification of intraperitoneal free tumor cells is a useful biomarker in gastric cancer patients with peritoneal metastasis. Ann Surg Oncol 2015;22:2336-2342.
15.
Roca E, Lacroix R, Judicone C, et al: Detection of EpCAM-positive microparticles in pleural fluid: a new approach to mini-invasively identify patients with malignant pleural effusions. Oncotarget 2016;7:3357-3366.
16.
Forster MD, Ormerod MG, Agarwal R, Kaye SB, Jackman AL: Flow cytometric method for determining folate receptor expression on ovarian carcinoma cells. Cytometry A 2007;71:945-950.
17.
Davidson B, Zhang Z, Kleinberg L, et al: Gene expression signatures differentiate ovarian/peritoneal serous carcinoma from diffuse malignant peritoneal mesothelioma. Clin Cancer Res 2006;12:5944-5950.
18.
Yuan Y, Nymoen DA, Dong HP, et al: Expression of the folate receptor genes FOLR1 and FOLR3 differentiates ovarian carcinoma from breast carcinoma and malignant mesothelioma in serous effusions. Hum Pathol 2009;40:1453-1460.
19.
Yuan Y, Dong HP, Nymoen DA, Nesland JM, Wu C, Davidson B: PINCH-2 expression in cancers involving the serosal cavities using quantitative PCR. Cytopathology 2011;22:22-29.
20.
Cornfield DB, Gheith SM: Flow cytometric quantitation of natural killer cells and T lymphocytes expressing T-cell receptors alpha/beta and gamma/delta is not helpful in distinguishing benign from malignant body cavity effusions. Cytometry B Clin Cytom 2009;76:213-217.
21.
Wang F, Yang L, Gao Q, et al: CD163+CD14+ macrophages, a potential immune biomarker for malignant pleural effusion. Cancer Immunol Immunother 2015;64:965-976.
22.
Scherpereel A, Grigoriu BD, Noppen M, et al: Defect in recruiting effector memory CD8+ T-cells in malignant pleural effusions compared to normal pleural fluid. BMC Cancer 2013;13:324.
23.
Desfrançois J, Derré L, Corvaisier M, et al: Increased frequency of nonconventional double positive CD4CD8 αβ T cells in human breast pleural effusions. Int J Cancer 2009;125:374-380.
24.
Giuntoli RL 2nd, Webb TJ, Zoso A, et al: Ovarian cancer-associated ascites demonstrates altered immune environment: implications for antitumor immunity. Anticancer Res 2009;29:2875-2884.
25.
Dong HP, Bunkholt Elstrand M, Holth A, et al: NK and B cell infiltration correlates with worse outcome in metastatic ovarian carcinoma. Am J Clin Pathol 2006;125:451-458.
26.
Lee SW, Kim YM, Lee HY, et al: Proliferation of CD4CD25Foxp3 regulatory T lymphocytes in ex vivo expanded ascitic fluid from primary and recurrent ovarian carcinoma. J Gynecol Oncol 2010;21:38-44.
27.
Shen LS, Wang J, Shen DF, et al: CD4+ CD25+CD127low/- regulatory T cells express Foxp3 and suppress effector T cell proliferation and contribute to gastric cancers progression. Clin Immunol 2009;131:109-118.
28.
Chen YQ, Shi HZ, Qin XJ, et al: CD4+CD25+ regulatory T lymphocytes in malignant pleural effusion. Am J Respir Crit Care Med 2005;172:1434-1439.
29.
Qin XJ, Shi HZ, Deng JM, Liang QL, Jiang J, Ye ZJ: CCL22 recruits CD4-positive CD25-positive regulatory T cells into malignant pleural effusion. Clin Cancer Res 2009;15:2231-2237.
30.
Yang G, Li H, Yao Y, Xu F, Bao Z, Zhou J: Treg/Th17 imbalance in malignant pleural effusion partially predicts poor prognosis. Oncol Rep 2015;33:478-484.
31.
Prado-Garcia H, Romero-Garcia S, Rumbo-Nava U, Lopez-Gonzalez JS: Predominance of Th17 over regulatory T-cells in pleural effusions of patients with lung cancer implicates a proinflammatory profile. Anticancer Res 2015;35:1529-1535.
32.
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.
33.
Davidson B, Dong HP, Holth A, Berner A, Risberg B: The chemokine receptor CXCR4 is more frequently expressed in breast compared to other metastatic adenocarcinomas in effusions. Breast J 2008;14:476-482.
34.
Davidson B, Dong HP, Holth A, Berner A, Risberg B: Chemokine receptors are infrequently expressed in malignant or benign mesothelial cells. Am J Clin Pathol 2007;127:752-759.
35.
Atanackovic D, Block A, de Weerth A, Faltz C, Hossfeld DK, Hegewisch-Becker S: Characterization of effusion-infiltrating T cells: benign versus malignant effusions. Clin Cancer Res 2004;10:2600-2608.
36.
Kaczmarek M, Nowicka A, Kozłowska M, Zurawski J, Batura-Gabryel H, Sikora J: Evaluation of the phenotype pattern of macrophages isolated from malignant and non-malignant pleural effusions. Tumour Biol 2011;32:1123-1132.
37.
Davidson B, Goldberg I, Reich R, et al: αV- and β1-integrin subunits are commonly expressed in malignant effusions from ovarian carcinoma patients. Gynecol Oncol 2003;90:248-257.
38.
Givant-Horwitz V, Davidson B, van de Putte G, et al: Expression of the 67 kDa laminin receptor and the α6 integrin subunit in serous ovarian carcinoma. Clin Exp Metastasis 2003;20:599-609.
39.
Sigstad E, Dong HP, Nielsen S, Berner A, Davidson B, Risberg B: Quantitative analysis of integrin expression in effusions using flow cytometric immunophenotyping. Diagn Cytopathol 2005;33:321-331.
40.
Koyama S, Maruyama T, Adachi S: Expression of epidermal growth factor receptor and CD44 splicing variants sharing exons 6 and 9 on gastric and esophageal carcinomas: a two-color flow-cytometric analysis. J Cancer Res Clin Oncol 1999;125:47-54.
41.
Taylor DD, Gercel-Taylor C, Gall SA: Expression and shedding of CD44 variant isoforms in patients with gynecologic malignancies. J Soc Gynecol Investig 1996;3:289-294.
42.
Koyama S: Coordinate cell-surface expression of matrix metalloproteinases and their inhibitors on cancer-associated myofibroblasts from malignant ascites in patients with gastric carcinoma. J Cancer Res Clin Oncol 2005;131:809-814.
43.
Lanitis E, Dangaj D, Hagemann IS, et al: Primary human ovarian epithelial cancer cells broadly express HER2 at immunologically-detectable levels. PLoS One 2012;7:e49829.
44.
Zhang J, Qiu S, Zhang Y, et al: Loss of mesothelin expression by mesothelioma cells grown in vitro determines sensitivity to anti-mesothelin immunotoxin SS1P. Anticancer Res 2012;32:5151-5158.
45.
Meng E, Mitra A, Tripathi K, et al: ALDH1A1 maintains ovarian cancer stem cell-like properties by altered regulation of cell cycle checkpoint and DNA repair network signaling. PLoS One 2014;9:e107142.
46.
Arend RC, Londoño-Joshi AI, Samant RS, et al: Inhibition of Wnt/β-catenin pathway by niclosamide: a therapeutic target for ovarian cancer. Gynecol Oncol 2014;13:112-120.
47.
Hu L, McArthur C, Jaffe RB: Ovarian cancer stem-like side-population cells are tumourigenic and chemoresistant. Br J Cancer 2010;102:1276-1283.
48.
Yao X, Labelle M, Lamb CR, et al: Determination of 35 cell surface antigen levels in malignant pleural effusions identifies CD24 as a marker of disseminated tumor cells. Int J Cancer 2013;133:2925-2933.
49.
Wang X, Osada T, Wang Y, et al: CSPG4 protein as a new target for the antibody-based immunotherapy of triple-negative breast cancer. J Natl Cancer Inst 2010;102:1496-1512.
50.
Dong HP, Kleinberg L, Davidson B, Risberg B: Methods for simultaneous measurement of apoptosis and cell surface phenotype of epithelial cells in effusions by flow cytometry. Nat Protoc 2008;3:955-964.
51.
Kleinberg L, Dong HP, Holth A, et al: Cleaved caspase-3 and nuclear factor-κB p65 are prognostic factors in metastatic serous ovarian carcinoma. Hum Pathol 2009;40:795-806.
52.
Dong HP, Holth A, Kleinberg L, et al: Evaluation of cell surface expression of phosphatidylserine in ovarian carcinoma effusions using the annexin-V/7-AAD assay: clinical relevance and comparison with other apoptosis parameters. Am J Clin Pathol 2009;132:756-762.
53.
Dong HP, Holth A, Ruud MG, Emilsen E, Risberg B, Davidson B: Measurement of apoptosis in cytological specimens by flow cytometry: comparison of Annexin V, caspase cleavage and dUTP incorporation assays. Cytopathology 2011;22:365-372.
54.
Dong HP, Ree Rosnes AK, Bock AJ, et al: Flow cytometric measurement of cellular FLICE-inhibitory protein (c-FLIP) in ovarian carcinoma effusions. Cytopathology 2011;22:373-382.
55.
Dong HP, Kleinberg L, Silins I, et al: Death receptor expression is associated with poor response to chemotherapy and shorter survival in metastatic ovarian carcinoma. Cancer 2008;112:84-93.
56.
Koyama S, Koike N, Adachi S: Expression of TNF-related apoptosis-inducing ligand (TRAIL) and its receptors in gastric carcinoma and tumor-infiltrating lymphocytes: a possible mechanism of immune evasion of the tumor. J Cancer Res Clin Oncol 2002;128:73-79.
57.
Zhou Y, He J, Gou LT, et al: Expression of CD40 and growth-inhibitory activity of CD40 agonist in ovarian carcinoma cells. Cancer Immunol Immunother 2012;61:1735-1743.
58.
Bock AJ, Dong HP, Tropé CG, Staff AC, Risberg B, Davidson B: Nucleoside transporters are widely expressed in ovarian carcinoma effusions. Cancer Chemother Pharmacol 2012;69:467-475.
59.
Bunkholt Elstrand M, Dong HP, Ødegaard E, et al: Mammalian target of rapamycin is a biomarker of poor survival in metastatic serous ovarian carcinoma. Hum Pathol 2010;41:794-804.
Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.