Malignant peritoneal effusion acting as a tumor environment in ovarian cancer progression: Impact and significance

Until recently, ovarian cancer research has mainly focused on the tumor cells themselves ignoring for the most part the surrounding tumor environment which includes malignant peritoneal effusions. However, one of the major conceptual advances in oncology over the last few years has been the appreciation that cancer progression cannot be explained by aberrations in cancer cells themselves and is strongly influenced by the surrounding tumor environment. The mechanisms of ovarian cancer progression differ from that of other solid tumors because ovarian cancer cells primarily disseminate within the peritoneal cavity. Malignant peritoneal effusion accumulates in the peritoneal cavity during ovarian cancer progression. These exudative fluids act as a unique tumor environment providing a framework that orchestrates cellular and molecular changes contributing to aggressiveness and disease progression. The composition of ascites, which includes cellular and acellular components, constantly adapts during the course of the disease in response to various cellular cues originating from both tumor and stromal cells. The tumor environment that represents peritoneal effusions closely constitute an ecosystem, with specific cell types and signaling molecules increasing and decreasing during the course of the disease progression creating a single complex network. Although recent advances aiming to understand the ovarian tumor environment have focused one at a time on components, the net impact of the whole environment cannot be understood simply from its parts or outside is environmental context.

[1]  M. Pocard,et al.  CCL2/CCL5 secreted by the stroma induce IL-6/PYK2 dependent chemoresistance in ovarian cancer , 2018, Molecular Cancer.

[2]  A. Piché,et al.  Ascites in Ovarian Cancer Progression: Opportunities for Biomarker Discovery and New Avenues for Targeted Therapies , 2017 .

[3]  Anna Norberg,et al.  How to make more out of community data? A conceptual framework and its implementation as models and software. , 2017, Ecology letters.

[4]  Wang Min,et al.  Tumor-associated macrophages drive spheroid formation during early transcoelomic metastasis of ovarian cancer. , 2016, The Journal of clinical investigation.

[5]  E. Lengyel,et al.  Exosomes Promote Ovarian Cancer Cell Invasion through Transfer of CD44 to Peritoneal Mesothelial Cells , 2016, Molecular Cancer Research.

[6]  C. Rancourt,et al.  CCL18 from ascites promotes ovarian cancer cell migration through proline-rich tyrosine kinase 2 signaling , 2016, Molecular Cancer.

[7]  C. Rancourt,et al.  Mesothelial cells interact with tumor cells for the formation of ovarian cancer multicellular spheroids in peritoneal effusions , 2016, Clinical & Experimental Metastasis.

[8]  A. Sood,et al.  Targeting the Tumor Microenvironment in Ovarian Cancer , 2016 .

[9]  M. Ohmichi,et al.  Hepatocyte growth factor secreted by ovarian cancer cells stimulates peritoneal implantation via the mesothelial-mesenchymal transition of the peritoneum. , 2015, Gynecologic oncology.

[10]  Stephen T. C. Wong,et al.  Cellular and molecular processes in ovarian cancer metastasis. A Review in the Theme: Cell and Molecular Processes in Cancer Metastasis. , 2015, American journal of physiology. Cell physiology.

[11]  C. Rancourt,et al.  Ovarian cancer ascites enhance the migration of patient‐derived peritoneal mesothelial cells via cMet pathway through HGF‐dependent and ‐independent mechanisms , 2015, International journal of cancer.

[12]  H. Kleinman,et al.  In Vitro Microtumors Provide a Physiologically Predictive Tool for Breast Cancer Therapeutic Screening , 2015, PloS one.

[13]  C. Rancourt,et al.  MUC16 mucin (CA125) regulates the formation of multicellular aggregates by altering β-catenin signaling. , 2015, American journal of cancer research.

[14]  S. Kellouche,et al.  Ascites-induced shift along epithelial-mesenchymal spectrum in ovarian cancer cells: enhancement of their invasive behavior partly dependant on αv integrins , 2014, Clinical & Experimental Metastasis.

[15]  C. Morrison,et al.  Ovarian Cancer Spheroid Cells with Stem Cell-Like Properties Contribute to Tumor Generation, Metastasis and Chemotherapy Resistance through Hypoxia-Resistant Metabolism , 2014, PloS one.

[16]  D. Bachvarov,et al.  Role of malignant ascites on human mesothelial cells and their gene expression profiles , 2014, BMC Cancer.

[17]  Yan Zhou,et al.  Effective Inhibition of c-MET-mediated Signaling, Growth, and Migration of Ovarian Cancer Cells is Influenced by the Ovarian Tissue Microenvironment , 2013, Oncogene.

[18]  N. Ahmed,et al.  Getting to Know Ovarian Cancer Ascites: Opportunities for Targeted Therapy-Based Translational Research , 2013, Front. Oncol..

[19]  S. Kaye,et al.  Meeting the challenge of ascites in ovarian cancer: new avenues for therapy and research , 2013, Nature Reviews Cancer.

[20]  C. Rancourt,et al.  Ovarian cancer ascites increase Mcl-1 expression in tumor cells through ERK1/2-Elk-1 signaling to attenuate TRAIL-induced apoptosis , 2012, Molecular Cancer.

[21]  E. Lengyel,et al.  HOXA9 promotes ovarian cancer growth by stimulating cancer-associated fibroblasts. , 2012, The Journal of clinical investigation.

[22]  Jing Cai,et al.  Fibroblasts in omentum activated by tumor cells promote ovarian cancer growth, adhesion and invasiveness. , 2012, Carcinogenesis.

[23]  C. Rancourt,et al.  Profiling of cytokines in human epithelial ovarian cancer ascites. , 2012, American journal of cancer research.

[24]  C. Rancourt,et al.  Prognostic significance of IL-6 and IL-8 ascites levels in ovarian cancer patients , 2011, BMC Cancer.

[25]  J. Malek,et al.  Tumor associated mesenchymal stem cells protects ovarian cancer cells from hyperthermia through CXCL12 , 2011, International journal of cancer.

[26]  C. Rancourt,et al.  Ovarian cancer ascites protects from TRAIL-induced cell death through αvβ5 integrin-mediated focal adhesion kinase and Akt activation , 2010, Oncogene.

[27]  R. Bristow,et al.  Ovarian cancer-associated ascites demonstrates altered immune environment: implications for antitumor immunity. , 2009, Anticancer research.

[28]  G. Rice,et al.  Multicellular spheroids in ovarian cancer metastases: Biology and pathology. , 2009, Gynecologic oncology.

[29]  B. Nelson,et al.  The impact of T‐cell immunity on ovarian cancer outcomes , 2008, Immunological reviews.

[30]  G. Coukos,et al.  Immunotherapy opportunities in ovarian cancer , 2008, Expert review of anticancer therapy.

[31]  A. Mes-Masson,et al.  Global gene expression analysis of early response to chemotherapy treatment in ovarian cancer spheroids , 2008, BMC Genomics.

[32]  C. Rancourt,et al.  Delivery of herpes simplex thymidine kinase bystander effect by engineered human mesothelial cells for the treatment of ovarian cancer. , 2003, Cytotherapy.

[33]  G. Mills,et al.  The emerging role of lysophosphatidic acid in cancer , 2003, Nature Reviews Cancer.

[34]  L. Karabon,et al.  IL-6 production in ovarian carcinoma is associated with histiotype and biological characteristics of the tumour and influences local immunity , 2000, British Journal of Cancer.