Editorial Open Access
Copyright ©The Author(s) 2018. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Oncol. Dec 20, 2018; 9(8): 167-171
Published online Dec 20, 2018. doi: 10.5306/wjco.v9.i8.167
Malignant peritoneal effusion acting as a tumor environment in ovarian cancer progression: Impact and significance
Alain Piché, Département de Microbiologie et Infectiologie, Faculté de Médecine, Université de Sherbrooke, Sherbrooke QC J1H 5N4, Canada
ORCID number: Alain Piché, (0000-0002-6313-4538).
Author contributions: Piché A drafted the manuscript.
Conflict-of-interest statement: The authors have no conflict of interest to declare.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Corresponding author to: Alain Piché, BSc, FRCP (C), MD, MSc, Professor, Département de Microbiologie et Infectiologie, Faculté de Médecine, Université de Sherbrooke, 3001, 12ième Avenue Nord, Sherbrooke QC J1H 5N4, Canada. alain.piche@usherbrooke.ca
Telephone: +1-819-3461110-75734 Fax: +1-819-5645392
Received: July 27, 2018
Peer-review started: July 30, 2018
First decision: October 8, 2018
Revised: October 11, 2018
Accepted: November 4, 2018
Article in press: November 4, 2018
Published online: December 20, 2018
Processing time: 147 Days and 6.4 Hours

Abstract

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.

Key Words: Ovarian cancer; Tumor environment; Peritoneal effusions; Ascites; Dissemination; Multicellular spheroids

Core tip: The malignant peritoneal effusion that accumulates during ovarian cancer dissemination and progression constitutes a unique tumor environment. Bidirectional communications between tumor cells and their surrounding environment influence ovarian cancer dissemination, progression and patient prognosis. To solve the complexity of this tumor environment and understand how it affects cancer progression, a paradigm shift is necessary. Peritoneal effusions should be studied as integrated systems with innovative modeling approaches.



MAIN TEXT

Malignant peritoneal effusions (ascites) commonly occur during the progression of certain types of cancers such as ovarian and pancreatic adenocarcinomas[1]. These exudative fluids contain a large number of tumor cells and are by definition metastatic. For example, patients with ovarian cancer will often present at diagnostic with multiple omental and peritoneal tumor implants. These patients will usually have accumulation of large amount of peritoneal fluids (often several liters). The presence of malignant effusion is associated with a poor prognosis. Because of their nature, malignant effusions, in which tumor cells proliferate and metastasize, constitute a distinct and unique tumor environment compared to solid cancers that develop in a tissue microenvironment. The cell-free fraction of malignant effusions is rich in cytokines, chemokines, growth factors and extracellular matrix components[2-4]. In addition to tumor cells, effusions contain of a large number of non-malignant cells such as fibroblasts and mesothelial cells, and various immune cells including macrophages and lymphocytes[5]. Although the importance of the bidirectional communication between tumor cells and their adjacent tissue microenvironment for supporting cancer progression is well recognized now, much less is known regarding the role peritoneal effusions in cancer progression. Nonetheless, it has been shown that extracellular cues provided by soluble factors contain in effusions can drive certain aspects of cancer progression such as cell proliferation, migration, and invasion. For example, effusion-associated hepatocyte growth factor[6-8], lysophosphatidic acid[9] and CCL18[10] have been shown to promote ovarian cancer cell growth and survival. Furthermore, cell-free ascites can promote drug resistance in ovarian cancer cells[11,12]. In support of the critical role of cell-free ascites, a number of factors contained in effusions are predictor of clinical prognosis[13,14]. The overall effect of cell-free ascites results from the complex integration of various extracellular cues, which leading to receptor-induced signaling in both tumor cells and stromal cells. For example, in contact with ascites, mesothelial cells undergo a pro-tumoral shift[15] whereas ovarian cancer cells display a more invasive phenotype with a shift along an epithelial-to-mesenchymal transition (EMT)[16]. As a result of this extensive cellular crosstalk, peritoneal effusions are constantly adapting during the course of the disease in response to the different cues[1]. The functional role of these changes in effusion composition during cancer dissemination and progression is mostly unknown.

Beyond the potential contribution of specific soluble factors to cancer progression, non-malignant cells present in peritoneal effusions probably play an essential supporting role, particularly in the early stages of dissemination, by preventing anoikis and stimulating proliferation of tumor cells. Paracrine signaling through mesenchymal stem cells can confer resistance to chemotherapy in ovarian cancer cells via IL-6/IL-6 R pathway and CXCL12[17,18]. Similarly, paracrine secretion of anti-apoptotic from ascites-primed mesothelial cells promote ovarian cancer cell drug resistance[15]. Cancer-associated fibroblasts isolated from omentum can be activated by tumor cells to promote ovarian cancer growth, adhesion and invasiveness through TGFβ1 pathway[19]. Expression of HOXA9 by ovarian cancer cells induce TGFβ2 secretion, which in turn, promote by secretion of IL-6 and CXCL12 by fibroblasts leading to tumor growth[20]. The presence of tumor-infiltrating CD8+ T cells in primary tumors is associated with prolonged progression-free and overall survival in ovarian cancer patients[21,22]. These data suggest that several cell types within peritoneal effusions may have a robust influence on ovarian cancer cells. Bidirectional communications between tumor cells and their surrounding environment are at play, however identifying these crosstalks and validating their functional roles in a systemic approach remains challenging because of complex interactions among multiple cell types in effusions.

Getting a better understanding of the complex ecosystem that constitute malignant peritoneal effusions also have the potential to enhance our insight to limit the accumulation of these effusions and to overcome platinum resistance in ovarian cancer, which represents a yet unmet need. Both soluble factors and cancer-associated stromal and immune cells constitute potential targets for novel therapies. Examples of soluble factors in the tumor environment of ovarian cancer targeted by novel therapies (mostly humanized monoclonal antibodies) include VEGF, VEGFR, PDFR, PDGFR, c-kit, PD-1 and PD-L1[23]. However, drug-associated toxicity and resistance often limit the clinical benefits associated with these novel therapies. These studies highlight the fact that targeting a single component of the tumor environment is likely to have limited clinical benefits and the secretome of malignant peritoneal effusions should be considered as a whole that cannot be understood simply from its parts. Given the progression-enhancing role of cancer-associated stromal cells in malignant peritoneal effusions, cell therapy may represent of novel approach to limit ovarian cancer dissemination. For example, it has been shown that genetically-engineered human mesothelial cells with the Herpes Simplex virus thymidime kinase (HSV-tk) efficiently deliver anticancer modalities to ovarian cancer cells within the peritoneal cavity when mice were treated with ganciclovir[24]. Conversion of ganciclovir pro-drug in a toxic apoptosis-inducing drug by HSV-TK results in the transfer of apoptotic bodies to adjacent tumor cells. Alternatively, the cells could be used to locally deliver in the peritoneal cavity other anti-cancer molecules potentially circumventing the toxicity associated with systemic delivery.

Tumor such as ovarian cancer does not usually use the classic patterns of metastasis via the hematogenous route[25]. The current view is that ovarian cancer spreads mainly via the peritoneal circulation with a high affinity for the omentum. Cells detach from the primary tumor to form free-floating multicellular spheroids, which then travel through the peritoneal fluid at distant sites onto the mesothelial lining where metastatic outgrowth occurs[26]. It is believed that the formation of multicellular spheroids is an essential step in the initiation of peritoneal implantation metastasis for ovarian cancer. In this context, spheroids can be considered the primary vehicles of ovarian cancer dissemination. However, the mechanisms of spheroid formation are for the most part unknown. Before tumor cells detach, they will usually undergo an EMT characterized by decreased expression of E-cadherin acquiring therefore a proliferative and invasive phenotype. However, it is not clear if tumor cells detached as single cell entity and then aggregate together to form homotypic multicellular spheroids or if clumps of the primary tumor exfoliate and stay together to form heterotypic spheroids. Alternatively, exfoliated single tumor cells could aggregate with floating mesothelial cells forming heterotypic unvascularized multicellular spheroids. Recent studies aiming to characterize ascites-derived spheroids suggested that spheroids are heterotypic containing mesothelial-derived myofibroblatic cells[27] and macrophages[28]. The binding of ovarian cancer cells to mesothelial cells to form spheroids could be mediated by integrins, CD44 and MUC16[27,29,30]. Further work however is needed to gain a comprehensive understanding of the cellular composition of the multicellular spheroids found in ascites and how this influences cancer progression. Another interesting hypothesis is that multicellular spheroids potentially constitute a chemoresistant niche that continuously repopulates the peritoneal cavity despite chemotherapy treatments as they are potentially endowed with a high tumor-initiating potential and increased drug resistance linked to the expression of stemness-associated genes[31]. However, the presence of mesenchymal stem cells in ovarian cancer spheroids remains to be demonstrated. The generation of unvascularized 3D spheroids would generate a structure with a metabolite density gradient that can inhibit the access of chemotherapy agents to internal cells[32]. Ovarian cancer cell grown as spheroids display enhanced resistance to common chemotherapeutic drugs such as taxol and cisplatin compared monolayers[31,33]. Because of their role in ovarian cancer dissemination and their potential role in disease recurrence after chemotherapy, a much better understanding of multicellular spheroid biology is necessary.

PERSPECTIVE

As highlighted here, most research on the ovarian cancer environment has focused one at a time on components, such as particular signaling cascades, specific ascites factors or specific cell types, etc. This knowledge is essential, but obviously not sufficient. The tumor environment that represents peritoneal effusions closely resembles an ecosystem, with specific cell types and signaling molecules increasing and decreasing during the course of the disease progression creating a single complex network. This environment is an archetypical complex system in which the functioning of the whole cannot be understood simply from its parts or outside is environmental context. Perhaps to understand the role of peritoneal effusions on cancer progression, a paradigm shift is necessary and effusions should be studied as integrated systems using bioinformatic modeling to quantify system-level biodiversity changes[34].

Footnotes

Manuscript source: Invited manuscript

Specialty type: Oncology

Country of origin: Canada

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C, C, C, C

Grade D (Fair): D

Grade E (Poor): 0

P- Reviewer: Huo Q, Kanat O, Su CC, Wion D, Yokoyama S S- Editor: Ji FF L- Editor: A E- Editor: Bian YN

References
1.  Matte I, Bessette P, Piché A. Ascites in ovarian cancer progression: opportunities for biomarker discovery and new avenues for targeted therapies. INTECH. 2017;Chapter 9:146-163.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Matte I, Lane D, Laplante C, Rancourt C, Piché A. Profiling of cytokines in human epithelial ovarian cancer ascites. Am J Cancer Res. 2012;2:566-580.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Ahmed N, Stenvers KL. Getting to know ovarian cancer ascites: opportunities for targeted therapy-based translational research. Front Oncol. 2013;3:256.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 246]  [Cited by in F6Publishing: 320]  [Article Influence: 29.1]  [Reference Citation Analysis (0)]
4.  Giuntoli RL 2nd, Webb TJ, Zoso A, Rogers O, Diaz-Montes TP, Bristow RE, Oelke M. Ovarian cancer-associated ascites demonstrates altered immune environment: implications for antitumor immunity. Anticancer Res. 2009;29:2875-2884.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Kipps E, Tan DS, Kaye SB. Meeting the challenge of ascites in ovarian cancer: new avenues for therapy and research. Nat Rev Cancer. 2013;13:273-282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 399]  [Cited by in F6Publishing: 399]  [Article Influence: 36.3]  [Reference Citation Analysis (0)]
6.  Nakamura M, Ono YJ, Kanemura M, Tanaka T, Hayashi M, Terai Y, Ohmichi M. Hepatocyte growth factor secreted by ovarian cancer cells stimulates peritoneal implantation via the mesothelial-mesenchymal transition of the peritoneum. Gynecol Oncol. 2015;139:345-354.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 43]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
7.  Matte I, Lane D, Laplante C, Garde-Granger P, Rancourt C, Piché A. Ovarian cancer ascites enhance the migration of patient-derived peritoneal mesothelial cells via cMet pathway through HGF-dependent and -independent mechanisms. Int J Cancer. 2015;137:289-298.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 30]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
8.  Kwon Y, Smith BD, Zhou Y, Kaufman MD, Godwin AK. Effective inhibition of c-MET-mediated signaling, growth and migration of ovarian cancer cells is influenced by the ovarian tissue microenvironment. Oncogene. 2015;34:144-153.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 53]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
9.  Mills GB, Moolenaar WH. The emerging role of lysophosphatidic acid in cancer. Nat Rev Cancer. 2003;3:582-591.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 875]  [Cited by in F6Publishing: 894]  [Article Influence: 42.6]  [Reference Citation Analysis (0)]
10.  Lane D, Matte I, Laplante C, Garde-Granger P, Carignan A, Bessette P, Rancourt C, Piché A. CCL18 from ascites promotes ovarian cancer cell migration through proline-rich tyrosine kinase 2 signaling. Mol Cancer. 2016;15:58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 46]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
11.  Lane D, Goncharenko-Khaider N, Rancourt C, Piché A. Ovarian cancer ascites protects from TRAIL-induced cell death through alphavbeta5 integrin-mediated focal adhesion kinase and Akt activation. Oncogene. 2010;29:3519-3531.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 69]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
12.  Goncharenko-Khaider N, Matte I, Lane D, Rancourt C, Piché A. Ovarian cancer ascites increase Mcl-1 expression in tumor cells through ERK1/2-Elk-1 signaling to attenuate TRAIL-induced apoptosis. Mol Cancer. 2012;11:84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 37]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
13.  Kryczek I, Gryboś M, Karabon L, Klimczak A, Lange A. IL-6 production in ovarian carcinoma is associated with histiotype and biological characteristics of the tumour and influences local immunity. Br J Cancer. 2000;82:621-628.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 33]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
14.  Lane D, Matte I, Rancourt C, Piché A. Prognostic significance of IL-6 and IL-8 ascites levels in ovarian cancer patients. BMC Cancer. 2011;11:210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 129]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
15.  Matte I, Lane D, Bachvarov D, Rancourt C, Piché A. Role of malignant ascites on human mesothelial cells and their gene expression profiles. BMC Cancer. 2014;14:288.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 28]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
16.  Carduner L, Leroy-Dudal J, Picot CR, Gallet O, Carreiras F, Kellouche S. Ascites-induced shift along epithelial-mesenchymal spectrum in ovarian cancer cells: enhancement of their invasive behavior partly dependant on αv integrins. Clin Exp Metastasis. 2014;31:675-688.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 36]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
17.  Pasquier J, Gosset M, Geyl C, Hoarau-Véchot J, Chevrot A, Pocard M, Mirshahi M, Lis R, Rafii A, Touboul C. CCL2/CCL5 secreted by the stroma induce IL-6/PYK2 dependent chemoresistance in ovarian cancer. Mol Cancer. 2018;17:47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 49]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
18.  Lis R, Touboul C, Mirshahi P, Ali F, Mathew S, Nolan DJ, Maleki M, Abdalla SA, Raynaud CM, Querleu D. Tumor associated mesenchymal stem cells protects ovarian cancer cells from hyperthermia through CXCL12. Int J Cancer. 2011;128:715-725.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 79]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
19.  Cai J, Tang H, Xu L, Wang X, Yang C, Ruan S, Guo J, Hu S, Wang Z. Fibroblasts in omentum activated by tumor cells promote ovarian cancer growth, adhesion and invasiveness. Carcinogenesis. 2012;33:20-29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 140]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
20.  Ko SY, Barengo N, Ladanyi A, Lee JS, Marini F, Lengyel E, Naora H. HOXA9 promotes ovarian cancer growth by stimulating cancer-associated fibroblasts. J Clin Invest. 2012;122:3603-3617.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 105]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
21.  Chu CS, Kim SH, June CH, Coukos G. Immunotherapy opportunities in ovarian cancer. Expert Rev Anticancer Ther. 2008;8:243-257.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 54]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
22.  Nelson BH. The impact of T-cell immunity on ovarian cancer outcomes. Immunol Rev. 2008;222:101-116.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 119]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
23.  Hansen JM, Coleman RL, Sood AK. Targeting the tumour microenvironment in ovarian cancer. Eur J Cancer. 2016;56:131-143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 83]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
24.  Rancourt C, Bergeron C, Lane D, Garon G, Piché A. Delivery of herpes simplex thymidine kinase bystander effect by engineered human mesothelial cells for the treatment of ovarian cancer. Cytotherapy. 2003;5:509-522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 6]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
25.  Yeung TL, Leung CS, Yip KP, Au Yeung CL, Wong ST, Mok SC. Cellular and molecular processes in ovarian cancer metastasis. A Review in the Theme: Cell and Molecular Processes in Cancer Metastasis. Am J Physiol Cell Physiol. 2015;309:C444-C456.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 188]  [Cited by in F6Publishing: 244]  [Article Influence: 27.1]  [Reference Citation Analysis (0)]
26.  Shield K, Ackland ML, Ahmed N, Rice GE. Multicellular spheroids in ovarian cancer metastases: Biology and pathology. Gynecol Oncol. 2009;113:143-148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 267]  [Cited by in F6Publishing: 287]  [Article Influence: 19.1]  [Reference Citation Analysis (0)]
27.  Matte I, Legault CM, Garde-Granger P, Laplante C, Bessette P, Rancourt C, Piché A. Mesothelial cells interact with tumor cells for the formation of ovarian cancer multicellular spheroids in peritoneal effusions. Clin Exp Metastasis. 2016;33:839-852.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 28]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
28.  Yin M, Li X, Tan S, Zhou HJ, Ji W, Bellone S, Xu X, Zhang H, Santin AD, Lou G. Tumor-associated macrophages drive spheroid formation during early transcoelomic metastasis of ovarian cancer. J Clin Invest. 2016;126:4157-4173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 261]  [Article Influence: 32.6]  [Reference Citation Analysis (0)]
29.  Giannakouros P, Comamala M, Matte I, Rancourt C, Piché A. MUC16 mucin (CA125) regulates the formation of multicellular aggregates by altering β-catenin signaling. Am J Cancer Res. 2014;5:219-230.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Nakamura K, Sawada K, Kinose Y, Yoshimura A, Toda A, Nakatsuka E, Hashimoto K, Mabuchi S, Morishige KI, Kurachi H. Exosomes Promote Ovarian Cancer Cell Invasion through Transfer of CD44 to Peritoneal Mesothelial Cells. Mol Cancer Res. 2017;15:78-92.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 154]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
31.  Liao J, Qian F, Tchabo N, Mhawech-Fauceglia P, Beck A, Qian Z, Wang X, Huss WJ, Lele SB, Morrison CD. Ovarian cancer spheroid cells with stem cell-like properties contribute to tumor generation, metastasis and chemotherapy resistance through hypoxia-resistant metabolism. PLoS One. 2014;9:e84941.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 219]  [Cited by in F6Publishing: 251]  [Article Influence: 25.1]  [Reference Citation Analysis (0)]
32.  Benton G, DeGray G, Kleinman HK, George J, Arnaoutova I. In vitro microtumors provide a physiologically predictive tool for breast cancer therapeutic screening. PLoS One. 2015;10:e0123312.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 48]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
33.  L’Espérance S, Bachvarova M, Tetu B, Mes-Masson AM, Bachvarov D. Global gene expression analysis of early response to chemotherapy treatment in ovarian cancer spheroids. BMC Genomics. 2008;9:99.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 86]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
34.  Ovaskainen O, Tikhonov G, Norberg A, Guillaume Blanchet F, Duan L, Dunson D, Roslin T, Abrego N. How to make more out of community data? A conceptual framework and its implementation as models and software. Ecol Lett. 2017;20:561-576.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 429]  [Cited by in F6Publishing: 335]  [Article Influence: 47.9]  [Reference Citation Analysis (0)]